Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Int J Mol Sci ; 22(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946381

ABSTRACT

Understanding the host anti-fungal immunity induced by beta-glucan has been one of the most challenging conundrums in the field of biomedical research. During the last couple of decades, insights on the role of beta-glucan in fungal disease progression, susceptibility, and resistance have been greatly augmented through the utility of various beta-glucan cognate receptor-deficient mouse models. Analysis of dectin-1 knockout mice has clarified the downstream signaling pathways and adaptive effector responses triggered by beta-glucan in anti-fungal immunity. On the other hand, assessment of CR3-deficient mice has elucidated the compelling action of beta-glucans in neutrophil-mediated fungal clearance, and the investigation of EphA2-deficient mice has highlighted its novel involvement in host sensing and defense to oral mucosal fungal infection. Based on these accounts, this review focuses on the recent discoveries made by these gene-targeted mice in beta-glucan research with particular emphasis on the multifaceted aspects of fungal immunity.


Subject(s)
Fungi/immunology , Mycoses/immunology , beta-Glucans/immunology , Adaptive Immunity , Animals , Disease Models, Animal , Gene Deletion , Humans , Immunity , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Mice , Mice, Knockout , Mycoses/genetics , Mycoses/microbiology , Receptor, EphA2/genetics , Receptor, EphA2/immunology
2.
Anal Chem ; 90(5): 3024-3029, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29443500

ABSTRACT

Upconversion nanoparticles (UCNPs) are new optical probes for biological applications. For specific biomolecular recognition to be realized for diagnosis and imaging, the key lies in developing a stable and easy-to-use bioconjugation method for antibody modification. Current methods are not yet satisfactory regarding conjugation time, stability, and binding efficiency. Here, we report a facile and high-yield approach based on a bispecific antibody (BsAb) free of chemical reaction steps. One end of the BsAb is designed to recognize methoxy polyethylene glycol-coated UCNPs, and the other end of the BsAb is designed to recognize the cancer antigen biomarker. Through simple vortexing, BsAb-UCNP nanoprobes form within 30 min and show higher (up to 54%) association to the target than that of the traditional UCNP nanoprobes in the ELISA-like assay. We further demonstrate its successful binding to the cancer cells with high efficiency and specificity for background-free fluorescence imaging under near-infrared excitation. This method suggests a general approach broadly suitable for functionalizing a range of nanoparticles to specifically target biomolecules.


Subject(s)
Antibodies, Bispecific/immunology , Immunoconjugates/immunology , Nanoparticles/chemistry , Antibodies, Bispecific/chemistry , Cell Line, Tumor , Fluorescence , Humans , Immunoconjugates/chemistry , Light , Microscopy, Confocal/methods , Nanoparticles/radiation effects , Polyethylene Glycols/chemistry , Receptor, EphA2/immunology
3.
J Neurooncol ; 130(3): 517-527, 2016 12.
Article in English | MEDLINE | ID: mdl-27624914

ABSTRACT

Recurrent high-grade gliomas (HGGs) of childhood have an exceedingly poor prognosis with current therapies. Accordingly, new treatment approaches are needed. We initiated a pilot trial of vaccinations with peptide epitopes derived from glioma-associated antigens (GAAs) overexpressed in these tumors in HLA-A2+ children with recurrent HGG that had progressed after prior treatments. Peptide epitopes for three GAAs (EphA2, IL13Rα2, survivin), emulsified in Montanide-ISA-51, were administered subcutaneously adjacent to intramuscular injections of poly-ICLC every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-cell responses against the GAA epitopes, assessed by enzyme-linked immunosorbent spot (ELISPOT) analysis. Treatment response was evaluated clinically and by magnetic resonance imaging. Twelve children were enrolled, 6 with glioblastoma, 5 with anaplastic astrocytoma, and one with malignant gliomatosis cerebri. No dose-limiting non-CNS toxicity was encountered. ELISPOT analysis, in ten children, showed GAA responses in 9: to IL13Rα2 in 4, EphA2 in 9, and survivin in 3. One child had presumed symptomatic pseudoprogression, discontinued vaccine therapy, and responded to subsequent treatment. One other child had a partial response that persisted throughout 2 years of vaccine therapy, and continues at >39 months. Median progression-free survival (PFS) from the start of vaccination was 4.1 months and median overall survival (OS) was 12.9 months. 6-month PFS and OS were 33 and 73 %, respectively. GAA peptide vaccination in children with recurrent malignant gliomas is generally well tolerated, and has preliminary evidence of immunological and modest clinical activity.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy, Active/methods , Adolescent , Antigens, Neoplasm/chemistry , Carboxymethylcellulose Sodium/analogs & derivatives , Child , Child, Preschool , Female , Glioma/immunology , Glioma/metabolism , Humans , Infant , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/immunology , Interleukin-13 Receptor alpha1 Subunit , Male , Peptides/immunology , Pilot Projects , Poly I-C/immunology , Polylysine/analogs & derivatives , Polylysine/immunology , Receptor, EphA2/chemistry , Receptor, EphA2/immunology , Receptors, Interleukin-13/chemistry , Receptors, Interleukin-13/immunology , Survivin , Treatment Outcome , Young Adult
4.
Mol Ther ; 23(1): 171-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25142939

ABSTRACT

Adoptive immunotherapy with antigen-specific T cells has shown promise for the treatment of malignancies. However, infused T cells are unable to redirect resident T cells, limiting potential benefit. While the infusion of bispecific T-cell engagers can redirect resident T cells to tumors, these molecules have a short half-life, and do not self amplify. To overcome these limitations, we generated T cells expressing a secretable T-cell engager specific for CD3 and EphA2, an antigen expressed on a broad range of human tumors (EphA2-ENG T cells). EphA2-ENG T cells were activated and recognized tumor cells in an antigen-dependent manner, redirected bystander T cells to tumor cells, and had potent antitumor activity in glioma and lung cancer severe combined immunodeficiency (SCID) xenograft models associated with a significant survival benefit. This new class of tumor-specific T cells, with the unique ability to redirect bystander T cells, may be a promising alternative to current immunotherapies for cancer.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Bystander Effect/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line, Tumor , Gene Expression , Genetic Vectors , Glioma/genetics , Glioma/immunology , Glioma/pathology , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred ICR , Mice, SCID , Receptor, EphA2/genetics , Receptor, EphA2/immunology , Retroviridae/genetics , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
5.
Mol Cell Proteomics ; 13(12): 3320-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25149096

ABSTRACT

Many forms of antibody-based targeted therapeutics, including antibody drug conjugates, utilize the internalizing function of the targeting antibody to gain intracellular entry into tumor cells. Ideal antibodies for developing such therapeutics should be capable of both tumor-selective binding and efficient endocytosis. The macropinocytosis pathway is capable of both rapid and bulk endocytosis, and recent studies have demonstrated that it is selectively up-regulated by cancer cells. We hypothesize that receptor-dependent macropinocytosis can be achieved using tumor-targeting antibodies that internalize via the macropinocytosis pathway, improving potency and selectivity of the antibody-based targeted therapeutic. Although phage antibody display libraries have been utilized to find antibodies that bind and internalize to target cells, no methods have been described to screen for antibodies that internalize specifically via macropinocytosis. We hereby describe a novel screening strategy to identify phage antibodies that bind and rapidly enter tumor cells via macropinocytosis. We utilized an automated microscopic imaging-based, High Content Analysis platform to identify novel internalizing phage antibodies that colocalize with macropinocytic markers from antibody libraries that we have generated previously by laser capture microdissection-based selection, which are enriched for internalizing antibodies binding to tumor cells in situ residing in their tissue microenvironment (Ruan, W., Sassoon, A., An, F., Simko, J. P., and Liu, B. (2006) Identification of clinically significant tumor antigens by selecting phage antibody library on tumor cells in situ using laser capture microdissection. Mol. Cell. Proteomics. 5, 2364-2373). Full-length human IgG molecules derived from macropinocytosing phage antibodies retained the ability to internalize via macropinocytosis, validating our screening strategy. The target antigen for a cross-species binding antibody with a highly active macropinocytosis activity was identified as ephrin type-A receptor 2. Antibody-toxin conjugates created using this macropinocytosing IgG were capable of potent and receptor-dependent killing of a panel of EphA2-positive tumor cell lines in vitro. These studies identify novel methods to screen for and validate antibodies capable of receptor-dependent macropinocytosis, allowing further exploration of this highly efficient and tumor-selective internalization pathway for targeted therapy development.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Immunoglobulin G/pharmacology , Peptide Library , Receptor, EphA2/immunology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/metabolism , Antibody Affinity , Antibody Specificity , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Biomarkers/metabolism , Cell Line , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , HEK293 Cells , High-Throughput Screening Assays , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunotoxins/chemistry , Immunotoxins/immunology , Laser Capture Microdissection , Molecular Targeted Therapy , Pinocytosis , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Ribosome Inactivating Proteins, Type 1/chemistry , Ribosome Inactivating Proteins, Type 1/immunology , Saporins
6.
Bioconjug Chem ; 26(10): 2085-96, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26340339

ABSTRACT

Antibody-drug conjugates (ADCs) have emerged as an important class of therapeutics for cancer treatment that combine the target specificity of antibodies with the killing activity of anticancer chemotherapeutics. Early conjugation technologies relied upon random conjugation to either lysine or cysteine residues, resulting in heterogeneous ADCs. Recent technology advancements have resulted in the preparation of homogeneous ADCs through the site-specific conjugation at engineered cysteines, glycosylated amino acids, and bioorthogonal unnatural amino acids. Here we describe for the first time the conjugation of an anti-mitotic drug to an antibody following the mild and selective oxidation of a serine residue engineered at the N-terminus of the light chain. Using an alkoxyamine-derivatized monomethyl auristatine E payload, we have prepared a hydrolytically stable ADC that retains binding to its antigen and displays potent in vitro cytotoxicity and in vivo tumor growth inhibition.


Subject(s)
Antibodies/chemistry , Antibodies/pharmacology , Protein Engineering/methods , Animals , Antibodies/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Hydrolysis , Mice, Nude , Oximes/chemistry , Protein Stability , Rats , Receptor, EphA2/immunology , Receptor, EphA2/metabolism , Serine/chemistry , Xenograft Model Antitumor Assays
7.
Mol Pharm ; 12(9): 3490-501, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26176328

ABSTRACT

Targeting Eph (erythropoietin producing hepatoma) receptors with monoclonal antibodies is being explored as therapy for several types of cancer. To test whether simultaneous targeting of EphA2, EphA4, and EphB4 would be an effective approach to cancer therapy, we generated a recombinant trispecific antibody using the variable domain genes of anti-EphA2, anti-EphA4, and anti-EphB4 monoclonal antibodies. A multidisciplinary approach combining biochemical, biophysical, and cellular-based assays was used to characterize the trispecific antibody in vitro and in vivo. Here we demonstrate that the trispecific antibody is expressed at high levels by mammalian cells, monodispersed in solution, thermostable, capable of simultaneously binding the three receptors, and able to activate the three targets effectively as evidenced by receptor internalization and degradation both in vitro and in vivo. Furthermore, pharmacokinetic analysis using tumor-bearing nude mice showed that the trispecific antibody remains in the circulation similarly to its respective parental antibodies. These results indicate that simultaneous blockade of EphA2, EphA4, and EphB4 could be an attractive approach to cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens/immunology , Drug Design , Pancreatic Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA4/antagonists & inhibitors , Receptor, EphB4/antagonists & inhibitors , Animals , Calorimetry, Differential Scanning , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Male , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Receptor, EphA2/immunology , Receptor, EphA4/immunology , Receptor, EphB4/immunology , Surface Plasmon Resonance , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Growth Factors ; 32(6): 214-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25494541

ABSTRACT

Recombinant antibody phage library technology provides multiple advantages, including that human antibodies can be generated against proteins that are highly conserved between species. We used this technology to isolate and characterize an anti-EphA2 single-chain antibody. We show that the antibody binds the antigen with 1:1 stoichiometry and has high specificity for EphA2. The crystal structure of the complex reveals that the antibody targets the same receptor surface cavity as the ephrin ligand. Specifically, a lengthy CDR-H3 loop protrudes deep into the ligand-binding cavity, with several hydrophobic residues at its tip forming an anchor-like structure buried within the hydrophobic Eph pocket, in a way similar to the ephrin receptor-binding loop in the Eph/ephrin structures. Consequently, the antibody blocks ephrin binding to EphA2. Furthermore, it induces apoptosis and reduces cell proliferation in lymphoma cells lines. Since Ephs are important mediators of tumorigenesis, such antibodies could have applications both in research and therapy.


Subject(s)
Receptor, EphA2/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antibody Affinity , Binding Sites, Antibody , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , Humans , Molecular Sequence Data , Single-Chain Antibodies/chemistry
9.
Invest New Drugs ; 31(1): 77-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22370972

ABSTRACT

BACKGROUND: Targeting the cell-surface receptor EphA2, which is highly expressed in some solid tumors, is a novel approach for cancer therapy. We aimed to evaluate the safety profile, maximum tolerated dose (MTD), pharmacokinetics, and antitumor activity of MEDI-547, an antibody drug conjugate composed of the cytotoxic drug auristatin (toxin) linked to a human anti-EphA2 monoclonal antibody (1C1), in patients with solid tumors relapsed/refractory to standard therapy. METHODS: In this phase 1, open-label study with planned dose-escalation and dose-expansion cohorts, patients received a 1-h intravenous infusion of MEDI-547 (0.08 mg/kg) every 3 weeks. RESULTS: Six patients received 0.08 mg/kg; all discontinued treatment. Dose escalation was not pursued. The study was stopped before cohort 2 enrollment due to treatment-related bleeding and coagulation events (hemorrhage-related, n = 3; epistaxis, n = 2). Therefore, lower doses were not explored and an MTD could not be selected. The most frequently reported treatment-related adverse events (AEs) were increased liver enzymes, decreased hemoglobin, decreased appetite, and epistaxis. Three patients (50%) experienced treatment-related serious AEs, including conjunctival hemorrhage, pain (led to study drug discontinuation), liver disorder, and hemorrhage. Best response included progressive disease (n = 5; 83.3%) and stable disease (n = 1; 16.7%). Minimal or no dissociation of toxin from 1C1 conjugate occurred in the blood. Serum MEDI-547 concentrations decreased rapidly, ~70% by 3 days post-dose. No accumulation of MEDI-547 was observed at 0.08 mg/kg upon administration of a second dose 3 weeks following dose 1. CONCLUSIONS: The safety profile of MEDI-547 does not support further clinical investigation in patients with advanced solid tumors.


Subject(s)
Aminobenzoates/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Aged , Aminobenzoates/adverse effects , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm , Epistaxis/chemically induced , Female , Hemorrhage/chemically induced , Humans , Middle Aged , Neoplasms/metabolism , Receptor, EphA2/immunology
10.
Cell Immunol ; 272(1): 102-6, 2011.
Article in English | MEDLINE | ID: mdl-22032907

ABSTRACT

EphA2 is a receptor tyrosine kinase and can be acted as an attractive antigen for glioma vaccines. In addition, LIGHT plays an important role on enhancing T cell proliferation and cytokine production. To improve the CTL mediated immune response against glioma cells, we prepared the novel vaccine containing EphA2(883-891) peptide (TLADFDPRV) and LIGHT plasmid and utilized it to immunize the HLA-A2 transgenic HHD mice. In addition, trimera mice were immunized with the novel vaccine to elicit the antitumor immune response. The results demonstrated that the novel vaccine could induce robust cellular immunity against glioma U251 cells without lysing autologous lymphocytes. Moreover, the novel vaccine could significantly inhibit the tumor growth and prolong the life span of tumor bearing mice. These findings suggested that the novel vaccine containing EphA2 epitope and LIGHT plasmid could induce anti-tumor immunity against U251 cells expressing EphA2, and provided a promising strategy for glioma immunotherapy.


Subject(s)
Cancer Vaccines/administration & dosage , Central Nervous System Neoplasms/prevention & control , Glioma/prevention & control , Immunity, Cellular/drug effects , Immunization , Immunotherapy/methods , Oligopeptides/pharmacology , Receptor, EphA2/immunology , T-Lymphocytes, Cytotoxic/drug effects , Amino Acid Sequence , Animals , COS Cells , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/pathology , Chlorocebus aethiops , Enzyme-Linked Immunospot Assay , Epitopes/immunology , Glioma/immunology , Glioma/mortality , Glioma/pathology , HLA-A2 Antigen/analysis , HLA-A2 Antigen/immunology , Immunity, Cellular/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/immunology , Plasmids/chemistry , Plasmids/immunology , Receptor, EphA2/chemistry , Survival Rate , T-Lymphocytes, Cytotoxic/immunology
11.
Cancer Gene Ther ; 28(3-4): 321-334, 2021 04.
Article in English | MEDLINE | ID: mdl-32873870

ABSTRACT

Chimeric Antigen Receptor (CAR) T-cell therapy, as an approved treatment option for patients with B cell malignancies, demonstrates that genetic modification of autologous immune cells is an effective anti-cancer regimen. Erythropoietin-producing Hepatocellular receptor tyrosine kinase class A2 (EphA2) is a tumour associated antigen expressed on a range of sarcomas, including paediatric osteosarcoma (OS) and Ewing sarcoma (ES). We tested human EphA2 directed CAR T cells for their capacity to target and kill human OS and ES tumour cells using in vitro and in vivo assays, demonstrating that EphA2 CAR T cells have potent anti-tumour efficacy in vitro and can eliminate established OS and ES tumours in vivo in a dose and delivery route dependent manner. Next, in an aggressive metastatic OS model we demonstrated that systemically infused EphA2 CAR T cells can traffic to and eradicate tumour deposits in murine livers and lungs. These results support further pre-clinical evaluation of EphA2 CAR T cells to inform the design of early phase clinical trial protocols to test the feasibility and safety of this immune cell therapy in paediatric bone sarcoma patients.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy, Adoptive/methods , Receptor, EphA2/immunology , T-Lymphocytes/immunology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Female , Humans , Mice , Mice, Inbred NOD , Molecular Targeted Therapy
12.
Sci Immunol ; 6(61)2021 07 30.
Article in English | MEDLINE | ID: mdl-34330813

ABSTRACT

Human γδ T cells contribute to tissue homeostasis and participate in epithelial stress surveillance through mechanisms that are not well understood. Here, we identified ephrin type-A receptor 2 (EphA2) as a stress antigen recognized by a human Vγ9Vδ1 TCR. EphA2 is recognized coordinately by ephrin A to enable γδ TCR activation. We identified a putative TCR binding site on the ligand-binding domain of EphA2 that was distinct from the ephrin A binding site. Expression of EphA2 was up-regulated upon AMP-activated protein kinase (AMPK)-dependent metabolic reprogramming of cancer cells, and coexpression of EphA2 and active AMPK in tumors was associated with higher CD3 T cell infiltration in human colorectal cancer tissue. These results highlight the potential of the human γδ TCR to cooperate with a co-receptor to recognize non-MHC-encoded proteins as signals of cellular dysregulation, potentially allowing γδ T cells to sense metabolic energy changes associated with either viral infection or cancer.


Subject(s)
AMP-Activated Protein Kinases/immunology , Antigens/immunology , Intraepithelial Lymphocytes/immunology , Neoplasms/immunology , Receptor, EphA2/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , AMP-Activated Protein Kinases/genetics , Animals , Antibodies, Monoclonal/immunology , Cell Line , Humans , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics
13.
Bioengineered ; 12(2): 10379-10400, 2021 12.
Article in English | MEDLINE | ID: mdl-34709992

ABSTRACT

Enhanced EphA2 expression is observed in a variety of epithelial-derived malignancies and is an important target for anti-tumor therapy. Currently, Therapeutic monoclonal antibodies against immune checkpoints have shown good efficacy for tumor treatment. In this study, we constructed an immune single-chain fragment variable (scFv) library using peripheral blood mononuclear cells (PBMCs) from 200 patients with a variety of malignant tumors. High affinity scFvs against EphA2 can be easily screened from the immune library using phage display technology. Anti-EphA2 scFvs can be modified into any form of recombinant antibody, including scFv-Fc and full-length IgG1 antibodies, and the recombinant antibody affinity was improved following modification. Among the modified anti-EphA2 antibodies the affinity of 77-IgG1 was significantly increased, reaching a pmol affinity level (10-12). We further demonstrated the binding activity of recombinant antibodies to the EphA2 protein, tumor cells, and tumor tissues using macromolecular interaction techniques, flow cytometry and immunohistochemistry. Most importantly, both the constructed scFvs-Fc, as well as the IgG1 antibodies against EphA2 were able to inhibit the growth of tumor cells to some extent. These results suggest that the immune libraries from patients with malignant tumors are more likely to screen for antibodies with high affinity and therapeutic effect. The constructed fully human scFv immune library has broad application prospects for specific antibody screening. The screened scFv-Fc and IgG1 antibodies against EphA2 can be used for the further study of tumor immunotherapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Peptide Library , Receptor, EphA2/immunology , Recombinant Proteins/pharmacology , Antigens, Surface/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Surface Display Techniques , Gene Expression Regulation, Neoplastic , Homologous Recombination/genetics , Humans , Immunoglobulin G , Neoplasms/genetics , Neoplasms/pathology , Prognosis , Protein Binding/drug effects , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Single-Chain Antibodies/immunology , Survival Analysis
14.
Cancer Immunol Immunother ; 59(5): 759-67, 2010 May.
Article in English | MEDLINE | ID: mdl-19943047

ABSTRACT

The prognosis of liver cancer remains poor, but recent advances in nanotechnology offer promising possibilities for cancer treatment. Novel adjuvant, amphiphilic nanoparticles (NPs) composed of L: -phenylalanine (Phe)-conjugated poly(gamma-glutamic acid) (gamma-PGA-Phe NPs) having excellent capacity for carrying peptides, were found to have the potential for use as a peptide vaccine against tumor models overexpressing artificial antigens, such as ovalbumin (OVA). However, the anti-tumor potential of gamma-PGA-Phe NPs vaccines using much less immunogenic tumor-associated antigen (TAA)-derived peptide needs to be clarified. In this study, we evaluated the effectiveness of immunization with EphA2, recently identified TAA, derived peptide-immobilized gamma-PGA-Phe NPs (Eph-NPs) against mouse liver tumor of MC38 cells (EphA2-positive colon cancer cells). Immunization of normal mice with Eph-NPs resulted in generation of EphA2-specific type-1 CD8+ T cells. Immunization with Eph-NPs tended to provide a degree of anti-MC38 liver tumor protection more than that observed for immunization with the mixture of EphA2-derived peptide and complete Freund's adjuvant (Eph + CFA). Neither Eph-NPs nor Eph + CFA vaccines inhibited tumor growth of BL6, EphA2-negative melanoma cells. Splenocytes isolated from MC38-bearing mice treated with Eph-NPs showed strong and specific cytotoxic activity against MC38 cells. Immunization with Eph + CFA induced liver damage as evidenced by elevation of serum alanine aminotransferase, while Eph-NPs vaccination did not exhibit any toxic damage to the liver. These results demonstrated that immunization with Eph-NPs displayed anti-tumor effects against liver tumor by generating acquired immunity equivalent to the toxic adjuvant CFA, suggesting that safe gamma-PGA-Phe NPs could be applied clinically for the vaccine treatment of liver cancer.


Subject(s)
Cancer Vaccines/immunology , Liver Neoplasms/therapy , Nanoparticles , Polyglutamic Acid/analogs & derivatives , Receptor, EphA2/immunology , Adjuvants, Immunologic , Animals , Cancer Vaccines/administration & dosage , Female , Freund's Adjuvant , Mice , Mice, Inbred C57BL , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/immunology , Surface-Active Agents/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
15.
J Immunol ; 181(11): 7721-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017961

ABSTRACT

The EphA2 receptor tyrosine kinase is an attractive therapeutic target that is commonly overexpressed on solid tumors, with the degree of overexpression associated with disease progression, metastatic potential, and poor prognosis. Agonistic mAbs or ligand (ephrinA1)-Fc fusion protein are capable of inducing EphA2 internalization and degradation, thereby (at least transiently) eliminating the influence of this oncoprotein. We and others have also shown that EphA2 contains multiple peptide epitopes that can be recognized by effector CD4(+) and CD8(+) T cells isolated from tumor-bearing patients. Herein, we show that "agonist" reagents that trigger the proteasome-dependent degradation of tumor cell EphA2 result in the improved presentation of peptides derived from (both the extracellular and intracellular domains of) EphA2 in MHC class I complexes expressed on the tumor cell membrane for at least 48 h, as manifested by increased recognition by EphA2-specific CD8(+) T cells in vitro. We also observed that while delivery of ephrinA1-Fc fusion protein or agonist mAb into EphA2(+) tumor lesions promotes EphA2 degradation in situ, this single administration of agent does not dramatically alter tumor progression in a humanized SCID model. However, when combined with the adoptive transfer of normally nontherapeutic (human) anti-EphA2 CD8(+) CTL, this dual-agent regimen results in complete tumor eradication. These results suggest that strategies targeting the conditional proteasome-mediated destruction of tumor cell EphA2 may enable EphA2-specific CD8(+) T cells (of modest functional avidity) to realize improved therapeutic potential.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Neoplasm Proteins/immunology , Peptides/immunology , Prostatic Neoplasms/immunology , Receptor, EphA2/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Antigen Presentation/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Prostatic Neoplasms/therapy , Proteasome Endopeptidase Complex/immunology , Protein Structure, Tertiary , Receptor, EphA2/antagonists & inhibitors
16.
Article in English | MEDLINE | ID: mdl-20516612

ABSTRACT

The recombinant N-terminal domain of human ephrin type-A receptor 2 (rEphA2) has been crystallized in complex with the recombinantly produced Fab fragment of a fully human antibody (1C1; IgG1/kappa). These are the first reported crystals of an ephrin receptor bound to an antibody. The orthorhombic crystals belonged to space group C222(1) (the 00l reflections obey the l = 2n rule), with unit-cell parameters a = 78.93, b = 120.79, c = 286.20 A. The diffraction of the crystals extended to 2.0 A resolution. However, only data to 2.55 A resolution were considered to be useful owing to spot overlap caused by the long unit-cell parameter. The asymmetric unit is most likely to contain two 1C1 Fab-rEphA2 complexes. This corresponds to a crystal volume per protein weight (V(M)) of 2.4 A(3) Da(-1) and a solvent content of 49.5%. The three-dimensional structure of this complex will shed light on the molecular basis of 1C1 specificity. This will also contribute to a better understanding of the mechanism of action of this antibody, the current evaluation of which as an antibody-drug conjugate in cancer therapy makes it a particularly interesting case study.


Subject(s)
Antigen-Antibody Complex/chemistry , Immunoglobulin Fab Fragments/chemistry , Peptide Fragments/chemistry , Receptor, EphA2/chemistry , Antigen-Antibody Complex/immunology , Crystallization , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/immunology , Peptide Fragments/immunology , Receptor, EphA2/immunology
17.
Cancer Res ; 67(8): 3927-35, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440108

ABSTRACT

The EphA2 receptor tyrosine kinase is frequently overexpressed and functionally altered in malignant cells and thus provides opportunities for selective targeting of tumor cells. We describe here the development of a novel, bispecific single-chain antibody (bscAb) referred to as bscEphA2xCD3. This molecule simultaneously targets EphA2 on tumor cells and the T-cell receptor/CD3 complex on T cells and possesses structural and functional characteristics of the recently developed BiTE technology. An EphA2-specific single-chain antibody was selected for recognition of an epitope that is preferentially exposed on malignant cells based on the concept of epitope exclusion; this was fused to a CD3-specific single-chain antibody to generate bscEphA2xCD3. The resultant bscAb redirected unstimulated human T cells to lyse EphA2-expressing tumor cells both in vitro and in vivo. In separate experiments, efficient tumor cell lysis was achieved in vitro at drug concentrations

Subject(s)
Antibodies, Bispecific/pharmacology , CD3 Complex/immunology , Immunization, Passive/methods , Neoplasms/therapy , Receptor, EphA2/immunology , Animals , Antibodies, Bispecific/immunology , Antibody Specificity , CHO Cells , Cell Growth Processes/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Epitopes/immunology , Humans , Intercellular Junctions/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
18.
Mol Cancer Ther ; 18(6): 1092-1103, 2019 06.
Article in English | MEDLINE | ID: mdl-30962321

ABSTRACT

Cell-type-specific intracellular payload delivery is desired for antibody-based-targeted therapy development. However, tumor-specific internalizing antigens are rare to find, and even rarer for those that are expressed at uniformly high levels. We constructed a bispecific antibody that is composed of a rapidly internalizing antibody binding to a tumor-associated antigen, ephrin receptor A2 (EphA2), and a noninternalizing antibody binding to a highly expressed tumor-associated antigen, activated leukocyte cell adhesion molecule (ALCAM). We found that the overall internalization property of the bispecific is profoundly impacted by the relative surface expression level (antigen density ratio) of EphA2 versus ALCAM. When the EphA2-to-ALCAM ratio is greater than a threshold level (1:5), the amount of the bispecific taken into the tumor cell exceeds what is achieved by either the monoclonal internalizing antibody or a mixture of the two antibodies, showing a bispecific-dependent amplification effect where a small amount of the internalizing antigen EphA2 induces internalization of a larger amount of the noninternalizing antigen ALCAM. When the ratio is below the threshold, EphA2 can be rendered noninternalizing by the presence of excess ALCAM on the same cell surface. We constructed a bispecific antibody-drug conjugate (ADC) based on the above bispecific design and found that the bispecific ADC is more potent than monospecific ADCs in tumor cell killing both in vitro and in vivo Thus, the internalizing property of a cell surface antigen can be manipulated in either direction by a neighboring antigen, and this phenomenon can be exploited for therapeutic targeting.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Neoplasms/therapy , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules, Neuronal/immunology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Survival/drug effects , Female , Fetal Proteins/immunology , Fetal Proteins/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , PC-3 Cells , Receptor, EphA2/immunology , Receptor, EphA2/metabolism , Transduction, Genetic , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
19.
Int Immunopharmacol ; 72: 176-185, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30986645

ABSTRACT

Eph receptor tyrosine kinases have a wide range of biological functions and have gradually been recognized increasingly as key regulators of inflammation and injury diseases. Although previous studies suggested that EphA2 receptor may be involved in the regulation of inflammation and vascular permeability in injured lung, the detailed effects of EphA2 on LPS-induced acute lung injury (ALI) are still inadequate and the underlying mechanism remains poorly understood. In this study, we detected the effects of EphA2 antagonism on inflammation, pulmonary vascular permeability and oxidative stress in LPS-induced ALI and investigate the potential mechanism. Our results showed that EphA2 antagonism markedly inhibited the cytokines release and inflammatory cells infiltration in BALF, prevented the LPS-induced elevations of MPO activity and MDA level in lung tissues. Our study also found that EphA2 antagonism significantly decreased the wet/dry ratios, reduced the Evans blue albumin extravasation in lung tissues and obviously alleviated the LPS-induced increment of pulmonary vascular permeability. Mechanistically, EphA2 antagonism significantly increased the activation of Nrf2 along with its target antioxidant enzyme HO-1 and inhibited the expressions of TLR4/MyD88 in lung tissues and A549 alveolar epithelial cells. Furthermore, EphA2 antagonism dramatically inhibited the LPS-evoked activations of RhoA/ROCK in lung tissues. In conclusion, our data indicate that EphA2 receptor plays an essential role in LPS-induced ALI and EphA2 antagonism has protective effects against LPS-induced ALI via Nrf2/HO-1, TLR4/MyD88 and RhoA/ROCK pathways. These results suggest that antagonism of EphA2 may be an effective therapeutic strategy for the treatment of ALI.


Subject(s)
Acute Lung Injury/drug therapy , Antibodies, Monoclonal/therapeutic use , Receptor, EphA2/antagonists & inhibitors , A549 Cells , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Antibodies, Monoclonal/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Humans , Lipopolysaccharides , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Rats, Sprague-Dawley , Receptor, EphA2/immunology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
20.
Mol Immunol ; 44(11): 3049-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17241664

ABSTRACT

We report here the humanization of two mouse monoclonal antibodies (mAb) using framework shuffling of human germline genes. mAbs EA2 and 47 were raised against the human receptor tyrosine kinase EphA2 and EphB4, respectively, which exhibit increased expression levels in many cancer cell lines. One- and two-step strategies were carried out, in which the light and heavy chains of each parental mAb were simultaneously or sequentially humanized. We characterized in detail these newly humanized antibodies in terms of binding affinity to their respective antigen, functional activity, thermostability, electric charge and expression yields. Three previously described framework shuffled, humanized versions of another mouse anti-human EphA2 antibody (mAb B233) were similarly characterized. We show that several of these parameters were either maintained or improved in all humanized molecules when compared with their respective chimaeric counterpart. Therefore, this humanization approach is generally applicable to non-human IgGs and allows for the specific selection of antibodies and antibody fragments exhibiting favorable functional, biochemical and biophysical properties.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Receptor, EphA2/immunology , Receptor, EphB4/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antigen-Antibody Reactions , Cell Line, Tumor , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Mice , Molecular Sequence Data , Sequence Analysis , Structure-Activity Relationship , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL