Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(22): 15443-15456, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36331527

ABSTRACT

Overexpression of the receptor tyrosine kinase EphA2 is invariably associated with poor prognosis and development of aggressive metastatic cancers. Guided by our recently solved X-ray structure of the complex between an agonistic peptide and EphA2-LBD, we report on a novel agent, targefrin, that binds to EphA2-LBD with a 21 nM dissociation constant by isothermal titration calorimetry and presents an IC50 value of 10.8 nM in a biochemical assay. In cell-based assays, a dimeric version of the agent is as effective as the natural dimeric ligands (ephrinA1-Fc) in inducing cellular receptor internalization and degradation in several pancreatic cancer cell lines. When conjugated with chemotherapy, the agents can effectively deliver paclitaxel to pancreatic cancers in a mouse xenograft study. Given the pivotal role of EphA2 in tumor progression, we are confident that the agents reported could be further developed into innovative EphA2-targeting therapeutics.


Subject(s)
Peptides , Receptor, EphA2 , Animals , Humans , Mice , Cell Line , Ligands , Peptides/pharmacology , Receptor Protein-Tyrosine Kinases , Receptor, EphA2/drug effects , Receptor, EphA2/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
2.
Invest New Drugs ; 40(4): 747-755, 2022 08.
Article in English | MEDLINE | ID: mdl-35404015

ABSTRACT

Ephrin type-A 2 (EphA2) is a transmembrane receptor expressed in epithelial cancers. We report on a phase I dose escalation and biodistribution study of DS-8895a, an anti-EphA2 antibody, in patients with advanced EphA2 positive cancers. DS-8895a was administered at 1, 3, 10 or 20 mg/kg every 2 weeks to determine safety, pharmacokinetics and anti-tumor efficacy. All patients underwent 89Zr trace-labelled infusion of DS-8895a (89Zr-DS-8995a) positron emission tomography imaging to determine the biodistribution of DS-8895a, and correlate findings with EphA2 expression, receptor saturation and response. Nine patients were enrolled on study. Of patients enrolled, seven patients received at least one infusion of DS-8895a: four patients received 1 mg/kg dose (Cohort 1) and three patients received 3 mg/kg dose (Cohort 2). Median age was 67.0 years (range 52-81), majority male (71%), and median number of prior systemic therapies was three (range 0-8). The primary cancer diagnosis was colorectal cancer (two patients) and one patient each had gastric, head and neck, high-grade serous adenocarcinoma, lung, and pancreatic cancers. No dose-limiting toxicities or treatment-related adverse events reported. The best response for the patients in Cohort 1 was stable disease and in Cohort 2 was progressive disease. 89Zr-DS-8895a demonstrated no normal tissue uptake and specific low-grade uptake in most tumours. DS-8895a had limited therapeutic efficacy at doses evaluated and 89Zr-DS-8895a demonstrated low tumour uptake. The biodistribution data from this study were key in halting further development of DS-8895a, highlighting the importance of biodistribution studies in drug development. (Trial registration: ClinicalTrials.gov Identifier NCT02252211).


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Neoplasms , Aged , Aged, 80 and over , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Ephrin-A2/immunology , Humans , Male , Middle Aged , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Receptor, EphA2/drug effects , Tissue Distribution
3.
Nat Microbiol ; 3(2): 1-8, 2018 02.
Article in English | MEDLINE | ID: mdl-29292383

ABSTRACT

Epstein-Barr virus (EBV) is causally associated with nasopharyngeal carcinoma, 10% of gastric carcinoma and various B cell lymphomas 1 . EBV infects both B cells and epithelial cells 2 . Recently, we reported that epidermal growth factor and Neuropilin 1 markedly enhanced EBV entry into nasopharyngeal epithelial cells 3 . However, knowledge of how EBV infects epithelial cells remains incomplete. To understand the mechanisms through which EBV infects epithelial cells, we integrated microarray and RNA interference screen analyses and found that Ephrin receptor A2 (EphA2) is important for EBV entry into the epithelial cells. EphA2 short interfering RNA knockdown or CRISPR-Cas9 knockout markedly reduced EBV epithelial cell infection, which was mostly restored by EphA2 complementary DNA rescue. EphA2 overexpression increased epithelial cell EBV infection. Soluble EphA2 protein, antibodies against EphA2, soluble EphA2 ligand EphrinA1, or the EphA2 inhibitor 2,5-dimethylpyrrolyl benzoic acid efficiently blocked EBV epithelial cell infection. Mechanistically, EphA2 interacted with EBV entry proteins gH/gL and gB to facilitate EBV internalization and fusion. The EphA2 Ephrin-binding domain and fibronectin type III repeats domain were essential for EphA2-mediated EBV infection, while the intracellular domain was dispensable. This is distinct from Kaposi's sarcoma-associated herpesvirus infection through EphA2 4 . Taken together, our results identify EphA2 as a critical player for EBV epithelial cell entry.


Subject(s)
Ephrin-A2/metabolism , Epithelial Cells/virology , Herpesvirus 4, Human/pathogenicity , Receptor, EphA2/metabolism , Virus Internalization , Animals , Benzoates/antagonists & inhibitors , CHO Cells , CRISPR-Cas Systems , Cricetulus , Gene Knockdown Techniques , HEK293 Cells , Humans , Membrane Glycoproteins , Molecular Chaperones , RNA Interference , Receptor, EphA2/drug effects , Receptor, EphA2/genetics , Viral Proteins
4.
J Periodontal Res ; 52(5): 913-921, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28590061

ABSTRACT

OBJECTIVE: EphrinA2-EphA2 and ephrinB2-EphB4 critically engage in bidirectional signalling to modulate alveolar bone remodelling. The present study aimed to investigate the effects of lipopolysaccharides (LPS) derived from Porphyromonas gingivalis on ephrin/Eph signalling in periodontal ligament fibroblasts (PDLFs). MATERIAL AND METHODS: The primary cultured PDLFs were incubated in the absence (as a control) or presence of P. gingivalisLPS at 0.001-10 µg/mL for 24 hours. The PDLFs were then stimulated with P. gingivalisLPS at the optimal concentration (0.1 µg/mL) for different periods (6-48 hours). The expression of ephrinA2, ephrinB2, EphA2 and EphB4 was assessed by quantitative reverse-transcription real-time polymerase chain reaction and western blotting. The osteoblastic markers alkaline phosphatase, osteocalcin and Runt-related transcription factor 2 (Runx2), and the osteoclastogenesis-related factors receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin were also evaluated. RESULTS: The ephrinA2 and EphA2 expression was upregulated and EphB4 expression was downregulated by stimulation of P. gingivalisLPS. EphrinA2 mRNA expression in the PDLFs was significantly upregulated from 12 to 48 hours (P<.05), whereas EphA2 exhibited no change for the first 24 hours, after which there was a significant increase at 48 hours (P<.05). EphB4 exhibited lower mRNA expression at 12 and 24 hours than did the control (P<.05), but the change was insignificant at 48 hours. In contrast, the expression of ephrinB2 remained unchanged. The expressions of ephrinA2, EphA2, ephrinB2 and EphB4 at the protein level showed a similar pattern to that at the mRNA level. The expression of Runx2 and osteocalcin significantly decreased, whereas that of RANKL/osteoprotegerin increased. CONCLUSION: The present study suggest that P. gingivalisLPS would contribute to a dysregulation of bone remodelling, whereby ephrinA2/EphA2 expression is stimulated and EphB4 expression is inhibited.


Subject(s)
Ephrins/drug effects , Fibroblasts/drug effects , Lipopolysaccharides/pharmacology , Periodontal Ligament/drug effects , Porphyromonas gingivalis/metabolism , Receptors, Eph Family/drug effects , Adolescent , Alkaline Phosphatase , Bone Remodeling/drug effects , Cell Culture Techniques , Child , Core Binding Factor Alpha 1 Subunit , Ephrin-A2/drug effects , Ephrin-A2/genetics , Ephrin-B2/drug effects , Ephrin-B2/genetics , Ephrins/genetics , Gene Expression Regulation , Humans , Osteoblasts , Osteocalcin , Osteogenesis , Osteoprotegerin , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptor, EphA2/drug effects , Receptor, EphA2/genetics , Receptor, EphB4/drug effects , Receptor, EphB4/genetics , Receptors, Eph Family/genetics
5.
Expert Opin Ther Targets ; 15(1): 31-51, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21142802

ABSTRACT

IMPORTANCE OF THE FIELD: High mortality rates with cancers warrant further development of earlier diagnostics and better treatment strategies. Membrane-bound erythropoietin-producing hepatocellular receptor tyrosine kinase class A2 (EphA2) is overexpressed in breast, prostate, urinary bladder, skin, lung, ovary and brain cancers. AREAS COVERED IN THIS REVIEW: EphA2 overexpression in cancers, its signaling mechanisms and strategies to target its deregulation. WHAT THE READER WILL GAIN: High EphA2 expression in cancer cells is correlated with a poor prognosis associated with recurrence due to enhanced metastasis. Interaction of the EphA2 receptor with its ligand (e.g., ephrinA1) triggers events that are deregulated and implicated in carcinogenesis. EphrinA1-independent oncogenic activity and ephrinA1-dependent tumor suppressor roles for EphA2 are described. Molecular interactions of EphA2 with signaling proteins are associated with the modulation of cytoskeleton dynamics, cell adhesion, proliferation, differentiation and metastasis. The deregulated signaling by EphA2 and its involvement in oncogenesis provide multiple avenues for the rational design of intervention approaches. TAKE HOME MESSAGE: EphA2 has been tested as a drug target using multiple approaches such as agonist antibodies, RNA interference, immunotherapy, virus vector-mediated gene transfer, small-molecule inhibitors and nanoparticles. With over a decade of research, encouraging results with targeting of EphA2 expression in various pre-clinical cancer models necessitate further studies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Receptor, EphA2/drug effects , Animals , Drug Delivery Systems , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neoplasm Recurrence, Local , Neoplasms/physiopathology , Prognosis , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Signal Transduction
6.
In Vivo ; 24(4): 393-400, 2010.
Article in English | MEDLINE | ID: mdl-20668305

ABSTRACT

BACKGROUND: The aim of this study was to determine the effects of soy phytoestrogens on the methylation of promoter genes in prostate tumors. The incidence of prostate cancer in Asia is thirty percent lower than in Western countries. Since soy phytoestrogens represent a large portion of the Asian diet, evidence suggests their protective effect against prostate cancer. MATERIALS AND METHODS: In three human prostate cancer cell lines, methylation-specific-PCR was used to determine the effect of soy isoflavones (genistein and daidzein), compared to known demethylating agent 5-azacytidine as control in the promoter regions of glutathione S-transferase P1 (GSTP1), Ras association domain family 1 (RASSF1A), ephrin B2 (EPHB2) and breast cancer 1 (BRCA1) genes. In parallel, immunohistochemistry was used to assess the effects of genistein, daidzein and 5-azacytidine treatment on the corresponding protein expression. RESULTS: All studied promoters, with the exception of that for BRCA1, were strongly methylated without treatment. After treatment by phytoestrogens, demethylation of GSTP1 and EPHB2 promoter regions was observed and an increase in their protein expression was demonstrated by immunohistochemistry. CONCLUSION: Epigenetic modifications of DNA, such as the promoter CpG island demethylation of tumor suppressor genes, might be related to the protective effect of soy on prostate cancer.


Subject(s)
BRCA1 Protein/genetics , DNA Methylation/drug effects , Genes, BRCA1/drug effects , Glutathione S-Transferase pi/genetics , Phytoestrogens/pharmacology , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Receptor, EphA2/genetics , Tumor Suppressor Proteins/genetics , BRCA1 Protein/drug effects , Cell Line, Tumor , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glutathione S-Transferase pi/drug effects , Humans , Immunohistochemistry , Male , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/pathology , Receptor, EphA2/drug effects , Glycine max , Tumor Suppressor Proteins/drug effects
7.
Mol Cancer Ther ; 6(12 Pt 1): 3208-18, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18089715

ABSTRACT

We have previously shown that the EphA2 receptor tyrosine kinase is overexpressed in glioblastoma multiforme (GBM) and represents a novel, attractive therapeutic target for the treatment of brain tumors. Here, we have developed an EphA2-targeted agent, ephrinA1-PE38QQR, a novel cytotoxin composed of ephrinA1, a ligand for EphA2, and PE38QQR, a mutated form of Pseudomonas aeruginosa exotoxin A. EphrinA1-PE38QQR showed potent and dose-dependent killing of GBM cells overexpressing the EphA2 receptor in cell viability and clonogenic survival assays, with an average IC(50) of approximately 10(-11) mol/L. The conjugate was also highly effective in killing breast and prostate cancer cells overexpressing EphA2. The cytotoxic effect of ephrinA1-PE38QQR was specific, as it was neutralized by an excess of EphA2 ligands. Moreover, normal human endothelial cells and breast cancer cells that do not overexpress EphA2, as well as GBM cells that have down-regulated EphA2, were not susceptible to the cytotoxin. EphrinA1-PE38QQR-mediated cytotoxicity induced caspase-dependent apoptosis, which was, however, not responsible for cell death in response to the conjugate. In addition, the conjugate elicited no changes in the activity of survival pathways such as phosphoinositide 3-kinase, measured by AKT phosphorylation. This is the first attempt to create a cytotoxic therapy using any of the ephrin ligands of either class (A or B) conjugated to a bacterial toxin. EphrinA1-PE38QQR is very potent and specific, produces cell death that is caspase independent, and forms the basis for the further development of clinically applicable EphA2-targeted cytotoxins.


Subject(s)
Cytotoxins/pharmacology , Ephrin-A1/chemistry , Receptor, EphA2/drug effects , Blotting, Western , Breast Neoplasms/pathology , Cell Line , Cell Survival , Humans , Male , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology
8.
Curr Cancer Drug Targets ; 5(3): 149-57, 2005 May.
Article in English | MEDLINE | ID: mdl-15892616

ABSTRACT

Eph receptors are a unique family of receptor tyrosine kinases (RTK) that play critical roles in embryonic patterning, neuronal targeting, and vascular development during normal embryogenesis. Eph RTKs and their ligands, the ephrins, are also frequently overexpressed in a variety of cancers and tumor cell lines. In particular, one family member, EphA2, is overexpressed in breast, prostate, lung, and colon cancers. Unlike traditional oncogenes that often function only in tumor cells, recent data show that Eph receptors mediate cell-cell interactions both in tumor cells and in the tumor microenvironment, namely the tumor stroma and tumor vasculature. Thus, EphA2 receptors are attractive targets for drug design, as targeting these molecules could simultaneously inhibit several aspects of tumor progression. This review focuses on the multiple roles of EphA2 in cancer progression, the mechanisms by which EphA2 inhibition may halt this progression, and the pre-clinical results of EphA2 inhibition in various cancer model systems.


Subject(s)
Antineoplastic Agents/pharmacology , Receptor, EphA2/drug effects , Animals , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Receptor, EphA2/biosynthesis
9.
Mol Cell Neurosci ; 24(3): 632-45, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14664814

ABSTRACT

Ephrin-As are repulsive axonal guidance cues that regulate retinotectal projection. EphA tyrosine kinases, which are the receptors of ephrin-As, activate signaling cascades leading to cytosckeleton reorganization. Here, we address the role of cyclin-dependent kinase (Cdk) 5 in Eph receptor signaling induced by ephrin-A5. Ephrin-A5 induced a cell morphological response in PC-3M cells that endogenously express Cdk5 and EphA2, a receptor for ephrin-A5. This response was augmented by the transfection of p35, which is a neuronal regulator of Cdk5. While the morphological response of native PC-3M cells was not affected by olomoucine, an inhibitor of Cdk, the response was inhibited in the p35-transfected cells. In retinal ganglion cells, either olomoucine at 20 microM or Y-27632 at 10 microM, an inhibitor of Rho-kinase/ROKalpha/ROCKII, showed maximum inhibitory effect against ephrin-A5 (10 microg/ml)-induced growth cone collapse. Combined application of olomoucine and Y-27632 further suppressed the ephrin-A5-induced response. Ephrin-A5 evoked phosphorylation of Cdk5 at Tyr15 and tau, a substrate of Cdk5 in retinal growth cones. Recombinant herpes simplex virus expressing Cdk5 mutant (kinase-negative or Tyr15 to Ala) showed a dominant-negative effect on the ephrin-A5-induced growth cone collapse. These findings demonstrate that both Cdk5 and the Rho kinase pathway independently contribute to the downstream of ephrin-A-induced signaling in retinal ganglion cells.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Ephrin-A5/metabolism , Growth Cones/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinal Ganglion Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Size/drug effects , Cell Size/physiology , Cells, Cultured , Chick Embryo , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Enzyme Inhibitors/pharmacology , Ephrin-A5/pharmacology , Growth Cones/drug effects , Growth Cones/ultrastructure , Humans , Intracellular Signaling Peptides and Proteins , Mutation/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, EphA2/drug effects , Receptor, EphA2/metabolism , Retina/cytology , Retina/embryology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Superior Colliculi/cytology , Superior Colliculi/embryology , Superior Colliculi/metabolism , Visual Pathways/cytology , Visual Pathways/embryology , Visual Pathways/metabolism , rho-Associated Kinases
10.
J Cancer Res Clin Oncol ; 129(12): 703-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14564515

ABSTRACT

PURPOSE: The EphA2 receptor protein tyrosine kinase gene has been shown to be over-expressed or functionally altered in a number of human tumors, including colon cancer, but little is known about the regulation of this new oncoprotein. In order to explore the mechanism of EphA2 up-regulation in cancer cells, we examined the change of expression of EphA2 gene induced by deoxycholic acid (DCA) and elucidated its possible pathways in human colon cancer cells. METHODS: Western blot and RT-PCR were used to assess the protein expression and messenger RNA in several colon cancer cell lines, which harbor various p53 status. The inhibition study to interfere the MAPK pathway was performed by using various chemicals and by transfecting dominant negative mutant plasmids. RESULTS: Up-regulation of EphA2 induced by DCA was observed in a dose- and time-dependent fashion both in mRNA and protein levels. This regulation is constant regardless of p53 status including wild, mutant or knocked out in the colon cell lines used. This induction was in part blocked by either erk1/2 inhibitors or dominant negative mutants erk1/2 plasmids. CONCLUSIONS: These results suggest that DCA induced up-regulation of EphA2 in colon cancer cells is due to activation of erk1/2 cascade, and is p53-independent. Taken together with the roles of EphA2 and DCA in tumorigenesis, which have been independently reported, our observation will provide a new mechanistic basis of DCA commitment in carcinogenesis.


Subject(s)
Carcinoma/metabolism , Colonic Neoplasms/metabolism , Deoxycholic Acid/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Receptor, EphA2/drug effects , Receptor, EphA2/metabolism , Blotting, Western , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitogen-Activated Protein Kinases/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
11.
Oncogene ; 21(50): 7690-9, 2002 Oct 31.
Article in English | MEDLINE | ID: mdl-12400011

ABSTRACT

Intracellular signaling by receptor tyrosine kinases regulates many different aspects of cell behavior. Recent studies in our laboratory and others have demonstrated that the EphA2 receptor tyrosine kinase critically regulates tumor cell growth, migration and invasiveness. Although the cellular consequences of EphA2 signaling have been the focus of recent attention, the biochemical changes that are triggered by ligand-mediated activation of EphA2 remain largely unknown. Herein, we demonstrate that ligand stimulation of EphA2 promotes the nucleus translocation and phosphorylation of ERK kinases, followed by an increase in nuclear induction of the Elk-1 transcription factor. Ligand-mediated activation allows EphA2 to form a molecular complex with the SHC and GRB2 adaptor proteins. Specifically, we demonstrate that tyrosine phosphorylated EphA2 interacts with the PTB and SH2 domains of SHC. We also show that the interaction of EphA2 with GRB2 is indirect and mediated by SHC and that this complex is necessary for EphA2-mediated activation of ERK kinases. These studies provide a novel mechanism to demonstrate how EphA2 can convey information from the cell exterior to the nucleus.


Subject(s)
Adaptor Proteins, Signal Transducing , DNA-Binding Proteins , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptor, EphA2/metabolism , Transcription Factors , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Enzyme Activation/drug effects , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-A1/pharmacology , Extracellular Matrix/metabolism , Female , GRB2 Adaptor Protein , Humans , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinases/drug effects , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, EphA2/drug effects , Receptor, EphA2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tumor Cells, Cultured , ets-Domain Protein Elk-1 , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...