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










Publication year range
1.
Blood Adv ; 2(23): 3492-3505, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30530776

ABSTRACT

In this study, we assessed the sensitivity of myeloma cells to the oncolytic measles virus (MV) in relation to p53 using 37 cell lines and 23 primary samples. We showed that infection and cell death were correlated with CD46 expression, which was associated with TP53 status; TP53 abn cell lines highly expressed CD46 and were preferentially infected by MV when compared with the TP53 wt cell lines (P = .046 and P = .045, respectively). Infection of myeloma cells was fully dependent on CD46 expression in both cell lines and primary cells. In the TP53 wt cell lines, but not the TP53 abn cell lines, activation of the p53 pathway with nutlin3a inhibited both CD46 expression and MV infection, while TP53 silencing reciprocally increased CD46 expression and MV infection. We showed using a p53 chromatin immunoprecipitation assay and microRNA assessment that CD46 gene expression was directly and indirectly regulated by p53. Primary myeloma cells overexpressed CD46 as compared with normal cells and were highly infected and killed by MV. CD46 expression and MV infection were inhibited by nutlin3a in primary p53-competent myeloma cells, but not in p53-deficient myeloma cells, and the latter were highly sensitive to MV infection. In summary, myeloma cells were highly sensitive to MV and infection inhibition by the p53 pathway was abrogated in p53-deficient myeloma cells. These results argue for an MV-based clinical trial for patients with p53 deficiency.


Subject(s)
Measles virus/physiology , Membrane Cofactor Protein/metabolism , Multiple Myeloma/pathology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/genetics , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
2.
J Clin Invest ; 126(12): 4640-4653, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27841764

ABSTRACT

Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Immunoconjugates/pharmacology , Membrane Cofactor Protein/antagonists & inhibitors , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Cell Line, Tumor , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/immunology , Gene Dosage/immunology , Humans , Immunoconjugates/immunology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mice , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
3.
J Thorac Oncol ; 9(8): 1101-10, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25157763

ABSTRACT

INTRODUCTION: Oncolytic virus therapy is a promising therapy for numerous tumor types. Edmonston-strain measles virus (MV) has been tested in clinical trials for ovarian cancer, glioma, and myeloma. Therefore, the antitumor activity of MV against non-small-cell lung cancer (NSCLC) was assessed. METHODS: Human NSCLC cells and immortalized lung epithelial cell lines, Beas2B, were infected with either MV-producing green fluorescent protein or MV-producing carcinoembryonic antigen. Cells were assessed for viability, induction of apoptosis by caspase and poly-ADP ribose polymerase cleavage, and for viral transgene production. The dependency of MV entry on CD46 and nectin-4 were determined using blocking antibodies. The role of host translational activity on viral replication was assessed by overexpression of eIF4E and translation inhibition. Antitumor activity was assessed by measuring treated NSCLC xenografts from flanks of nude mice. RESULTS: MV infection of NSCLC cells results in potent cell killing in most of the cell lines compared with immortalized Beas2B cells and induces apoptosis. MV infection was prevented by blocking of CD46, however independent of nectin-4 blockade. Tumor weights are diminished after intratumoral injections of MV-producing carcinoembryonic antigen in one of two cell lines and result in detectable viral transgene in serum of mice. CONCLUSIONS: These data indicate that MV is oncolytic for human NSCLC and this was independent of nectin-4 expression. Dysregulated protein translational machinery may play a role in determining tumor tropism in NSCLC. MV combined with gemcitabine could be explored further as chemovirotherapy for NSCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy , Tumor Burden , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoembryonic Antigen/drug effects , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Epithelial Cells/virology , Eukaryotic Initiation Factor-4E/metabolism , Green Fluorescent Proteins/genetics , Humans , Hydrazones/pharmacology , Lung , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Measles Vaccine , Measles virus/genetics , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/metabolism , Mice , Phosphoproteins/metabolism , Sirolimus/pharmacology , Thiazoles/pharmacology , Virus Replication , Gemcitabine
4.
Adv Exp Med Biol ; 735: 55-81, 2013.
Article in English | MEDLINE | ID: mdl-23402019

ABSTRACT

For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.


Subject(s)
Complement System Proteins/drug effects , Complement System Proteins/physiology , Lupus Erythematosus, Systemic/drug therapy , Membrane Proteins/drug effects , Membrane Proteins/physiology , Animals , Biomarkers , CD55 Antigens/physiology , CD59 Antigens/drug effects , CD59 Antigens/physiology , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/physiology , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/physiology
5.
J Immunol ; 188(11): 5303-10, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544928

ABSTRACT

The ubiquitous protein CD46, a regulator of complement activity, promotes T cell activation and differentiation toward a regulatory Tr1-like phenotype. The CD46-mediated differentiation pathway is defective in several chronic inflammatory diseases, underlying the importance of CD46 in controlling T cell function and the need to understand its regulatory mechanisms. Using an RNA interference-based screening approach in primary T cells, we have identified that two members of the G protein-coupled receptor kinases were involved in regulating CD46 expression at the surface of activated cells. We have investigated the role of PGE(2), which binds to the E-prostanoid family of G protein-coupled receptors through four subtypes of receptors called EP 1-4, in the regulation of CD46 expression and function. Conflicting roles of PGE(2) in T cell functions have been reported, and the reasons for these apparent discrepancies are not well understood. We show that addition of PGE(2) strongly downregulates CD46 expression in activated T cells. Moreover, PGE(2) differentially affects T cell activation, cytokine production, and phenotype depending on the activation signals received by the T cells. This was correlated with a distinct pattern of the PGE(2) receptors expressed, with EP4 being preferentially induced by CD46 activation. Indeed, addition of an EP4 antagonist could reverse the effects observed on cytokine production after CD46 costimulation. These data demonstrate a novel role of the PGE(2)-EP4 axis in CD46 functions, which might at least partly explain the diverse roles of PGE(2) in T cell functions.


Subject(s)
Dinoprostone/physiology , Lymphocyte Activation/immunology , Membrane Cofactor Protein/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Cell Proliferation , Cells, Cultured , Dinoprostone/metabolism , G-Protein-Coupled Receptor Kinase 1/physiology , Gene Expression Regulation/immunology , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/biosynthesis , RNA Interference/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
7.
Curr Cancer Drug Targets ; 10(8): 922-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20879979

ABSTRACT

The efficacy of cancer-immunotherapy with complement-activating monoclonal antibodies is limited by over-expression of one or more membrane-bound complement regulatory proteins (mCRPs: CD46, CD55, CD59) on the surface of neoplastic cells. In this study we designed small interfering RNAs (siRNAs) for posttranscriptional gene knock down of CD46, CD55 and CD59 aiming at to sensitize tumor cells to complement attack and thereby to better exploit complement for tumor cell destruction. Tumor cell lines of different origin, such as Du145 (prostate), BT474 (breast) and K562 (erythroleukemia) were selected for the study. FACS-analysis demonstrated that siRNA anti-CD46(301) reduced CD46 protein expression up to 80%, siRNA anti-CD55(255) diminished CD55 protein expression up to 49%, and CD59 protein expression was inhibited up to 82% by siRNA anti-CD59(1339). Time course experiments revealed a long-lasting silencing effect with >50% complement regulator inhibition up to day 13. Upon mCRP knock down, complement-dependent cytotoxicity (CDC) was augmented by 20-30% for CD46, by up to 24% for CD55 and by up to 55% for CD59. The combined inhibition of all three inhibitors further enhanced CDC by up to 66%. Dependent on the cell line, CD46 and CD55 downregulation increased significantly C3 ospsonization, which is known to support cell-mediated defense mechanisms. mCRP blocking antibodies were only partly able to further augment the tumor cells' susceptibility to complement lysis. Thus, siRNA-induced inhibition of complement regulator expression clearly sensitizes malignant cells to complement attack and, if specifically targeted to the tumor, appears suited as adjuvant to improve antibody-based cancer immunotherapy.


Subject(s)
CD55 Antigens/metabolism , CD59 Antigens/metabolism , Complement Activation/genetics , Membrane Cofactor Protein/metabolism , Neoplasms/genetics , Neoplasms/immunology , RNA, Small Interfering/genetics , CD55 Antigens/chemistry , CD55 Antigens/genetics , CD59 Antigens/chemistry , CD59 Antigens/genetics , Complement Activation/immunology , Flow Cytometry , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/genetics , Neoplasms/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Expert Opin Biol Ther ; 8(6): 759-68, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18476787

ABSTRACT

The effective and practical use of mAbs in cancer therapy became a reality with the development of the chimeric anti-CD20 mAb, rituximab. Several additional mAbs have since been approved for clinical use. Despite these successes, the mechanisms by which mAbs mediate antitumor activity are still unclear. Preclinical studies indicate complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) both can contribute to mAb-induced tumor cell lysis. However, evidence related to the relative clinical importance of each mechanism, and whether they are synergistic or antagonistic, is conflicting. New ways to enhance both CDC and ADCC are being developed in attempt to develop a more effective anticancer mAb. Continued research on the mechanisms of mAb therapy will be necessary if we are to take optimal advantage of the current mAbs and develop more effective mAbs in the future.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Complement Pathway, Classical , Cytotoxicity, Immunologic , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal, Murine-Derived , CD55 Antigens/drug effects , CD59 Antigens/drug effects , Drug Design , Humans , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/therapeutic use , Membrane Cofactor Protein/antagonists & inhibitors , Mice , Neoplasms/immunology , Rituximab , Xenograft Model Antitumor Assays
9.
Mol Cancer Res ; 5(8): 823-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17699108

ABSTRACT

CD46 is one of the complement-regulatory proteins expressed on the surface of normal and tumor cells for protection against complement-dependent cytotoxicity. Cancer cells need to access the blood circulation for continued growth and metastasis, thus exposing themselves to destruction by complement system components. Previous studies have established that the signal transducers and activators of transcription 3 (STAT3) transcription factor is persistently activated in a wide variety of human cancer cells and primary tumor tissues compared with their normal counterparts. Using microarray gene expression profiling, we identified the CD46 gene as a target for activated STAT3 signaling in human breast and prostate cancer cells. The CD46 promoter contains two binding sites for activated STAT3 and mutations introduced into the major site abolished STAT3 binding. Chromatin immunoprecipitation confirms binding of STAT3 to the CD46 promoter. CD46 promoter activity is induced by activation of STAT3 and blocked by a dominant-negative form of STAT3 in luciferase reporter assays. CD46 mRNA expression is induced by interleukin-6 and by transient transfection of normal human epithelial cells with a persistently active mutant construct of STAT3, STAT3C. Furthermore, we show that inhibition of STAT3-mediated CD46 cell surface expression sensitizes DU145 prostate cancer cells to cytotoxicity in an in vitro complement lysis assay using rabbit anti-DU145 antiserum and rabbit complement. These results show that activated STAT3 signaling induces the CD46 promoter and protects human cancer cells from complement-dependent cytotoxicity, suggesting a potential mechanism whereby oncogenic signaling contributes to tumor cell evasion of antibody-mediated immunity.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/metabolism , Complement System Proteins/pharmacology , Membrane Cofactor Protein/metabolism , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Dominant , Humans , Immunoglobulin G/immunology , Interleukin-6/metabolism , Luciferases/metabolism , Male , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/genetics , Microarray Analysis , Phosphorylation , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Rabbits , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transfection
10.
J Immunol ; 178(9): 5991-8, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17442984

ABSTRACT

Malignant cells are often resistant to complement activation through the enhanced expression of complement inhibitors. In this work, we examined the protective role of factor H, CD46, CD55, and CD59 in two non-small cell lung cancer cell lines, H1264 and A549, upon activation of the classical pathway of complement. Complement was activated with polyclonal Abs raised against each cell line. After blocking factor H activity with a neutralizing Ab, C3 deposition and C5a release were more efficient. Besides, a combined inhibition of factor H and CD59 significantly increased complement-mediated lysis. CD46 and CD55 did not show any effect in the control of complement activation. Factor H expression was knockdown on A549 cells using small interfering RNA. In vivo growth of factor H-deficient cells in athymic mice was significantly reduced. C3 immunocytochemistry on explanted xenografts showed an enhanced activation of complement in these cells. Besides, when mice were depleted of complement with cobra venom factor, growth was recovered, providing further evidence that complement was important in the reduction of in vivo growth. In conclusion, we show that expression of the complement inhibitor factor H by lung cancer cells can prevent complement activation and improve tumor development in vivo. This may have important consequences in the efficiency of complement-mediated immunotherapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Complement Activation , Complement Factor H/antagonists & inhibitors , Complement Factor H/immunology , Lung Neoplasms/immunology , Animals , CD55 Antigens/drug effects , CD55 Antigens/immunology , CD59 Antigens/drug effects , CD59 Antigens/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Complement Activation/genetics , Complement C3/analysis , Complement C3/immunology , Complement C5a/immunology , Complement Factor H/genetics , Cytotoxicity, Immunologic , Down-Regulation , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/immunology , Mice , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...