Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 1399, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446805

ABSTRACT

SHP2 is a ubiquitous tyrosine phosphatase involved in regulating both tumor and immune cell signaling. In this study, we discovered a novel immune modulatory function of SHP2. Targeting this protein with allosteric SHP2 inhibitors promoted anti-tumor immunity, including enhancing T cell cytotoxic function and immune-mediated tumor regression. Knockout of SHP2 using CRISPR/Cas9 gene editing showed that targeting SHP2 in cancer cells contributes to this immune response. Inhibition of SHP2 activity augmented tumor intrinsic IFNγ signaling resulting in enhanced chemoattractant cytokine release and cytotoxic T cell recruitment, as well as increased expression of MHC Class I and PD-L1 on the cancer cell surface. Furthermore, SHP2 inhibition diminished the differentiation and inhibitory function of immune suppressive myeloid cells in the tumor microenvironment. SHP2 inhibition enhanced responses to anti-PD-1 blockade in syngeneic mouse models. Overall, our study reveals novel functions of SHP2 in tumor immunity and proposes that targeting SHP2 is a promising strategy for cancer immunotherapy.


Subject(s)
Immunity, Cellular , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction/genetics
2.
Mol Cancer Ther ; 19(10): 2089-2104, 2020 10.
Article in English | MEDLINE | ID: mdl-32847974

ABSTRACT

The sole inhibitory Fcγ receptor CD32b (FcγRIIb) is expressed throughout B and plasma cell development and on their malignant counterparts. CD32b expression on malignant B cells is known to provide a mechanism of resistance to rituximab that can be ameliorated with a CD32b-blocking antibody. CD32b, therefore, represents an attractive tumor antigen for targeting with a monoclonal antibody (mAb). To this end, two anti-CD32b mAbs, NVS32b1 and NVS32b2, were developed. Their complementarity-determining regions (CDR) bind the CD32b Fc binding domain with high specificity and affinity while the Fc region is afucosylated to enhance activation of FcγRIIIa on immune effector cells. The NVS32b mAbs selectively target CD32b+ malignant cells and healthy B cells but not myeloid cells. They mediate potent killing of opsonized CD32b+ cells via antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP) as well as complement-dependent cytotoxicity (CDC). In addition, NVS32b CDRs block the CD32b Fc-binding domain, thereby minimizing CD32b-mediated resistance to therapeutic mAbs including rituximab, obinutuzumab, and daratumumab. NVS32b mAbs demonstrate robust antitumor activity against CD32b+ xenografts in vivo and immunomodulatory activity including recruitment of macrophages to the tumor and enhancement of dendritic cell maturation in response to immune complexes. Finally, the activity of NVS32b mAbs on CD32b+ primary malignant B and plasma cells was confirmed using samples from patients with B-cell chronic lymphocytic leukemia (CLL) and multiple myeloma. The findings indicate the promising potential of NVS32b mAbs as a single agent or in combination with other mAb therapeutics for patients with CD32b+ malignant cells.


Subject(s)
Lymphoma, B-Cell/genetics , Neoplasms, Plasma Cell/genetics , Receptors, IgG/immunology , Animals , CHO Cells , Cricetulus , Humans
3.
J Med Chem ; 59(10): 4711-23, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27187609

ABSTRACT

MELK kinase has been implicated in playing an important role in tumorigenesis. Our previous studies suggested that MELK is involved in the regulation of cell cycle and its genetic depletion leads to growth inhibition in a subset of high MELK-expressing basal-like breast cancer cell lines. Herein we describe the discovery and optimization of novel MELK inhibitors 8a and 8b that recapitulate the cellular effects observed by short hairpin ribonucleic acid (shRNA)-mediated MELK knockdown in cellular models. We also discovered a novel fluorine-induced hydrophobic collapse that locked the ligand in its bioactive conformation and led to a 20-fold gain in potency. These novel pharmacological inhibitors achieved high exposure in vivo and were well tolerated, which may allow further in vivo evaluation.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/standards , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
4.
Cancer Res ; 75(22): 4937-48, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26490646

ABSTRACT

Patients with lung tumors harboring activating mutations in the EGF receptor (EGFR) show good initial treatment responses to the EGFR tyrosine kinase inhibitors (TKI) erlotinib or gefitinib. However, acquired resistance invariably develops. Applying a focused shRNA screening approach to identify genes whose knockdown can prevent and/or overcome acquired resistance to erlotinib in several EGFR-mutant non-small cell lung cancer (NSCLC) cell lines, we identified casein kinase 1 α (CSNK1A1, CK1α). We found that CK1α suppression inhibits the NF-κB prosurvival signaling pathway. Furthermore, downregulation of NF-κB signaling by approaches independent of CK1α knockdown can also attenuate acquired erlotinib resistance, supporting a role for activated NF-κB signaling in conferring acquired drug resistance. Importantly, CK1α suppression prevented erlotinib resistance in an HCC827 xenograft model in vivo. Our findings suggest that patients with EGFR-mutant NSCLC might benefit from a combination of EGFR TKIs and CK1α inhibition to prevent acquired drug resistance and to prolong disease-free survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Casein Kinase I/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Erlotinib Hydrochloride/pharmacology , Female , Gene Knockdown Techniques , Genes, erbB-1/genetics , Humans , Immunoblotting , Lung Neoplasms/enzymology , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 111(8): 3128-33, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24520176

ABSTRACT

Defects in epigenetic regulation play a fundamental role in the development of cancer, and epigenetic regulators have recently emerged as promising therapeutic candidates. We therefore set out to systematically interrogate epigenetic cancer dependencies by screening an epigenome-focused deep-coverage design shRNA (DECODER) library across 58 cancer cell lines. This screen identified BRM/SMARCA2, a DNA-dependent ATPase of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex, as being essential for the growth of tumor cells that harbor loss of function mutations in BRG1/SMARCA4. Depletion of BRM in BRG1-deficient cancer cells leads to a cell cycle arrest, induction of senescence, and increased levels of global H3K9me3. We further demonstrate the selective dependency of BRG1-mutant tumors on BRM in vivo. Genetic alterations of the mSWI/SNF chromatin remodeling complexes are the most frequent among chromatin regulators in cancers, with BRG1/SMARCA4 mutations occurring in ∼10-15% of lung adenocarcinomas. Our findings position BRM as an attractive therapeutic target for BRG1 mutated cancers. Because BRG1 and BRM function as mutually exclusive catalytic subunits of the mSWI/SNF complex, we propose that such synthetic lethality may be explained by paralog insufficiency, in which loss of one family member unveils critical dependence on paralogous subunits. This concept of "cancer-selective paralog dependency" may provide a more general strategy for targeting other tumor suppressor lesions/complexes with paralogous subunits.


Subject(s)
DNA Helicases/deficiency , Epigenesis, Genetic/physiology , Multiprotein Complexes/genetics , Neoplasms/genetics , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Transcription Factors/genetics , Blotting, Western , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cellular Senescence/genetics , Gene Knockdown Techniques , Gene Library , Histones/metabolism , Humans , Immunoprecipitation , Multiprotein Complexes/metabolism , RNA, Small Interfering/genetics , Transcription Factors/metabolism
6.
Cancer Res ; 73(19): 6024-35, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23928993

ABSTRACT

HER2/HER3 dimerization resulting from overexpression of HER2 or neuregulin (NRG1) in cancer leads to HER3-mediated oncogenic activation of phosphoinositide 3-kinase (PI3K) signaling. Although ligand-blocking HER3 antibodies inhibit NRG1-driven tumor growth, they are ineffective against HER2-driven tumor growth because HER2 activates HER3 in a ligand-independent manner. In this study, we describe a novel HER3 monoclonal antibody (LJM716) that can neutralize multiple modes of HER3 activation, making it a superior candidate for clinical translation as a therapeutic candidate. LJM716 was a potent inhibitor of HER3/AKT phosphorylation and proliferation in HER2-amplified and NRG1-expressing cancer cells, and it displayed single-agent efficacy in tumor xenograft models. Combining LJM716 with agents that target HER2 or EGFR produced synergistic antitumor activity in vitro and in vivo. In particular, combining LJM716 with trastuzumab produced a more potent inhibition of signaling and cell proliferation than trastuzumab/pertuzumab combinations with similar activity in vivo. To elucidate its mechanism of action, we solved the structure of LJM716 bound to HER3, finding that LJM716 bound to an epitope, within domains 2 and 4, that traps HER3 in an inactive conformation. Taken together, our findings establish that LJM716 possesses a novel mechanism of action that, in combination with HER2- or EGFR-targeted agents, may leverage their clinical efficacy in ErbB-driven cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/pathology , Neuregulin-1/metabolism , Protein Conformation/drug effects , Receptor, ErbB-3/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphorylation/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Nat Med ; 16(12): 1429-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21076395

ABSTRACT

Aberrant activation of the Hedgehog (Hh) pathway can drive tumorigenesis. To investigate the mechanism by which glioma-associated oncogene family zinc finger-1 (GLI1), a crucial effector of Hh signaling, regulates Hh pathway activation, we searched for GLI1-interacting proteins. We report that the chromatin remodeling protein SNF5 (encoded by SMARCB1, hereafter called SNF5), which is inactivated in human malignant rhabdoid tumors (MRTs), interacts with GLI1. We show that Snf5 localizes to Gli1-regulated promoters and that loss of Snf5 leads to activation of the Hh-Gli pathway. Conversely, re-expression of SNF5 in MRT cells represses GLI1. Consistent with this, we show the presence of a Hh-Gli-activated gene expression profile in primary MRTs and show that GLI1 drives the growth of SNF5-deficient MRT cells in vitro and in vivo. Therefore, our studies reveal that SNF5 is a key mediator of Hh signaling and that aberrant activation of GLI1 is a previously undescribed targetable mechanism contributing to the growth of MRT cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Rhabdoid Tumor/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , Immunoblotting , In Situ Hybridization , Mass Spectrometry , Mice , Microarray Analysis , SMARCB1 Protein , Transcription Factors/genetics , Zinc Finger Protein GLI1
8.
Cancer Biol Ther ; 7(12): 1959-67, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18981727

ABSTRACT

The MUC1 oncoprotein interacts with the c-Abl tyrosine kinase and blocks nuclear targeting of c-Abl in the apoptotic response to DNA damage. Mutation of the MUC1 cytoplasmic domain at Tyr-60 disrupts the MUC1-c-Abl interaction. The present results demonstrate that the MUC1(Y60F) mutant is a potent inducer of the ARF tumor suppressor. MUC1(Y60F) induces transcription of the ARF locus by a c-Abl-dependent mechanism that promotes CUL-4A-mediated nuclear export of the replication protein Cdc6. The functional significance of these findings is that MUC1(Y60F)-induced ARF expression and thereby inhibition of MDM2 results in the upregulation of p53 and the homeodomain interacting protein kinase 2 (HIPK2) serine/threonine kinase. HIPK2-mediated phosphorylation of p53 on Ser-46 was further associated with a shift from expression of the cell cycle arrest-related p21 gene to the apoptosis-related PUMA gene. We also show that the MUC1(Y60F) mutant functions as dominant negative inhibitor of tumorigenicity. These findings indicate that the oncogenic function of MUC1 is conferred by suppressing activation of the ARF-MDM2-p53 pathway.


Subject(s)
ADP-Ribosylation Factor 1/genetics , Mucin-1/genetics , Mucin-1/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Cell Cycle/genetics , Cell Line, Tumor , Colorectal Neoplasms , HCT116 Cells , Hematologic Neoplasms/genetics , Humans , Proto-Oncogene Proteins c-abl/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
9.
J Biol Chem ; 282(27): 19321-30, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17500061

ABSTRACT

The MUC1 heterodimeric transmembrane protein is aberrantly overexpressed by most human carcinomas. The MUC1 C-terminal subunit (MUC1-C) is devoid of a classical nuclear localization signal and is targeted to the nucleus by an unknown mechanism. The present results demonstrate that MUC1-C associates with importin beta and not importin alpha. The results also show that, like importin beta, MUC1-C binds to Nup62 (nucleoporin p62). MUC1-C binds directly to the Nup62 central domain and indirectly to the Nup62 C-terminal alpha-helical coiled-coil domain. We demonstrate that MUC1-C forms oligomers and that oligomerization is necessary for binding to Nup62. The MUC1-C cytoplasmic domain contains a CQC motif that when mutated to AQA abrogates oligomerization and binding to Nup62. Stable expression of MUC1 with the CQC --> AQA mutations was associated with targeting to the cell membrane and cytosol and attenuation of nuclear localization. The results further show that expression of MUC1(CQC-AQA) attenuates MUC1-induced (i) transcriptional coactivation, (ii) anchorage-independent growth, and (iii) tumorigenicity. These findings indicate that the MUC1-C oncoprotein is imported to the nucleus by a pathway involving Nup62.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Nucleus/metabolism , Membrane Glycoproteins/metabolism , Mucins/metabolism , Neoplasms/metabolism , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Amino Acid Motifs , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Gene Expression , Humans , Mucin-1 , Mucins/genetics , Mutation , Neoplasms/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins/genetics , Protein Binding/genetics , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Transcriptional Activation/genetics , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism
10.
Cancer Res ; 65(22): 10413-22, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288032

ABSTRACT

Dysregulation of beta-catenin is of importance to the development of diverse human malignancies. The MUC1 oncoprotein is aberrantly overexpressed by most human carcinomas and associates with beta-catenin. However, the functional significance of the MUC1-beta-catenin interaction is not known. Here, we show that MUC1 increases beta-catenin levels in the cytoplasm and nucleus of carcinoma cells. Previous studies have shown that glycogen synthase kinase 3beta (GSK3beta) phosphorylates beta-catenin and thereby targets it for proteosomal degradation. Consistent with the up-regulation of beta-catenin levels, our results show that MUC1 blocks GSK3beta-mediated phosphorylation and degradation of beta-catenin. To further define the interaction between MUC1 and beta-catenin, we identified a serine-rich motif (SRM) in the MUC1 cytoplasmic domain that binds directly to beta-catenin Armadillo repeats. Mutation of the SRM attenuated binding of MUC1 to beta-catenin and MUC1-mediated inhibition of beta-catenin degradation. Importantly, disruption of the MUC1-beta-catenin interaction with the SRM mutant also attenuated MUC1-induced anchorage-dependent and -independent growth and delayed MUC1-mediated tumorigenicity. These findings indicate that MUC1 promotes transformation, at least in part, by blocking GSK3beta-mediated phosphorylation and thereby degradation of beta-catenin.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Mucin-1/metabolism , beta Catenin/metabolism , Amino Acid Motifs , Cytoplasm/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Mucin-1/genetics , Phosphorylation , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Transfection , Up-Regulation
11.
Leuk Res ; 28(12): 1303-12, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15475072

ABSTRACT

Presentation of AML antigens by dendritic cells (DC) could potentially induce a T cell-mediated anti-leukemic immune response. In the present study, we generated DC from adherent (AD-DC) and non-adherent (NAD-DC) myeloblasts obtained from bone marrows of AML patients. Both cell populations displayed morphological, phenotypic and functional properties of DC. The functions of NAD-DC were compared to AD-DC that had been fused with autologous AML blasts (FC/AML). The FC/AML induced greater T cell proliferation and CTL activity against autologous AML blasts (9/10 cases) as compared to NAD-DC. FC/AML may thus represent a promising strategy for DC-based immunotherapy of patients with AML.


Subject(s)
Dendritic Cells/pathology , Granulocyte Precursor Cells/pathology , Leukemia/immunology , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Bone Marrow Cells , Cell Adhesion , Cell Culture Techniques , Cell Fusion , Cytotoxicity, Immunologic , Female , Humans , Immunotherapy/methods , Leukemia/pathology , Male , Middle Aged
12.
Cancer Cell ; 5(2): 163-75, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14998492

ABSTRACT

The MUC1 transforming protein is overexpressed by most human carcinomas. The present studies demonstrate that the MUC1 C-terminal subunit (MUC1 C-ter) localizes to mitochondria in HCT116/MUC1 colon carcinoma cells and that heregulin stimulates mitochondrial targeting of MUC1 C-ter. We also show that MUC1 attenuates cisplatin-induced (1) release of mitochondrial apoptogenic factors, (2) activation of caspase-3, and (3) induction of apoptosis. Moreover, knockdown of MUC1 expression in A549 lung and ZR-75-1 breast carcinoma cells by MUC1siRNA was associated with increased sensitivity to genotoxic drugs in vitro and in vivo. These findings indicate that MUC1 attenuates the apoptotic response to DNA damage and that this oncoprotein confers resistance to genotoxic anticancer agents.


Subject(s)
Antigens/metabolism , Apoptosis/physiology , Drug Resistance, Neoplasm/physiology , Glycoproteins/metabolism , Mitochondria/metabolism , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins , Caspase 3 , Caspases/metabolism , Cisplatin/pharmacology , Cloning, Molecular , Cytochromes c/metabolism , DNA Damage/physiology , Flow Cytometry , Membrane Glycoproteins/metabolism , Mucin-1 , Mucins , Neuregulin-1/pharmacology , Protein Subunits/metabolism , RNA, Small Interfering/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
13.
Cancer Biol Ther ; 2(6): 702-6, 2003.
Article in English | MEDLINE | ID: mdl-14688481

ABSTRACT

The DF3/MUC1 mucin-like transmembrane oncoprotein is overexpressed by most human carcinomas. The MUC1 cytoplasmic domain (CD) binds directly to the Wnt effector, beta-catenin, and colocalizes with beta-catenin in the nucleus; however, the nuclear function of MUC1 is unknown. The present results demonstrate that MUC1 coactivates transcription of beta-catenin-Tcf-binding sites in the pTOPFLASH reporter. Activation of transcription was abrogated by expression of MUC1 with a Y-46->F mutation in the CD that attenuates binding of MUC1 and beta-catenin. We also show that transcription of the Wnt responsive cyclin D1 promoter is activated by MUC1, but not MUC1(Y46F), and that the cyclin D1 gene is upregulated in MUC1-positive cells. In concert with these results, MUC1-induced anchorage-independent growth and tumorigenicity were also abrogated by mutating MUC1 at the Y-46 site. These findings support a model in which the MUC1 functions as a transforming protein by coactivating transcription of Wnt target genes.


Subject(s)
Mucin-1/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Transcriptional Activation , Amino Acid Sequence , Animals , Binding Sites , Cell Adhesion/genetics , Cell Nucleus/metabolism , Cell Transplantation , Clone Cells , Cyclin D1/genetics , Cytoplasm/chemistry , Cytoskeletal Proteins , Gene Expression Regulation, Neoplastic , Genes, Reporter , HCT116 Cells , Humans , Luciferases/metabolism , Mice , Mice, Nude , Mucin-1/chemistry , Mucin-1/genetics , Mutation , Promoter Regions, Genetic , Protein Binding , Trans-Activators , Transcription Factors/metabolism , Up-Regulation , Wnt Proteins , beta Catenin
14.
Oncogene ; 22(38): 6107-10, 2003 Sep 04.
Article in English | MEDLINE | ID: mdl-12955090

ABSTRACT

The human DF3/MUC1 mucin-like glycoprotein is aberrantly overexpressed by most carcinomas of the breast and other epithelia. The contribution of MUC1 overexpression to the malignant phenotype is, however, not known. In the present studies, we have stably expressed MUC1 in rat 3Y1 fibroblasts. MUC1-positive cells were selected from independent transfections. The results demonstrate that, as found in human carcinomas, MUC1 is expressed on the cell surface and as a complex with beta-catenin in the nucleus of the transfectants. Colony formation in soft agar demonstrates that cells expressing MUC1, but not the empty vector, exhibit anchorage-independent growth. The results also show that MUC1 expression confers tumor formation in nude mice. These findings provide the first evidence that MUC1 induces cellular transformation.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Oncogenes , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenicity Tests , Cell Membrane , Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Mice , Mice, Nude , Mucin-1/genetics , Mucin-1/metabolism , Rats , Trans-Activators/metabolism , Tumor Stem Cell Assay , beta Catenin
15.
Immunology ; 109(2): 300-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12757626

ABSTRACT

The tumour-associated antigen mucin 1 (MUC1) is a multifunctional protein involved in protection of mucous membranes, signal transduction, and modulation of the immune system. More than 70% of cancers overexpress MUC1, making MUC1 a potential target for immunotherapy. In the present study, MUC1 transgenic mice were crossed with syngeneic strains that express the polyomavirus middle-T oncogene (PyMT) driven by the mouse mammary tumour virus promoter long-terminal repeat (MMTV-LTR). The resultant breed (MMT mice) developed spontaneous MUC1-expressing mammary carcinomas with 100% penetrance at 8-15 weeks of age. As found in human breast cancer, the mammary carcinoma in MMT mice arose in multiple stages. Immunization with fusions of dendritic cells and MUC1-positive tumour cells (FC/MUC1) induced MUC1-specific immune responses that blocked or delayed the development of spontaneous breast carcinomas. In contrast, there was no delay of tumour development in MMT mice immunized with irradiated MC38/MUC1 tumour cells. The efficacy of fusion cells was closely correlated with the timing of initial immunization. Immunization with FC/MUC1 initiated in MMT mice at < 1, 1-2 and 2-3 months of age rendered 33, 5 and 0% of mice free of tumour, respectively, up to 6 months. Whereas mice immunized in the later stage of tumour development succumbed to their disease, immunization resulted in control of tumour progression and prolongation of life. These results indicate that immunization with FC/MUC1 can generate an anti-MUC1 response that is sufficient to delay the development of spontaneous mammary carcinomas and control tumour progression in MMT mice.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Mammary Neoplasms, Experimental/prevention & control , Mucin-1/immunology , Peptide Fragments/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Cell Fusion , Cell Transplantation , Disease Progression , Female , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucin-1/metabolism , Neoplasm Transplantation , Peptide Fragments/metabolism
16.
Blood ; 99(7): 2512-7, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11895787

ABSTRACT

Fusions of cancer cells and dendritic cells (DCs) are effective in the treatment of animal tumor models and patients with metastatic renal carcinoma. In this study, we have fused DCs with mouse 4TOO plasmacytoma cells. The results demonstrate that vaccination of mice with the fusion cells (FC/4TOO) is associated with induction of antitumor humoral and cytotoxic T lymphocyte (CTL) responses. Immunization with FC/4TOO cells protected mice against tumor challenge. In addition, treatment of established multiple myeloma with FC/4TOO cells was associated with prolongation of survival but not with eradication of disease. As interleukin (IL)-12 potentiates the induction of immune responses, recombinant mouse IL-12 was administered with the FC/4TOO vaccine. Treatment of mice with FC/4TOO and IL-12 was associated with increased CTL activity and T-cell proliferation responses. Treatment with FC/4TOO and IL-12 also resulted in eradication of established disease. These findings demonstrate that immunization with FC/4TOO fusion cells and IL-12 potentiates antitumor immunity and the treatment of murine multiple myeloma.


Subject(s)
Dendritic Cells/transplantation , Immunization, Passive , Interleukin-12/pharmacology , Multiple Myeloma/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation , Cell Fusion , Cytotoxicity, Immunologic , Humans , Immunization Schedule , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Multiple Myeloma/prevention & control , Plasmacytoma/immunology , Tumor Cells, Cultured
17.
J Immunol ; 168(5): 2111-7, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11859096

ABSTRACT

Previous work has demonstrated that dendritic/tumor fusion cells induce potent antitumor immune responses in vivo and in vitro. However, little is known about the migration and homing of fusion cells after s.c. injection or the kinetics of CD4+ and CD8+ T cell activation. In the present study, fluorescence-labeled dendritic/MUC1-positive tumor fusion cells (FC/MUC1) were injected s.c. into MUC1-transgenic mice. The FC/MUC1 migrated to draining lymph nodes and were closely associated with T cells in a pattern comparable with that of unfused dendritic cells. Immunization of MUC1-transgenic mice with FC/MUC1 resulted in proliferation of T cells and induced MUC1-specific CD8+ CTL. Moreover, CD4+ T cells activated by FC/MUC1 were multifunctional effectors that produced IL-2, IFN-gamma, IL-4, and IL-10. These findings indicate that both CD4+ and CD8+ T cells can be primed in vivo by FC/MUC1 immunization.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Mucin-1/genetics , Mucin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Cell Fusion , Cell Movement , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Cytotoxicity Tests, Immunologic , Dendritic Cells/transplantation , Humans , Kinetics , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...