Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 6: 31740, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27545199

ABSTRACT

MUC1 is a shared tumor antigen expressed on >80% of human cancers. We completed the first prophylactic cancer vaccine clinical trial based on a non-viral antigen, MUC1, in healthy individuals at-risk for colon cancer. This trial provided a unique source of potentially effective and safe immunotherapeutic drugs, fully-human antibodies affinity-matured in a healthy host to a tumor antigen. We purified, cloned, and characterized 13 IgGs specific for several tumor-associated MUC1 epitopes with a wide range of binding affinities. These antibodies bind hypoglycosylated MUC1 on human cancer cell lines and tumor tissues but show no reactivity against fully-glycosylated MUC1 on normal cells and tissues. We found that several antibodies activate complement-mediated cytotoxicity and that T cells carrying chimeric antigen receptors with the antibody variable regions kill MUC1(+) target cells, express activation markers, and produce interferon gamma. Fully-human and tumor-specific, these antibodies are candidates for further testing and development as immunotherapeutic drugs.


Subject(s)
Antibodies/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Mucin-1/immunology , Adult , Amino Acid Sequence , Antibody Affinity/immunology , Base Sequence , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Epitopes/immunology , Female , HEK293 Cells , Humans , Immunoglobulin G/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , MCF-7 Cells , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
2.
PLoS One ; 8(4): e61125, 2013.
Article in English | MEDLINE | ID: mdl-23577196

ABSTRACT

Transforming growth factor (ß1TGFß1) can promote proliferation in late stage cancers but acts as a tumor suppressor in normal epithelial cells and in early stage cancers. Although, the TGFß pathway has been shown to play a key role in tumorigenesis and metastasis, only a limited number of models have been developed to understand this process. Here, we present a novel model system to discern this paradoxical role of TGFß1 using the MDA-MB-231 (MB-231) cell line. The MB-231 triple-negative breast cancer cell line has been extensively characterized and has been shown to continue to proliferate and undergo epithelial-to-mesenchymal transition (EMT) upon TGFß1 stimulation. We have previously shown by microarray analysis that expression of GATA3 in MB-231 cells results in reprogramming of these cells from a basal to a luminal subtype associated with a reduction of metastasis and tumorigenesis when implanted as xenografts. We now demonstrate that GATA3 overexpression in these cells results in a reduction of TGFß1 response, reversal of EMT, and most importantly, restoration of sensitivity to the inhibitory effects on proliferation of TGFß1. Microarray analysis revealed that TGFß1 treatment resulted in reduction of several cell cycle effectors in 231-GATA3 cells but not in control cells. Furthermore, our microarray analysis revealed a significant increase of BMP5 in 231-GATA3 cells. We demonstrate that combined treatment of MB-231 control cells with TGFß1 and BMP5 results in a significant reduction of cellular proliferation. Thus, this model offers a means to further investigate potentially novel mechanisms involved in the switch in response to TGFß1 from tumor promoter to tumor suppressor through the reprogramming of a triple-negative breast cancer cell line by the GATA3 transcription factor.


Subject(s)
Breast Neoplasms/pathology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Transforming Growth Factor beta/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 5/metabolism , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism
3.
Breast Cancer Res ; 14(4): R109, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22812567

ABSTRACT

INTRODUCTION: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is diagnosed in approximately 15% of all human breast cancer (BrCa) patients. Currently, no targeted therapies exist for this subtype of BrCa and prognosis remains poor. Our laboratory has previously identified a proliferation/DNA repair/cell cycle gene signature (Tag signature) that is characteristic of human TNBC. We hypothesize that targeting the dysregulated biological networks in the Tag gene signature will lead to the identification of improved combination therapies for TNBC. METHODS: Cross-species genomic analysis was used to identify human breast cancer cell lines that express the Tag signature. Knock-down of the up-regulated genes in the Tag signature by siRNA identified several genes that are critical for TNBC cell growth. Small molecule inhibitors to two of these genes were analyzed, alone and in combination, for their effects on cell proliferation, cell cycle, and apoptosis in vitro and tumor growth in vivo. Synergy between the two drugs was analyzed by the Chou-Talalay method. RESULTS: A custom siRNA screen was used to identify targets within the Tag signature that are critical for growth of TNBC cells. Ribonucleotide reductase 1 and 2 (RRM1 and 2) and checkpoint kinase 1 (CHK1) were found to be critical targets for TNBC cell survival. Combination therapy, to simultaneously attenuate cell cycle checkpoint control through inhibition of CHK1 while inducing DNA damage with gemcitabine, improved therapeutic efficacy in vitro and in xenograft models of TNBC. CONCLUSIONS: This combination therapy may have translational value for patients with TNBC and improve therapeutic response for this aggressive form of breast cancer.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Ribonucleotide Reductases/antagonists & inhibitors , Triple Negative Breast Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Checkpoint Kinase 1 , Cluster Analysis , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Mice , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Retinoblastoma Protein/metabolism , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Gemcitabine
5.
Nat Rev Cancer ; 8(4): 253-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18354415

ABSTRACT

The cyclin-dependent kinase (Cdk) inhibitor p27 (also known as KIP1) regulates cell proliferation, cell motility and apoptosis. Interestingly, the protein can exert both positive and negative functions on these processes. Diverse post-translational modifications determine the physiological role of p27. Phosphorylation regulates p27 binding to and inhibition of cyclin-Cdk complexes, its localization and its ubiquitin-mediated proteolysis. In cancers, p27 is inactivated through impaired synthesis, accelerated degradation and by mislocalization. Moreover, studies in several tumour types indicate that p27 expression levels have both prognostic and therapeutic implications.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Humans , Neoplasms/drug therapy , Phosphorylation , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...