Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 48(8): 4013-4027, 2020 05 07.
Article in English | MEDLINE | ID: mdl-31989173

ABSTRACT

Libraries of single-stranded oligodeoxynucleotides (ssODNs) can be enriched for sequences that specifically bind molecules on naïve complex biological samples like cells or tissues. Depending on the enrichment strategy, the ssODNs can identify molecules specifically associated with a defined biological condition, for example a pathological phenotype, and thus are potentially useful for biomarker discovery. We performed ADAPT, a variant of SELEX, on exosomes secreted by VCaP prostate cancer cells. A library of ∼1011 ssODNs was enriched for those that bind to VCaP exosomes and discriminate them from exosomes derived from LNCaP prostate cancer cells. Next-generation sequencing (NGS) identified the best discriminating ssODNs, nine of which were resynthesized and their discriminatory ability confirmed by qPCR. Affinity purification with one of the sequences (Sequence 7) combined with LC-MS/MS identified its molecular target complex, whereof most proteins are part of or associated with the multiprotein ESCRT complex participating in exosome biogenesis. Within this complex, YBX1 was identified as the directly-bound target protein. ADAPT thus is able to differentiate exosomes from cancer cell subtypes from the same lineage. The composition of ESCRT complexes in exosomes from VCaP versus LNCaP cells might constitute a discriminatory element between these prostate cancer subtypes.


Subject(s)
Endosomal Sorting Complexes Required for Transport/chemistry , Exosomes/metabolism , Prostatic Neoplasms/chemistry , Aptamers, Nucleotide , Cell Line, Tumor , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Male , Prostatic Neoplasms/classification , Prostatic Neoplasms/metabolism , SELEX Aptamer Technique , Y-Box-Binding Protein 1/metabolism
2.
Cell Chem Biol ; 26(5): 756-764.e6, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30930163

ABSTRACT

Spliceosomal dysregulation dramatically affects many cellular processes, notably signal transduction, metabolism, and proliferation, and has led to the concept of targeting intracellular spliceosomal proteins to combat cancer. Here we show that a subset of lymphoma cells displays a spliceosomal complex on their surface, which we term surface spliceosomal complex (SSC). The SSC consists of at least 13 core components and was discovered as the binding target of the non-Hodgkin's lymphoma-specific aptamer C10.36. The aptamer triggers SSC internalization, causing global changes in alternative splicing patterns that eventually lead to necrotic cell death. Our study reveals an exceptional spatial arrangement of a spliceosomal complex and defines it not only as a potential target of anti-cancer drugs, but also suggests that its localization plays a fundamental role in cell survival.


Subject(s)
Alternative Splicing , Spliceosomes/metabolism , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Heterogeneous-Nuclear Ribonucleoprotein U/chemistry , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Lymphoma/metabolism , Lymphoma/pathology , Tandem Mass Spectrometry
3.
Sci Rep ; 7: 42741, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28218293

ABSTRACT

Technologies capable of characterizing the full breadth of cellular systems need to be able to measure millions of proteins, isoforms, and complexes simultaneously. We describe an approach that fulfils this criterion: Adaptive Dynamic Artificial Poly-ligand Targeting (ADAPT). ADAPT employs an enriched library of single-stranded oligodeoxynucleotides (ssODNs) to profile complex biological samples, thus achieving an unprecedented coverage of system-wide, native biomolecules. We used ADAPT as a highly specific profiling tool that distinguishes women with or without breast cancer based on circulating exosomes in their blood. To develop ADAPT, we enriched a library of ~1011 ssODNs for those associating with exosomes from breast cancer patients or controls. The resulting 106 enriched ssODNs were then profiled against plasma from independent groups of healthy and breast cancer-positive women. ssODN-mediated affinity purification and mass spectrometry identified low-abundance exosome-associated proteins and protein complexes, some with known significance in both normal homeostasis and disease. Sequencing of the recovered ssODNs provided quantitative measures that were used to build highly accurate multi-analyte signatures for patient classification. Probing plasma from 500 subjects with a smaller subset of 2000 resynthesized ssODNs stratified healthy, breast biopsy-negative, and -positive women. An AUC of 0.73 was obtained when comparing healthy donors with biopsy-positive patients.


Subject(s)
Breast Neoplasms/blood , Exosomes/genetics , Oligodeoxyribonucleotides/metabolism , Systems Biology/methods , Area Under Curve , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , SELEX Aptamer Technique , Sequence Analysis, DNA
4.
Cancer Immunol Immunother ; 61(11): 2055-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22543528

ABSTRACT

Monoclonal antibodies (mAbs) against tumor-associated antigens are useful anticancer agents. Antibody-dependent cellular cytotoxicity (ADCC) is one of the major mechanisms responsible for initiating natural killer cell (NK)-mediated killing of tumors. However, the regulation of ADCC via NK cells is poorly understood. We have investigated the cytolytic activity of NK cells against pancreatic cancer cells that were coated with an antibody directed against the human tumor antigen, Mucin-1 designated HMFG-2, either alone or conjugated to CpG oligodeoxynucleotide (CpG ODN). Conjugated antibodies were tested for their ability to elicit ADCC in vitro and in vivo against pancreatic cancer cells. NK cells cultured in the presence of immobilized CpG ODN, HMFG-2 Ab, or CpG ODN-conjugated HMFG-2 Ab were able to up-regulate perforin similarly. Interestingly, a significant higher ADCC was observed when CpG ODN-conjugated HMFG-2-coated tumor cells were co-cultured with NK cells compared to unconjugated HMFG-2 Ab or CpG ODN alone. Moreover, MyD88-deficient NK cells can perform ADCC in vitro. Furthermore, intratumoral injections of CpG ODN-conjugated HMFG-2 induced a significant reduction in tumor burden in vivo in an established model of pancreatic tumor in nude mice compared to CpG ODN or the HMFG-2 alone. Depletion of macrophages or NK cells before treatment confirmed that both cells were required for the anti-tumor response in vivo. Results also suggest that CpG ODN and HMFG-2 Ab could be sensed by NK cells on the mAb-coated tumor cells triggering enhanced ADCC in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Carcinoma, Pancreatic Ductal/therapy , CpG Islands/immunology , Killer Cells, Natural/immunology , Mucin-1/immunology , Pancreatic Neoplasms/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Myeloid Differentiation Factor 88/immunology , Oligodeoxyribonucleotides/immunology , Pancreatic Neoplasms/immunology , Perforin/biosynthesis , Perforin/immunology , Up-Regulation/immunology
5.
BMC Cancer ; 11: 365, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21859454

ABSTRACT

BACKGROUND: Several studies have demonstrated that sites of chronic inflammation are often associated with the establishment and growth of various malignancies. A common inflammatory condition in humans is autoimmune arthritis (AA). Although AA and cancer are different diseases, many of the underlying processes that contribute to the disorders of the joints and connective tissue that characterize AA also affect cancer progression and metastasis. Systemically, AA can lead to cellular infiltration and inflammation of the lungs. Several studies have reported statistically significant risk ratios between AA and breast cancer. Despite this knowledge being available, there has been minimal research linking breast cancer, arthritis, and metastasis associated with breast cancer. Notably both diseases are extremely prevalent in older post-menopausal women. METHODS: To establish the novel link between arthritis induced inflammation and secondary metastasis associated with breast cancer, PyV MT mice that spontaneously develop mammary gland carcinoma were injected with Type II collagen (CII) to induce arthritis at 9 and 18 weeks of age for pre-metastatic and metastatic condition. The sites of secondary metastasis and the associated inflammatory microenvironment were evaluated. RESULTS: A significant increase in breast cancer-associated secondary metastasis to the lungs and bones was observed in the arthritic versus the non-arthritic PyV MT mice along with an increase in primary tumor burden. We report significant increases in the levels of interstitial cellular infiltrates and pro-inflammatory cytokines such as interleukin-17 (IL-17), interleukin-6 (IL-6), Pro- Matrix metallopeptidase 9 (Pro-MMP9), insulin like growth factor-II (GF-II) and macrophage colony stimulating factor (M-CSF) in the arthritic lung and bone milieu as well as in the circulation. These pro-inflammatory cytokines along with the inflammatory microenvironment may be the underlying factors facilitating tumor progression and metastasis in arthritic PyV MT mice. This was further substantiated by treatment with celecoxib, an anti-inflammatory drug + αIL-17 antibody that significantly reduced the secondary metastasis to lung and bone. CONCLUSIONS: The data generated not only reveal the underlying mechanism of high susceptibility to bone and lung metastasis in an arthritic condition but our combination therapies may lead to treatment modalities that will be capable of reducing tumor burden, and preventing relapse and metastasis in arthritic patients with breast cancer.


Subject(s)
Arthritis, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Celecoxib , Collagen Type II , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Enzyme Precursors/metabolism , Female , Histocytochemistry , Inflammation/metabolism , Inflammation/pathology , Insulin-Like Growth Factor II/metabolism , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Interleukin-6/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophage Colony-Stimulating Factor/metabolism , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Neoplasm Metastasis , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism
6.
Cancer Res ; 71(13): 4432-42, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21558393

ABSTRACT

MUC1 is overexpressed and aberrantly glycosylated in more than 60% of pancreatic ductal adenocarcinomas. The functional role of MUC1 in pancreatic cancer has yet to be fully elucidated due to a dearth of appropriate models. In this study, we have generated mouse models that spontaneously develop pancreatic ductal adenocarcinoma (KC), which are either Muc1-null (KCKO) or express human MUC1 (KCM). We show that KCKO mice have significantly slower tumor progression and rates of secondary metastasis, compared with both KC and KCM. Cell lines derived from KCKO tumors have significantly less tumorigenic capacity compared with cells from KCM tumors. Therefore, mice with KCKO tumors had a significant survival benefit compared with mice with KCM tumors. In vitro, KCKO cells have reduced proliferation and invasion and failed to respond to epidermal growth factor, platelet-derived growth factor, or matrix metalloproteinase 9. Further, significantly less KCKO cells entered the G(2)-M phase of the cell cycle compared with the KCM cells. Proteomics and Western blotting analysis revealed a complete loss of cdc-25c expression, phosphorylation of mitogen-activated protein kinase (MAPK), as well as a significant decrease in nestin and tubulin-α2 chain expression in KCKO cells. Treatment with a MEK1/2 inhibitor, U0126, abrogated the enhanced proliferation of the KCM cells but had minimal effect on KCKO cells, suggesting that MUC1 is necessary for MAPK activity and oncogenic signaling. This is the first study to utilize a Muc1-null PDA mouse to fully elucidate the oncogenic role of MUC1, both in vivo and in vitro.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Mucin-1/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Butadienes/pharmacology , Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle/physiology , Cell Growth Processes/physiology , Epidermal Growth Factor , Humans , Intermediate Filament Proteins/biosynthesis , Matrix Metalloproteinase 9 , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mucin-1/genetics , Neoplasm Metastasis , Nerve Tissue Proteins/biosynthesis , Nestin , Nitriles/pharmacology , Pancreatic Neoplasms/enzymology , Platelet-Derived Growth Factor , Protein Kinase Inhibitors/pharmacology , Tubulin/biosynthesis
7.
Cancer Immunol Immunother ; 59(12): 1801-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20824430

ABSTRACT

Since epithelial mucin 1 (MUC1) is associated with several adenocarcinomas at the mucosal sites, it is pertinent to test the efficacy of a mucosally targeted vaccine formulation. The B subunit of the Vibrio cholerae cholera toxin (CTB) has great potential to act as a mucosal carrier for subunit vaccines. In the present study we evaluated whether a MUC1 tandem repeat (TR) peptide chemically linked to CTB would break self-antigen tolerance in the transgenic MUC1-tolerant mouse model (MUC1.Tg) through oral or parenteral immunizations. We report that oral immunization with the CTB-MUC1 conjugate along with mucosal adjuvant, unmethylated CpG oligodeoxynucleotide (ODN) and interleukin-12 (IL-12) did not break self-antigen tolerance in MUC1.Tg mice, but induced a strong humoral response in wild-type C57BL/6 mice. However, self-antigen tolerance in the MUC1.Tg mouse model was broken after parenteral immunizations with different doses of the CTB-MUC1 conjugate protein and with the adjuvant CpG ODN co-delivered with CTB-MUC1. Importantly, mice immunized systemically with CpG ODN alone and with CTB-MUC1 exhibited decreased tumor burden when challenged with a mammary gland tumor cell line that expresses human MUC1.


Subject(s)
Cholera Toxin/immunology , Mucin-1/immunology , Neoplasms, Experimental/therapy , Animals , Autoantigens/immunology , Cell Line, Tumor , Humans , Immune Tolerance , Immunization , Injections, Intraperitoneal , Interleukin-12/pharmacology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology
8.
Breast Cancer Res ; 11(4): R56, 2009.
Article in English | MEDLINE | ID: mdl-19643025

ABSTRACT

INTRODUCTION: Sites of chronic inflammation are often associated with the establishment and growth of various malignancies including breast cancer. A common inflammatory condition in humans is autoimmune arthritis (AA) that causes inflammation and deformity of the joints. Other systemic effects associated with arthritis include increased cellular infiltration and inflammation of the lungs. Several studies have reported statistically significant risk ratios between AA and breast cancer. Despite this knowledge, available for a decade, it has never been questioned if the site of chronic inflammation linked to AA creates a milieu that attracts tumor cells to home and grow in the inflamed bones and lungs which are frequent sites of breast cancer metastasis. METHODS: To determine if chronic inflammation induced by autoimmune arthritis contributes to increased breast cancer-associated metastasis, we generated mammary gland tumors in SKG mice that were genetically prone to develop AA. Two breast cancer cell lines, one highly metastatic (4T1) and the other non-metastatic (TUBO) were used to generate the tumors in the mammary fat pad. Lung and bone metastasis and the associated inflammatory milieu were evaluated in the arthritic versus the non-arthritic mice. RESULTS: We report a three-fold increase in lung metastasis and a significant increase in the incidence of bone metastasis in the pro-arthritic and arthritic mice compared to non-arthritic control mice. We also report that the metastatic breast cancer cells augment the severity of arthritis resulting in a vicious cycle that increases both bone destruction and metastasis. Enhanced neutrophilic and granulocytic infiltration in lungs and bone of the pro-arthritic and arthritic mice and subsequent increase in circulating levels of proinflammatory cytokines, such as macrophage colony stimulating factor (M-CSF), interleukin-17 (IL-17), interleukin-6 (IL-6), vascular endothelial growth factor (VEGF), and tumor necrosis factor-alpha (TNF-alpha) may contribute to the increased metastasis. Treatment with anti-IL17 + celecoxib, an anti-inflammatory drug completely abrogated the development of metastasis and significantly reduced the primary tumor burden. CONCLUSIONS: The data clearly has important clinical implications for patients diagnosed with metastatic breast cancer, especially with regards to the prognosis and treatment options.


Subject(s)
Arthritis/complications , Autoimmune Diseases/complications , Bone Neoplasms/secondary , Cyclooxygenase 2/physiology , Interleukin-17/physiology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/therapeutic use , Arthritis/chemically induced , Arthritis/drug therapy , Arthritis/immunology , Arthritis/physiopathology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Bone Neoplasms/physiopathology , Bronchoalveolar Lavage Fluid/immunology , Celecoxib , Cell Line, Tumor/transplantation , Cyclooxygenase 2 Inhibitors/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Female , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Lung Neoplasms/physiopathology , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Organ Specificity , Osteoclasts/physiology , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , ZAP-70 Protein-Tyrosine Kinase/deficiency , ZAP-70 Protein-Tyrosine Kinase/genetics
9.
J Immunol ; 182(1): 216-24, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109152

ABSTRACT

With a 5-year survival rate of <5%, pancreatic cancer is one of the most rapidly fatal malignancies. Current protocols for the treatment of pancreas cancer are not as effective as we desire. In this study, we show that a novel Mucin-1 (MUC1)-based vaccine in combination with a cyclooxygenase-2 inhibitor (celecoxib), and low-dose chemotherapy (gemcitabine) was effective in preventing the progression of preneoplastic intraepithelial lesions to invasive pancreatic ductal adenocarcinomas. The study was conducted in an appropriate triple transgenic model of spontaneous pancreatic cancer induced by the KRAS(G12D) mutation and that expresses human MUC1 as a self molecule. The combination treatment elicited robust antitumor cellular and humoral immune responses and was associated with increased apoptosis in the tumor. The mechanism for the increased immune response was attributed to the down-regulation of circulating prostaglandin E(2) and indoleamine 2, 3,-dioxygenase enzymatic activity, as well as decreased levels of T regulatory and myeloid suppressor cells within the tumor microenvironment. The preclinical data provide the rationale to design clinical trials with a combination of MUC1-based vaccine, celecoxib, and gemcitabine for the treatment of pancreatic cancer.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/prevention & control , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/prevention & control , Cyclooxygenase 2 Inhibitors/administration & dosage , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/prevention & control , Adenocarcinoma/pathology , Animals , Antibodies/blood , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/pathology , Celecoxib , Cyclooxygenase 2/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Progression , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Transgenic , Mucin-1/administration & dosage , Mucin-1/immunology , Pancreatic Neoplasms/pathology , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Gemcitabine
10.
J Immunol ; 181(5): 3116-25, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713982

ABSTRACT

MUC1, a membrane tethered mucin glycoprotein, is overexpressed and aberrantly glycosylated in >80% of human ductal pancreatic adenocarcinoma. However, the role of MUC1 in pancreatic cancer has been elusive, partly due to the lack of an appropriate model. We report the characterization of a novel mouse model that expresses human MUC1 as a self molecule (PDA.MUC1 mice). Pancreatic tumors arise in an appropriate MUC1-tolerant background within an immune-competent host. Significant enhancement in the development of pancreatic intraepithelial preneoplastic lesions and progression to adenocarcinoma is observed in PDA.MUC1 mice, possibly due to increased proliferation. Tumors from PDA.MUC1 mice express higher levels of cyclooxygenase-2 and IDO compared with PDA mice lacking MUC1, especially during early stages of tumor development. The increased proinflammatory milieu correlates with an increased percentage of regulatory T cells and myeloid suppressor cells in the pancreatic tumor and tumor draining lymph nodes. Data shows that during pancreatic cancer progression, MUC1-mediated mechanisms enhance the onset and progression of the disease, which in turn regulate the immune responses. Thus, the mouse model is ideally suited for testing novel chemopreventive and therapeutic strategies against pancreatic cancer.


Subject(s)
Adenocarcinoma , Immune Tolerance , Mucin-1/physiology , Pancreatic Neoplasms/pathology , Animals , Cyclooxygenase 2/genetics , Disease Progression , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Transgenic , Models, Animal , Mucin-1/genetics , Pancreatic Neoplasms/immunology , Tumor Escape , Up-Regulation
11.
J Immunol ; 177(4): 2391-402, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16888001

ABSTRACT

We report that administration of celecoxib, a specific cyclooxygenase-2 (COX-2) inhibitor, in combination with a dendritic cell-based cancer vaccine significantly augments vaccine efficacy in reducing primary tumor burden, preventing metastasis, and increasing survival. This combination treatment was tested in MMTV-PyV MT mice that develop spontaneous mammary gland tumors with metastasis to the lungs and bone marrow. Improved vaccine potency was associated with an increase in tumor-specific CTLs. Enhanced CTL activity was attributed to a significant decrease in levels of tumor-associated IDO, a negative regulator of T cell activity. We present data suggesting that inhibiting COX-2 activity in vivo regulates IDO expression within the tumor microenvironment; this is further corroborated in the MDA-MB-231 human breast cancer cell line. Thus, a novel mechanism of COX-2-induced immunosuppression via regulation of IDO has emerged that may have implications in designing future cancer vaccines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Animals , Cancer Vaccines/immunology , Celecoxib , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Male , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Breast Cancer Res ; 7(4): R422-35, 2005.
Article in English | MEDLINE | ID: mdl-15987447

ABSTRACT

INTRODUCTION: Inhibitors of cyclo-oxygenase (COX)-2 are being extensively studied as anticancer agents. In the present study we evaluated the mechanisms by which a highly selective COX-2 inhibitor, celecoxib, affects tumor growth of two differentially invasive human breast cancer cell lines. METHODS: MDA-MB-231 (highly invasive) and MDA-MB-468 (moderately invasive) cell lines were treated with varying concentrations of celecoxib in vitro, and the effects of this agent on cell growth and angiogenesis were monitored by evaluating cell proliferation, apoptosis, cell cycle arrest, and vasculogenic mimicry. The in vitro results of MDA-MB-231 cell line were further confirmed in vivo in a mouse xenograft model. RESULTS: The highly invasive MDA-MB-231 cells express higher levels of COX-2 than do the less invasive MDA-MB-468 cells. Celecoxib treatment inhibited COX-2 activity, indicated by prostaglandin E2 secretion, and caused significant growth arrest in both breast cancer cell lines. In the highly invasive MDA-MB-231 cells, the mechanism of celecoxib-induced growth arrest was by induction of apoptosis, associated with reduced activation of protein kinase B/Akt, and subsequent activation of caspases 3 and 7. In the less invasive MDA-MB-468 cells, growth arrest was a consequence of cell cycle arrest at the G0/G1 checkpoint. Celecoxib-induced growth inhibition was reversed by addition of exogenous prostaglandin E2 in MDA-MB-468 cells but not in MDA-MB-231 cells. Furthermore, MDA-MB-468 cells formed significantly fewer extracellular matrix associated microvascular channels in vitro than did the high COX-2 expressing MDA-MB-231 cells. Celecoxib treatment not only inhibited cell growth and vascular channel formation but also reduced vascular endothelial growth factor levels. The in vitro findings corroborated in vivo data from a mouse xenograft model in which daily administration of celecoxib significantly reduced tumor growth of MDA-MB-231 cells, which was associated with reduced vascularization and increased necrosis in the tumor mass. CONCLUSION: The disparate molecular mechanisms of celecoxib-induced growth inhibition in human breast cancer cells depends upon the level of COX-2 expression and the invasive potential of the cell lines examined. Data suggest a role for COX-2 not only in the growth of cancer cells but also in activating the angiogenic pathway through regulating levels of vascular endothelial growth factor.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Cycle/drug effects , Cyclooxygenase 2/biosynthesis , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/biosynthesis
13.
Mol Cancer Res ; 2(11): 632-42, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15561779

ABSTRACT

Cyclooxygenase-2 (COX-2) inhibitors are rapidly emerging as a new generation of therapeutic drug in combination with chemotherapy or radiation therapy for the treatment of cancer. The mechanisms underlying its antitumor effects are not fully understood and more thorough preclinical trials are needed to determine if COX-2 inhibition represents a useful approach for prevention and/or treatment of breast cancer. The purpose of this study was to evaluate the growth inhibitory mechanism of a highly selective COX-2 inhibitor, celecoxib, in an in vivo oncogenic mouse model of spontaneous breast cancer that resembles human disease. The oncogenic mice carry the polyoma middle T antigen driven by the mouse mammary tumor virus promoter and develop primary adenocarcinomas of the breast. Results show that oral administration of celecoxib caused significant reduction in mammary tumor burden associated with increased tumor cell apoptosis and decreased proliferation in vivo. In vivo apoptosis correlated with significant decrease in activation of protein kinase B/Akt, a cell survival signaling kinase, with increased expression of the proapoptotic protein Bax and decreased expression of the antiapoptotic protein Bcl-2. In addition, celecoxib treatment reduced levels of proangiogenic factor (vascular endothelial growth factor), suggesting a role of celecoxib in suppression of angiogenesis in this model. Results from these preclinical studies will form the basis for assessing the feasibility of celecoxib therapy alone or in combination with conventional therapies for treatment and/or prevention of breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/secondary , Cyclooxygenase Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Breast Neoplasms/metabolism , Celecoxib , Cell Division/drug effects , Dinoprostone/blood , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein
14.
Breast Dis ; 20: 53-63, 2004.
Article in English | MEDLINE | ID: mdl-15687707

ABSTRACT

To study immunology in breast tumors, we have utilized a mammary gland adenocarcinoma model in which mice develop spontaneous tumors of the mammary gland which are initiated at puberty and express a human tumor antigen, MUC1. MUC1 (CD227) is over-expressed in 90% of human breast cancers and its glycosylation status and pattern of expression in cancer cells is altered. Humoral and cellular responses to MUC1 have been reported in breast cancer patients and therefore, MUC1 is being evaluated as a target for immune intervention. This mouse model of spontaneous breast cancer allows the evaluation of anti-MUC1 immune responses at all stages of the disease. In this report, we review the model as it pertains to a) the development of the tumor, b) MUC1 expression, and the native immune responses against MUC1 as tumors progress, and c) the immune suppressive microenvironment within the developing tumor. Finally, we report our latest findings describing the therapeutic efficacy of adoptively transferred MUC1-specific cytotoxic T lymphocytes (MUC1-CTL) in these mice and discuss ways to increase their effectiveness by agonistic monoclonal antibody against CD137 T cell costimulatory molecule.


Subject(s)
Breast Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Mucin-1/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD , Breast Neoplasms/immunology , Disease Models, Animal , Humans , Immunotherapy , Mammary Neoplasms, Animal/immunology , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 9
15.
J Immunother ; 26(1): 47-62, 2003.
Article in English | MEDLINE | ID: mdl-12514429

ABSTRACT

Human mucin 1 (MUC1) is an epithelial mucin glycoprotein that is overexpressed in 90% of all adenocarcinomas including breast, lung, pancreas, prostate, stomach, colon, and ovary. MUC1 is a target for immune intervention, because, in patients with solid adenocarcinomas, low-level cellular and humoral immune responses to MUC1 have been observed, which are not sufficiently strong to eradicate the growing tumor. The hypothesis for this study is that enhancing MUC1-specific immunity will result in antitumor immunity. To test this, the authors have developed a clinically relevant breast cancer model that demonstrates peripheral and central tolerance to MUC1 and develops spontaneous tumors of the mammary gland. In these mice, the authors tested a vaccine formulation comprised of liposomal-MUC1 lipopeptide and human recombinant interleukin-2. Results indicate that when compared with untreated mice, immunized mice develop T cells that express intracellular IFN-gamma, are reactive with MHC class I H-2Db/MUC1 tetramer, and are cytotoxic against MUC1-expressing tumor cells in vitro. The presence of MUC1-specific CTL did not translate into a clinical response as measured by time of tumor onset, tumor burden, and survival. The authors demonstrate that some of the immune-evasion mechanisms used by the tumor cells include downregulation of MHC-class I molecule, expression of TGF-beta2, and decrease in IFN-gamma -expressing effector T cells as tumors progress. Finally, utilizing an injectable breast cancer model, the authors show that targeting a single tumor antigen may not be an effective antitumor treatment, but that immunization with dendritic cells fed with whole tumor lysate is effective in breaking tolerance and protecting mice from subsequent tumor challenge. A physiologically relevant spontaneous breast cancer model has been developed to test improved immunotherapeutic approaches.


Subject(s)
Immunotherapy/methods , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Mucin-1/immunology , Analysis of Variance , Animals , Base Sequence , Biopsy, Needle , Cancer Vaccines/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immune Tolerance , Immunohistochemistry , Male , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Polymerase Chain Reaction/methods , Sensitivity and Specificity , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...