Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cancer ; 5(8): 633-45, 2014.
Article in English | MEDLINE | ID: mdl-25157274

ABSTRACT

Bevacizumab, the recombinant antibody targeting vascular endothelial growth factor (VEGF), improves progression-free but not overall survival in metastatic breast cancer. To seek further insights in resistance mechanisms to bevacizumab at the molecular level, we developed VEGF and non-VEGF-driven ER-positive MCF7-derived xenograft models allowing comparison of tumor response at different timepoints. VEGF gene (MV165) overexpressing xenografts were initially sensitive to bevacizumab, but eventually acquired resistance. In contrast, parental MCF7 cells derived tumors were de novo insensitive to bevacizumab. Microarray analysis with qRT-PCR validation revealed that Follistatin (FST) and NOTCH were the top signaling pathways associated with resistance in VEGF-driven tumors (P<0.05). Based on the presence of VEGF, treatment with bevacizumab resulted in altered patterns of metagenes and PAM50 gene expression. In VEGF-driven model after short and long-term bevacizumab treatments, a change in the intrinsic subtype (luminal to myoepithelial/basal-like) was observed in association with increased expression of genes implicated with cancer stem cell phenotype (P<0.05). Our results show that the presence or absence of VEGF expression affects the response to bevacizumab therapy and gene pathways. In particular, long-term bevacizumab treatment shifts the cancer cells to a more aggressive myoepithelial/basal subtype in VEGF-expressing model, but not in non-VEGF model. These findings could shed light on variable results to anti-VEGF therapy in patients and emphasize the importance of patient stratification based on the VEGF expression. Our data strongly suggest consideration of patient subgroups for treatment and designing novel combinatory therapies in the clinical setting.

2.
Breast Cancer Res Treat ; 136(3): 673-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23085766

ABSTRACT

Aberrant activation of the mammalian target of rapamycin (mTOR) signaling plays an important role in breast cancer progression and represents a potential therapeutic target for breast cancer. In this study, we report the impact of the investigational drug MLN0128, a potent and selective small molecule active-site TORC1/2 kinase inhibitor, on tumor growth and metastasis using human breast cancer xenograft models. We assessed in vitro antiproliferative activity of MLN0128 in a panel of breast cancer cell lines. We next evaluated the impact of MLN0128 on tumor growth, angiogenesis and metastasis using mammary fat pad xenograft models of a non-VEGF (ML20) and a VEGF-driven (MV165) MCF-7 sublines harboring PIK3CA mutations. MLN0128 potently inhibited cell proliferation in various breast cancer cell lines harboring PIK3CA (IC(50): 1.5-53 nM), PTEN (IC(50): 1-149 nM), KRAS, and/or BRAF mutations (IC(50): 13-162 nM), and in human endothelial cells (IC(50): 33-40 nM) in vitro. In vivo, MLN0128 decreased primary tumor growth significantly in both non-VEGF (ML20; p = 0.05) and VEGF-driven MCF-7 (MV165; p = 0.014) xenograft models. MLN0128 decreased the phosphorylation of Akt, S6, 4E-BP1, and NDRG1 in both models. In contrast, rapamycin increased Akt activity and failed to reduce the phosphorylation of 4E-BP1, PRAS40, and NDRG1. VEGF-induced lung metastasis in MV165 is inhibited by MLN0128 and rapamycin. In conclusion, MLN0128 inhibits TORC1/2-dependent signaling in preclinical models of breast cancer. MLN0128 appears to be superior in blocking mTORC1/2 signaling in contrast to rapamycin. Our findings support the clinical research of MLN0128 in patients with breast cancer and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoxazoles/pharmacology , Breast Neoplasms/drug therapy , Multiprotein Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/blood , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Nude , Multiprotein Complexes/metabolism , Mutation , Neovascularization, Pathologic/drug therapy , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/blood , Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
3.
Lab Invest ; 92(11): 1564-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926645

ABSTRACT

Thymomas are low-grade epithelial tumors of the anterior mediastinum. The complexity of the disease and the lack of in vitro and in vivo models hamper the development of better therapeutics. In this study, we report a novel cell line, designated as IU-TAB-1, which was established from a patient with stage II thymoma (World Health Organization-type AB). The IU-TAB-1 cell line was established in vitro and characterized using histological and immunohistochemical staining, fluorescence-activated cell sorting, cytogenetic analyses and functional assays including in vitro and a NOD/SCID xenograft model. A whole-genome gene expression analysis (Illumina) was performed on the IU-TAB-1 cell line and 34 thymomas to determine the clinical relevance of the cell line. The IU-TAB-1 cell line was positive for epithelial markers (pan-cytokeratin and EpCAM/CD326) including thymic epithelial (TE) surface markers (such as CD29, CD9, CD54/ICAM-1, CD58 and CD24) and p63, and negative for B- and T-cell lineage markers. Gene expression profiling demonstrated overlapping and distinct genes between IU-TAB-1 and primary thymomas including the primary tumor (from which the cell line was derived). IU-TAB-1 cells are tumorigenic when implanted in immunodeficient mice with tumors reaching a volume of 1000 mm³ at around 130 days. The established cell line represents a biologically relevant new tool to investigate the molecular pathology of thymic malignancies and to evaluate the efficacy of novel therapeutics both in vitro and in vivo.


Subject(s)
Cell Line, Tumor , Thymoma/pathology , Thymus Neoplasms/pathology , Animals , Cell Proliferation , Chromosome Aberrations , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged
4.
Breast Cancer Res Treat ; 124(2): 327-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20099025

ABSTRACT

Protein kinase C betaII (PKCßII) represents a novel potential target for anticancer therapies in breast cancer. In order to identify patient subgroups which might benefit from PKC-targeting therapies, we investigated the expression of PKCßII in human breast cancer cell lines and in a tissue microarray (TMA). We first screened breast cancer cell line representatives of breast cancer subtypes for PKCßII expression at the mRNA and at the protein levels. We analyzed a TMA comprising of tumors from 438 patients with a median followup of 15.4 years for PKCßII expression by immunohistochemistry along with other prognostic factors in breast cancer. Among a panel of human breast cancer cell lines, only MDA-MB-436, a triple negative basal cell line, showed overexpression for PKCßII both at the mRNA and at the protein levels. In breast cancer patients, cytoplasmic expression of PKCßII correlated positively with human epidermal growth factor receptor-2 (HER-2; P = 0.01) and Ki-67 (P = 0.016), while nuclear PKCßII correlated positively with estrogen receptor (ER; P = 0.016). The positive correlation of CK5/6 with cytoplasmic PKCßII (P = 0.033) lost statistical significance after adjusting for multiple comparisons (P = 0.198). Cytoplasmic PKCßII did not correlate with cyclooxygenase (COX-2; P = 0.925) and vascular endothelial growth factor (P = 1). There was no significant association between PKCßII staining and overall survival. Cytoplasmic PKCßII correlates with HER-2 and Ki-67, while nuclear PKCßII correlates with ER in breast cancer. Our study suggests the necessity for assessing the subcellular localization of PKCßII in breast cancer subtypes when evaluating the possible effectiveness of PKCßII-targeting agents.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Protein Kinase C/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/enzymology , Cyclooxygenase 2/metabolism , Cytoplasm/enzymology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Keratin-5/metabolism , Keratin-6/metabolism , Ki-67 Antigen/metabolism , Prognosis , Protein Kinase C/genetics , Protein Kinase C beta , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Time Factors , Tissue Array Analysis , Vascular Endothelial Growth Factor A/metabolism
5.
Cancer Res ; 65(20): 9406-14, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230404

ABSTRACT

2-Methoxyestradiol is an estradiol metabolite with significant antiproliferative and antiangiogenic activity independent of estrogen receptor status. To identify a molecular basis for acquired 2-methoxyestradiol resistance, we generated a stable 2-methoxyestradiol-resistant (2ME2R) MDA-MB-435 human cancer cell line by stepwise exposure to increasing 2-methoxyestradiol concentrations. 2ME2R cells maintained in the presence of the drug and W435 cells maintained in the absence of the drug showed 32.34- to 40.07-fold resistance to 2-methoxyestradiol. Cross-resistance was observed to Vinca alkaloids, including vincristine, vinorelbine, and vinblastine (4.29- to 6.40-fold), but minimal resistance was seen to colchicine-binding agents including colchicine, colcemid, and AVE8062A (1.72- to 2.86-fold). No resistance was observed to paclitaxel and epothilone B, polymerizing agents (0.89- to 1.14-fold). Genomic sequencing identified two different heterozygous point mutations in the class I (M40) isotype of beta-tubulin at amino acids 197 (Dbeta197N) and 350 (Kbeta350N) in 2ME2R cells. Tandem mass spectrometry confirmed the presence of both wild-type and the mutant beta-tubulin in 2ME2R cells at the protein level. Consistently, treatment of parental P435 cells with 2-methoxyestradiol resulted in a dose-dependent depolymerization of microtubules, whereas 2ME2R cells remained unaffected. In contrast, paclitaxel affected both cell lines. In the absence of 2-methoxyestradiol, 2ME2R cells were characterized by an elevated level of detyrosination. Upon 2-methoxyestradiol treatment, levels of acetylated and detyrosinated tubulins decreased in P435 cells, while remaining constant in 2ME2R cells. These results, together with our structure-based modeling, show a tight correlation between the antitubulin and antiproliferative effects of 2-methoxyestradiol, consistent with acquired tubulin mutations contributing to 2-methoxyestradiol resistance.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estradiol/analogs & derivatives , Point Mutation , Tubulin Modulators/pharmacology , Tubulin/genetics , 2-Methoxyestradiol , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm , Estradiol/pharmacology , Humans , Mass Spectrometry , Microtubules/drug effects , Microtubules/metabolism , Models, Molecular , Molecular Sequence Data , Protein Isoforms , Structure-Activity Relationship , Tubulin/metabolism
6.
J Clin Immunol ; 24(4): 426-34, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15163899

ABSTRACT

Asthma is characterized by infiltration and shedding of the bronchial epithelium. The Th2 cytokines IL-4 and IL-13 are involved in the cellular recruitment and infiltration seen in asthma. The effects of IL-4 and IL-13 on cell-matrix interactions and epithelial shedding are unknown. We hypothesize that bronchial airway epithelial cells (BAEC) express paxillin, a structural focal adhesion protein, and downregulation of paxillin by Th2 cytokines lead to BAEC hyperpermeability. We showed by confocal microscopy the presence of paxillin in BAEC. We demonstrated by Western blot analysis that IL-4 and IL-13 stimulation results in downregulation of paxillin production. IL-4 and IL-13 stimulation decreased epithelial cell-matrix attachment as measured by electrical cell-substrate impedance sensing system (ECIS). Our results suggest that Th2 cytokines IL-4 and IL-13 downregulate paxillin production by BAEC, thereby disrupting the cell-matrix interactions. This may help explain the epithelial shedding and epithelial membrane hyperpermeability that occurs in asthma.


Subject(s)
Bronchi/cytology , Cytoskeletal Proteins/genetics , Epithelial Cells/drug effects , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Phosphoproteins/genetics , Cell Adhesion/drug effects , Cells, Cultured , Down-Regulation/drug effects , Humans , Paxillin , Permeability/drug effects , Th2 Cells , Vascular Endothelial Growth Factor A/pharmacology
7.
J Clin Immunol ; 23(1): 1-10, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12645855

ABSTRACT

In bacterial empyema, the recruited polymorphonuclear leukocytes (PMN) represent important phagocytic cells involved in antibacterial defense. In this study we demonstrate that pleural fluids (PF) obtained from patients with empyema (EMP) contains significantly higher levels of granulocyte colony stimulating factor (GM-CSF), and PMN incubated in empyema (EMP) pleural fluid (PF) showed significantly less apoptosis than congestive heart failure (CHF) PF. Staphylococcus aureus-stimulated PMC released significantly (P < 0.001) higher levels of GM-CSF than resting PMC. Staphylococcus aureus-stimulated PMC (SPMC)-CM significantly (P < 0.001) inhibited PMN apoptosis. In SPMC-CM-incubated PMN the antiapoptotic gene Bcl-xL mRNA and protein expression was up-regulated; Bak mRNA and protein expression was down-regulated compared to control PMN. The active caspases activity significantly decreased. When SPMC-CM and EMP PF were immunodepleted with GM-CSF antibody, PMN apoptosis was significantly higher. The delay in apoptosis of PMN is in part attributable to the release of cytokine GM-CSF by activated PMC. These findings suggest that S. aureus-activated PMC extend PMN life span by modulating Bcl-xL and Bak gene expression and active caspases activity during acute inflammation and empyema.


Subject(s)
Apoptosis/physiology , Empyema, Pleural/metabolism , Neutrophils/metabolism , Pleural Cavity/metabolism , Caspases/metabolism , Epithelium/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neutrophils/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Staphylococcus aureus/immunology , bcl-2 Homologous Antagonist-Killer Protein , bcl-X Protein
8.
J Clin Immunol ; 22(3): 144-52, 2002 May.
Article in English | MEDLINE | ID: mdl-12078856

ABSTRACT

The repair of an injured bronchial epithelial cell (BEC) monolayer requires proliferation and migration of BECs into the injured area. We hypothesized that BEC monolayer injury results in monocyte chemoattractant protein-1 (MCP-1) production, which initiates the repair process. BECs (BEAS-2B from ATCC) were utilized in this study. MCP-1 interacts with CCR2B receptor (CCR2B), resulting in cell proliferation, haptotaxis, and healing of the monolayer. Reverse transcriptase-polymerase chain reaction (RT-PCR) was employed to verify the presence of CCR2B. CCR2B was not merely present but also inducible by interleukin-2 (IL-2) and lipopolysaccharide (LPS). We demonstrated by immunohistochemistry that BECs express MCP-1 after injury and that receptor expression can be regulated by exposure to IL-2 and LPS. Haptotactic migration of cells was enhanced in the presence of MCP-1 and reduced in the presence of CCR2B antibody. This enhanced or depressed ability of the BECs to perform haptotactic migration was shown to be statistically significant (P < 0.05) when compared to controls. Finally, BECs proliferate in response to MCP-1 as proven by electric cell-substrate impedance sensing (ECIS) technology. MCP-1-specific antibodies were shown to neutralize the MCP-1-mediated BEC proliferation. This cascade of events following injury to the bronchial epithelium may provide insight into the mechanism of the repair process.


Subject(s)
Bronchi/cytology , Chemokine CCL2/physiology , Epithelial Cells/pathology , Cell Division , Cell Line, Transformed , Cell Movement , Chemokine CCL2/biosynthesis , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-2/pharmacology , Lipopolysaccharides/pharmacology , Receptors, CCR2 , Receptors, Chemokine/drug effects , Receptors, Chemokine/physiology
9.
Inflammation ; 26(2): 73-82, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11989790

ABSTRACT

Current knowledge implicates pleural mesothelial cells as mainly responsible for inflammatory responses in the pleural space. However, a vast body of recent evidence underscores the important role of fibroblasts in the process of inflammation in several types of tissues. We hypothesize that HPFBs (human pleural fibroblasts) play an important role in pleural responses and also when activated by bacterial endotoxin LPS (lipopolysaccharide), IL-1 beta (interleukin-1 beta), or TNF-alpha (tumor necrosis factor-alpha) release of C-C and C-X-C chemokines-specifically, MCP-1 and IL-8. Our results show that pleural fluid-isolated human fibroblasts release IL-8 and MCP-1 upon stimulation with IL-1 beta, TNF-alpha, and LPS in both a concentration- and time-dependent manner. RT-PCR (reverse-transcriptase-polymerase chain reaction) studies have also confirmed IL-8- and MCP-1-specific mRNA expression in activated pleural fibroblasts. On the time-dependent response curve, IL-8 was found in maximum concentrations at 144 hr, whereas MCP-1 continued to increase even after 196 hr following stimulation. IL-1 beta induced the maximum release of IL-8 (800-fold) and MCP-1 (164-fold), as compared to the controls. TNF-alpha induced a 95-fold increase in IL-8 and an 84-fold increase in MCP-1 levels, as compared to the controls. Collectively, our results show that human pleural fibroblasts contribute to the inflammatory cascade in the pleural space.


Subject(s)
Chemokine CCL2/biosynthesis , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Interleukin-8/biosynthesis , Pleura/cytology , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CCL2/genetics , Chemotaxis, Leukocyte/drug effects , DNA, Complementary/genetics , Fibroblasts/metabolism , Humans , Interleukin-8/genetics , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Neutrophils/drug effects , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...