Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Clin Cancer Res ; 27(5): 1278-1286, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33277370

ABSTRACT

PURPOSE: Immunotherapy is currently ineffective for nearly all pancreatic ductal adenocarcinomas (PDAC), largely due to its tumor microenvironment (TME) that lacks antigen-experienced T effector cells (Teff). Vaccine-based immunotherapies are known to activate antigen-specific Teffs in the peripheral blood. To evaluate the effect of vaccine therapy on the PDAC TME, we designed a neoadjuvant and adjuvant clinical trial of an irradiated, GM-CSF-secreting, allogeneic PDAC vaccine (GVAX). PATIENTS AND METHODS: Eighty-seven eligible patients with resectable PDAC were randomly assigned (1:1:1) to receive GVAX alone or in combination with two forms of low-dose cyclophosphamide. Resected tumors following neoadjuvant immunotherapy were assessed for the formation of tertiary lymphoid aggregates (TLA) in response to treatment. The clinical endpoints are disease-free survival (DFS) and overall survival (OS). RESULTS: The neoadjuvant treatment with GVAX either alone or with two forms of low-dose cyclophosphamide is safe and feasible without adversely increasing the surgical complication rate. Patients in Arm A who received neoadjuvant and adjuvant GVAX alone had a trend toward longer median OS (35.0 months) than that (24.8 months) in the historical controls who received adjuvant GVAX alone. However, Arm C, who received low-dose oral cyclophosphamide in addition to GVAX, had a significantly shorter DFS than Arm A. When comparing patients with OS > 24 months to those with OS < 15 months, longer OS was found to be associated with higher density of intratumoral TLA. CONCLUSIONS: It is safe and feasible to use a neoadjuvant immunotherapy approach for PDACs to evaluate early biologic responses. In-depth analysis of TLAs is warranted in future neoadjuvant immunotherapy clinical trials.


Subject(s)
Adjuvants, Vaccine/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/mortality , Cyclophosphamide/administration & dosage , Lymphocytes/pathology , Neoadjuvant Therapy/mortality , Pancreatic Neoplasms/mortality , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Feasibility Studies , Female , Follow-Up Studies , Humans , Immunotherapy , Lymphocytes/drug effects , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , Survival Rate
2.
J Oncol ; 2010: 856105, 2010.
Article in English | MEDLINE | ID: mdl-20414347

ABSTRACT

Id1, which belongs to the Id family of helix-loop-helix transcription factors has been most associated with tumor progression and metastatsis; however, its significance in lung cancers has not been extensively explored. Here we seek to evaluate the expression of Id1 in a pilot study of nonsmall-cell lung cancers (NSCLCs) and determine its diagnostic and functional significance in these tumors. Paired normal and malignant lung tissues as well as a panel of NSCLC primary tumors and cell lines were evaluated for Id1 expression using Western blotting and quantitative RT-PCR. Functional assays were performed to evaluate the role of Id1 in tumor cell growth, migration and progression. We find Id1 expression is upregulated in squamous cell carcinoma when compared to adenocarcinoma of the lung and that expression of Id1 versus the normal control is variable in NSCLCs. We also note that Id1 expression in NSCLC cells is largely growth factor dependant and constitutive expression of Id1 in NSCLC cells significantly increases tumor cell migration without affecting cell proliferation. We conclude that Id1, as a mediator of tumor cell migration, may be an indicator of aggressive potential in nonsmall-cell lung cancers.

3.
Mol Cancer Res ; 7(11): 1803-12, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903768

ABSTRACT

Although lung cancer is the leading cause of cancer death worldwide, the precise molecular mechanisms that give rise to lung cancer are incompletely understood. Here, we show that HMGA1 is an important oncogene that drives transformation in undifferentiated, large-cell carcinoma. First, we show that the HMGA1 gene is overexpressed in lung cancer cell lines and primary human lung tumors. Forced overexpression of HMGA1 induces a transformed phenotype with anchorage-independent cell growth in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA1 expression blocks anchorage-independent cell growth in the H1299 metastatic, undifferentiated, large-cell human lung carcinoma cells. We also show that the matrix metalloproteinase-2 (MMP-2) gene is a downstream target upregulated by HMGA1 in large-cell carcinoma cells. In chromatin immunoprecipitation experiments, HMGA1 binds directly to the MMP-2 promoter in vivo in large-cell lung cancer cells, but not in squamous cell carcinoma cells. In large-cell carcinoma cell lines, there is a significant, positive correlation between HMGA1 and MMP-2 mRNA. Moreover, interfering with MMP-2 expression blocks anchorage-independent cell growth in H1299 large-cell carcinoma cells, indicating that the HMGA1-MMP-2 pathway is required for this transformation phenotype in these cells. Blocking MMP-2 expression also inhibits migration and invasion in the H1299 large-cell carcinoma cells. Our findings suggest an important role for MMP-2 in transformation mediated by HMGA1 in large-cell, undifferentiated lung carcinoma and support the development of strategies to target this pathway in selected tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/metabolism , HMGA1a Protein/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/biosynthesis , Carcinoma, Large Cell/enzymology , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/metabolism , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/biosynthesis , HMGA1a Protein/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection , Up-Regulation
4.
Cancer Res ; 68(24): 10121-7, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19074878

ABSTRACT

Although HMGA1 (high-mobility group A1; formerly HMG-I/Y) is an oncogene that is widely overexpressed in aggressive cancers, the molecular mechanisms underlying transformation by HMGA1 are only beginning to emerge. HMGA1 encodes the HMGA1a and HMGA1b protein isoforms, which function in regulating gene expression. To determine how HMGA1 leads to neoplastic transformation, we looked for genes regulated by HMGA1 using gene expression profile analysis. Here, we show that the STAT3 gene, which encodes the signaling molecule signal transducer and activator of transcription 3 (STAT3), is a critical downstream target of HMGA1a. STAT3 mRNA and protein are up-regulated in fibroblasts overexpressing HMGA1a and activated STAT3 recapitulates the transforming activity of HMGA1a in fibroblasts. HMGA1a also binds directly to a conserved region of the STAT3 promoter in vivo in human leukemia cells by chromatin immunoprecipitation and activates transcription of the STAT3 promoter in transfection experiments. To determine if this pathway contributes to HMGA1-mediated transformation, we investigated STAT3 expression in our HMGA1a transgenic mice, all of which developed aggressive lymphoid malignancy. STAT3 expression was increased in the leukemia cells from our transgenics but not in control cells. Blocking STAT3 function induced apoptosis in the transgenic leukemia cells but not in controls. In primary human leukemia samples, there was a positive correlation between HMGA1a and STAT3 mRNA. Moreover, blocking STAT3 function in human leukemia or lymphoma cells led to decreased cellular motility and foci formation. Our results show that the HMGA1a-STAT3 axis is a potential Achilles heel that could be exploited therapeutically in hematopoietic and other malignancies overexpressing HMGA1a.


Subject(s)
HMGA1a Protein/genetics , Hematologic Neoplasms/genetics , STAT3 Transcription Factor/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HMGA1a Protein/biosynthesis , HMGA1a Protein/metabolism , Hematologic Neoplasms/metabolism , Humans , Mice , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/metabolism , Transfection , Up-Regulation
5.
Mol Cancer Res ; 6(5): 743-50, 2008 May.
Article in English | MEDLINE | ID: mdl-18505920

ABSTRACT

Although previous studies have established a prominent role for HMGA1 (formerly HMG-I/Y) in aggressive human cancers, the role of HMGA2 (formerly HMGI-C) in malignant transformation has not been clearly defined. The HMGA gene family includes HMGA1, which encodes the HMGA1a and HMGA1b protein isoforms, and HMGA2, which encodes HMGA2. These chromatin-binding proteins function in transcriptional regulation and recent studies also suggest a role in cellular senescence. HMGA1 proteins also appear to participate in cell cycle regulation and malignant transformation, whereas HMGA2 has been implicated primarily in the pathogenesis of benign, mesenchymal tumors. Here, we show that overexpression of HMGA2 leads to a transformed phenotype in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA2 expression blocks the transformed phenotype in metastatic human non-small cell lung cancer cells. Moreover, we show that HMGA2 mRNA and protein are overexpressed in primary human lung cancers compared with normal tissue or indolent tumors. In addition, there is a statistically significant correlation between HMGA2 protein staining by immunohistochemical analysis and tumor grade (P < 0.001). Our results indicate that HMGA2 is an oncogene important in the pathogenesis of human lung cancer. Although additional studies with animal models are needed, these findings suggest that targeting HMGA2 could be therapeutically beneficial in lung cancer and other cancers characterized by increased HMGA2 expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , HMGA2 Protein/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Chromatin/metabolism , HMGA2 Protein/metabolism , Humans , Immunohistochemistry/methods , Phenotype , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic
6.
Cancer Res ; 64(10): 3371-5, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15150086

ABSTRACT

HMG-I/Y is overexpressed in human cancer, although a direct role for this gene in transformation has not been established. We generated transgenic mice with HMG-I targeted to lymphoid cells. All seven informative founder HMG-I mice developed aggressive lymphoma by a mean age of 4.8 months. Tumors express T-cell markers and are transplantable. We also demonstrate that HMG-I mRNA and protein are increased in human acute lymphocytic leukemia samples. Our results show that HMG-I functions as an oncogene and suggest that it contributes to the pathogenesis of leukemia and other cancers with increased HMG-I expression.


Subject(s)
Cell Transformation, Neoplastic/genetics , HMGB1 Protein/genetics , Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Female , HMGB1 Protein/biosynthesis , Humans , Lymphoma/metabolism , Lymphoma/pathology , Male , Mice , Mice, Transgenic , Neoplasm Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transgenes
7.
Nat Med ; 10(1): 48-54, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702634

ABSTRACT

Although tumor progression involves processes such as tissue invasion that can activate inflammatory responses, the immune system largely ignores or tolerates disseminated cancers. The mechanisms that block initiation of immune responses during cancer development are poorly understood. We report here that constitutive activation of Stat-3, a common oncogenic signaling pathway, suppresses tumor expression of proinflammatory mediators. Blocking Stat-3 in tumor cells increases expression of proinflammatory cytokines and chemokines that activate innate immunity and dendritic cells, leading to tumor-specific T-cell responses. In addition, constitutive Stat-3 activity induces production of pleiotropic factors that inhibit dendritic cell functional maturation. Tumor-derived factors inhibit dendritic cell maturation through Stat-3 activation in progenitor cells. Thus, inhibition of antitumor immunity involves a cascade of Stat-3 activation propagating from tumor to dendritic cells. We propose that tumor Stat-3 activity can mediate immune evasion by blocking both the production and sensing of inflammatory signals by multiple components of the immune system.


Subject(s)
DNA-Binding Proteins/physiology , Immunity, Innate/physiology , Melanoma, Experimental/immunology , Signal Transduction/physiology , Trans-Activators/physiology , Animals , Cell Line, Tumor , Dendritic Cells/immunology , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Melanoma, Experimental/metabolism , Mice , STAT3 Transcription Factor
8.
J Immunol ; 172(1): 464-74, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14688356

ABSTRACT

Abnormal differentiation of myeloid cells is one of the hallmarks of cancer. However, the molecular mechanisms of this process remain elusive. In this study, we investigated the effect of tumor-derived factors on Janus kinase (Jak)/STAT signaling in myeloid cells during their differentiation into dendritic cells. Tumor cell conditioned medium induced activation of Jak2 and STAT3, which was associated with an accumulation of immature myeloid cells. Jak2/STAT3 activity was localized primarily in these myeloid cells, which prevented the differentiation of immature myeloid cells into mature dendritic cells. This differentiation was restored after removal of tumor-derived factors. Inhibition of STAT3 abrogated the negative effects of these factors on myeloid cell differentiation, and overexpression of STAT3 reproduced the effects of tumor-derived factors. Thus, this is a first demonstration that tumor-derived factors may affect myeloid cell differentiation in cancer via constitutive activation of Jak2/STAT3.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Proto-Oncogene Proteins , Signal Transduction/immunology , Trans-Activators/metabolism , Animals , Cell Aggregation/immunology , Cell Differentiation/immunology , Cell Division/immunology , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , DNA-Binding Proteins/physiology , Dendritic Cells/enzymology , Female , Growth Inhibitors/physiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Janus Kinase 2 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/pathology , NIH 3T3 Cells , Neoplasm Proteins/physiology , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Trans-Activators/physiology , Up-Regulation/immunology
9.
Cell Biol Int ; 26(2): 131-44, 2002.
Article in English | MEDLINE | ID: mdl-11846443

ABSTRACT

During corneal development, neural crest derivatives from the periocular mesenchyme migrate into the cornea and differentiate into corneal fibroblasts. During this time, these cells interact with a variety of extracellular matrices for proper orientation and development. In the present studies, we have examined the interaction of beta(1) integrins on periocular mesenchyme cells (POM) and corneal fibroblasts (CF) with fibronectin and tenascin by perturbing the function of this integrin. POM and CF attached and spread to a much greater extent on fibronectin than on tenascin. An antibody against beta(1) integrin, CSAT, decreased spreading and attachment, and resulted in a lack of immuno-detectable beta(1) integrin in focal adhesions on fibronectin; few beta(1) positive focal adhesions were observed in cells grown on tenascin. An anti-sense retroviral construct decreased endogenous levels of beta(1) integrin protein, and caused decreased attachment and spreading as well as sparse, disorganized focal adhesions. These data indicate that in vitro, both POM and CF have beta(1) integrins that interact with fibronectin and allow them to attach and spread, while tenascin is anti-adhesive. Further studies using both of these experimental paradigms will clarify whether these interactions also occur in vivo.


Subject(s)
Cell Culture Techniques/methods , Cornea/cytology , Fibronectins/metabolism , Integrin beta1/metabolism , Integrin beta1/physiology , Oligonucleotides, Antisense/pharmacology , Tenascin/metabolism , Animals , Cattle , Cell Adhesion , Cells, Cultured , Chick Embryo , Chickens , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Immunohistochemistry , Integrins/metabolism , Microscopy, Fluorescence , Protein Binding , Retroviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...