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










Publication year range
1.
Oncotarget ; 7(13): 15648-61, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26871292

ABSTRACT

We previously showed how key pathways in cancer-related inflammation and Notch signaling are part of an autocrine malignant cell network in ovarian cancer. This network, which we named the "TNF network", has paracrine actions within the tumor microenvironment, influencing angiogenesis and the immune cell infiltrate.The aim of this study was to identify critical regulators in the signaling pathways of the TNF network in ovarian cancer cells that might be therapeutic targets. To achieve our aim, we used a systems biology approach, combining data from phospho-proteomic mass spectrometry and gene expression array analysis. Among the potential therapeutic kinase targets identified was the protein kinase Casein kinase II (CK2).Knockdown of CK2 expression in malignant cells by siRNA or treatment with the specific CK2 inhibitor CX-4945 significantly decreased Notch signaling and reduced constitutive cytokine release in ovarian cancer cell lines that expressed the TNF network as well as malignant cells isolated from high grade serous ovarian cancer ascites. The expression of the same cytokines was also inhibited after treatment with CX-4945 in a 3D organotypic model. CK2 inhibition was associated with concomitant inhibition of proliferative activity, reduced angiogenesis and experimental peritoneal ovarian tumor growth.In conclusion, we have identified kinases, particularly CK2, associated with the TNF network that may play a central role in sustaining the cytokine network and/or mediating its effects in ovarian cancer.


Subject(s)
Inflammation/enzymology , Ovarian Neoplasms/pathology , Signal Transduction/immunology , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Female , Gene Expression Profiling/methods , Heterografts , Humans , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/immunology , Proteomics/methods , Systems Biology/methods , Transcriptome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
2.
Cancer Res ; 75(7): 1255-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25670170

ABSTRACT

Excess production of the proinflammatory IL6 has both local and systemic tumor-promoting activity in many cancers, including ovarian cancer. However, treatment of advanced ovarian cancer patients with a neutralizing IL6 antibody yielded little efficacy in a previous phase II clinical trial. Here, we report results that may explain this outcome, based on the finding that neutralizing antibodies to IL6 and STAT3 inhibition are sufficient to upregulate the EGFR pathway in high-grade serous and other ovarian cancer cells. Cell treatment with the EGFR inhibitor gefitinib abolished upregulation of the EGFR pathway. Combining neutralizing IL6 antibodies and gefitinib inhibited malignant cell growth in 2D and 3D culture. We found that ErbB-1 was localized predominantly in the nucleus of ovarian cancer cells examined, contrasting with plasma membrane localization in lung cancer cells. Treatment with anti-IL6, gefitinib, or their combination all led to partial restoration of ErbB-1 on the plasma membrane. In vivo experiments confirmed the effects of IL6 inhibition on the EGFR pathway and the enhanced activity of a combination of anti-IL6 antibodies and gefitinib on malignant cell growth. Taken together, our results offer a preclinical rationale to combine anti-IL6 and gefitinib to treat patients with advanced stage ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/metabolism , Ovarian Neoplasms/drug therapy , Animals , Antibodies, Neutralizing/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Nucleus , Drug Synergism , ErbB Receptors/genetics , Female , Gefitinib , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Interleukin-6/physiology , MAP Kinase Signaling System , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/metabolism , Protein Transport , Quinazolines/administration & dosage , STAT3 Transcription Factor/metabolism , Tumor Burden , Up-Regulation , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 111(34): E3562-70, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114209

ABSTRACT

Cancer-associated inflammation mobilizes a variety of leukocyte populations that can inhibit or enhance tumor cell growth in situ. These subsets include γδ T cells, which can infiltrate tumors and typically provide large amounts of antitumor cytokines, such as IFN-γ. By contrast, we report here that in a well-established transplantable (ID8 cell line) model of peritoneal/ovarian cancer, γδ T cells promote tumor cell growth. γδ T cells accumulated in the peritoneal cavity in response to tumor challenge and could be visualized within solid tumor foci. Functional characterization of tumor-associated γδ T cells revealed preferential production of interleukin-17A (IL-17), rather than IFN-γ. Consistent with this finding, both T cell receptor (TCR)δ-deficient and IL-17-deficient mice displayed reduced ID8 tumor growth compared with wild-type animals. IL-17 production by γδ T cells in the tumor environment was essentially restricted to a highly proliferative CD27((-)) subset that expressed Vγ6 instead of the more common Vγ1 and Vγ4 TCR chains. The preferential expansion of IL-17-secreting CD27((-)) Vγ6((+)) γδ T cells associated with the selective mobilization of unconventional small peritoneal macrophages (SPMs) that, in comparison with large peritoneal macrophages, were enriched for IL-17 receptor A, and for protumor and proangiogenic molecular mediators, which were up-regulated by IL-17. Importantly, SPMs were uniquely and directly capable of promoting ovarian cancer cell proliferation. Collectively, this work identifies an IL-17-dependent lymphoid/myeloid cross-talk involving γδ T cells and SPMs that promotes tumor cell growth and thus counteracts cancer immunosurveillance.


Subject(s)
Interleukin-17/biosynthesis , Macrophages, Peritoneal/immunology , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Inflammation Mediators/metabolism , Lymphocytes, Tumor-Infiltrating/classification , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Ovarian Neoplasms/pathology , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Interleukin-17/metabolism , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
4.
Cancer Res ; 72(1): 66-75, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22065722

ABSTRACT

Constitutive production of inflammatory cytokines is a characteristic of many human malignant cell lines; however, the in vitro and in vivo interdependence of these cytokines, and their significance to the human cancer microenvironment, are both poorly understood. Here, we describe for the first time how three key cytokine/chemokine mediators of cancer-related inflammation, TNF, CXCL12, and interleukin 6, are involved in an autocrine cytokine network, the "TNF network," in human ovarian cancer. We show that this network has paracrine actions on angiogenesis, infiltration of myeloid cells, and NOTCH signaling in both murine xenografts and human ovarian tumor biopsies. Neutralizing antibodies or siRNA to individual members of this TNF network reduced angiogenesis, myeloid cell infiltration, and experimental peritoneal ovarian tumor growth. The dependency of network genes on TNF was shown by their downregulation in tumor cells from patients with advanced ovarian cancer following the infusion of anti-TNF antibodies. Together, the findings define a network of inflammatory cytokine interactions that are crucial to tumor growth and validate this network as a key therapeutic target in ovarian cancer.


Subject(s)
Cytokines/metabolism , Ovarian Neoplasms/metabolism , Animals , Biopsy , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Real-Time Polymerase Chain Reaction
5.
Proc Natl Acad Sci U S A ; 108(26): 10662-7, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670304

ABSTRACT

The inflammatory cytokine TNF-α has been recognized as a critical tumor promoter, but the effector cells that mediate its action have not been fully characterized. Because B cells regulate squamous and prostate carcinogenesis, and Tnf(-/-) mice harbor B-cell defects, we investigated the hypothesis that B cells are important effector cells for TNF-α-mediated promotion of cancer development. Using an adoptive transfer strategy and the 7,12-dimethylbenz[α]anthracene/terephthalic acid (DMBA/TPA) two-stage model of skin carcinogenesis, we found that both B cells and TNF-α are critical for the development of DMBA/TPA-induced papilloma. Transfer of B cells from DMBA/TPA-treated wild-type mice to Tnf(-/-) mice rescued papilloma development to a wild-type level, a result not observed when B cells from Tnf(-/-) mice were transferred to Rag2(-/-) mice or when TNF-α was eliminated selectively in B cells. Resistance to papilloma development in Tnf(-/-) mice was associated with increased IFN-γ and CD8(+) T cells in skin and a significant reduction in IL-10-producing B regulatory cells alongside an increase in IFN-γ-producing CD8(+) T cells in the spleen. These data indicate that during DMBA/TPA-induced squamous carcinogenesis TNF-α mediates tumor-promoting activity via regulatory B cells that repress antitumor immunity.


Subject(s)
B-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Skin Neoplasms/immunology , Tumor Necrosis Factor-alpha/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Adoptive Transfer , Animals , Carcinogens/toxicity , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Immunohistochemistry , Mice , Mice, Knockout , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tumor Necrosis Factor-alpha/genetics
6.
J Clin Invest ; 119(10): 3011-23, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19741298

ABSTRACT

Cytokines orchestrate the tumor-promoting interplay between malignant cells and the immune system. In many experimental and human cancers, the cytokine TNF-alpha is an important component of this interplay, but its effects are pleiotropic and therefore remain to be completely defined. Using a mouse model of ovarian cancer in which either TNF receptor 1 (TNFR1) signaling was manipulated in different leukocyte populations or TNF-alpha was neutralized by antibody treatment, we found that this inflammatory cytokine maintained TNFR1-dependent IL-17 production by CD4+ cells and that this led to myeloid cell recruitment into the tumor microenvironment and enhanced tumor growth. Consistent with this, in patients with advanced cancer, treatment with the TNF-alpha-specific antibody infliximab substantially reduced plasma IL-17 levels. Furthermore, expression of IL-1R and IL-23R was downregulated in CD4+CD25- cells isolated from ascites of ovarian cancer patients treated with infliximab. We have also shown that genes ascribed to the Th17 pathway map closely with the TNF-alpha signaling pathway in ovarian cancer biopsy samples, showing particularly high levels of expression of genes encoding IL-23, components of the NF-kappaB system, TGF-beta1, and proteins involved in neutrophil activation. We conclude that chronic production of TNF-alpha in the tumor microenvironment increases myeloid cell recruitment in an IL-17-dependent manner that contributes to the tumor-promoting action of this proinflammatory cytokine.


Subject(s)
Interleukin-17/immunology , Ovarian Neoplasms/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Chimera/genetics , Chimera/immunology , Clinical Trials as Topic , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Infliximab , Interleukin-17/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics
7.
J Exp Med ; 205(6): 1261-8, 2008 Jun 09.
Article in English | MEDLINE | ID: mdl-18490490

ABSTRACT

The nuclear factor kappaB (NF-kappaB) signaling pathway is important in cancer-related inflammation and malignant progression. Here, we describe a new role for NF-kappaB in cancer in maintaining the immunosuppressive phenotype of tumor-associated macrophages (TAMs). We show that macrophages are polarized via interleukin (IL)-1R and MyD88 to an immunosuppressive "alternative" phenotype that requires IkappaB kinase beta-mediated NF-kappaB activation. When NF-kappaB signaling is inhibited specifically in TAMs, they become cytotoxic to tumor cells and switch to a "classically" activated phenotype; IL-12(high), major histocompatibility complex II(high), but IL-10(low) and arginase-1(low). Targeting NF-kappaB signaling in TAMs also promotes regression of advanced tumors in vivo by induction of macrophage tumoricidal activity and activation of antitumor activity through IL-12-dependent NK cell recruitment. We provide a rationale for manipulating the phenotype of the abundant macrophage population already located within the tumor microenvironment; the potential to "re-educate" the tumor-promoting macrophage population may prove an effective and novel therapeutic approach for cancer that complements existing therapies.


Subject(s)
Macrophages/physiology , NF-kappa B/physiology , Animals , Cell Differentiation , Humans , Immunosuppression Therapy , Macrophages/cytology , Macrophages/immunology , Macrophages/pathology , Mice , Neoplasms/pathology , Phenotype , Signal Transduction
8.
Cancer Immunol Immunother ; 57(2): 247-63, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17657488

ABSTRACT

Antibodies directed against tumor-associated antigens are emerging as effective treatments for a number of cancers, although the mechanism(s) of action for some are unclear and still under investigation. We have previously examined a chimeric IgE antibody (MOv18 IgE), against the ovarian tumor-specific antigen, folate binding protein (FBP), and showed that it can direct human PBMC to kill ovarian cancer cells. We have developed a three-color flow cytometric assay to investigate the mechanism by which IgE receptors on U937 monocytes target and kill ovarian tumor cells. U937 monocytes express three IgE receptors, the high-affinity receptor, FcepsilonRI, the low-affinity receptor, CD23, and galectin-3, and mediate tumor cell killing in vitro by two mechanisms, cytotoxicity, and phagocytosis. Our results suggest that CD23 mediates phagocytosis, which is enhanced by upregulation of CD23 on U937 cells with IL-4, whereas FcepsilonRI mediates cytotoxicity. We show that effector : tumor cell bridging is associated with both activities. Galectin-3 does not appear to be involved in tumor cell killing. U937 cells and IgE exerted ovarian tumor cell killing in vivo in our xenograft model in nude mice. Harnessing IgE receptors to target tumor cells suggests the potential of tumor-specific IgE antibodies to activate effector cells in immunotherapy of ovarian cancer.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/physiology , Immunotherapy/methods , Monocytes/immunology , Ovarian Neoplasms/therapy , Phagocytosis/physiology , Receptors, IgE/immunology , Animals , Cell Line, Tumor , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Nude
9.
J Immunol ; 179(5): 2832-43, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17709497

ABSTRACT

Abs have a paramount place in the treatment of certain, mainly lymphoid, malignancies, although tumors of nonhemopoietic origin have proved more refractory ones. We have previously shown that the efficacy of immunotherapy of solid tumors, in particular ovarian carcinoma, may be improved by the use of IgE Abs in place of the conventional IgG. An IgE Ab (MOv18 IgE) against an ovarian-tumor-specific Ag (folate binding protein), in combination with human PBMC, introduced into ovarian cancer xenograft-bearing mice, greatly exceeded the analogous IgG1 in promoting survival. In this study, we analyzed the mechanisms by which MOv18 IgE may exert its antitumor activities. Monocytes were essential IgE receptor-expressing effector cells that mediated the enhanced survival of tumor-bearing mice by MOv18 IgE and human PBMC. Monocytes mediated MOv18 IgE-dependent ovarian tumor cell killing in vitro by two distinct pathways, cytotoxicity and phagocytosis, acting respectively through the IgE receptors FcepsilonRI and CD23. We also show that human eosinophils were potent effector cells in MOv18 IgE Ab-dependent ovarian tumor cell cytotoxicity in vitro. These results demonstrate that IgE Abs can engage cell surface IgE receptors and activate effector cells against ovarian tumor cells. Our findings offer a framework for an improved immunotherapeutic strategy for combating solid tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Carcinoma/drug therapy , Immunoglobulin E/therapeutic use , Ovarian Neoplasms/drug therapy , Phagocytosis , Animals , Antibodies, Monoclonal, Murine-Derived , Carcinoma/immunology , Carcinoma/therapy , Cell Line, Tumor , Female , Humans , Immunotherapy , Mice , Monocytes/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptors, IgE/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 6(7): 1993-2002, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620429

ABSTRACT

In view of our previous findings that tumor cell-derived macrophage migration inhibitory factor (MIF) increased macrophage-mediated ovarian cancer cell invasiveness in vitro, we investigated the wider significance of ovarian cancer cell-derived MIF for tumor growth, metastasis, and angiogenesis. We found that MIF is expressed in borderline and malignant ovarian tumors, and active MIF is found in malignant ascitic fluid. We next investigated the expression and function of MIF in a syngeneic ovarian cancer model. Stable knockdown of MIF in the murine ovarian cancer cell line ID8 decreased in vivo tumor burden and overall survival. Tumors arising from MIF knockdown cells had decreased proliferation and significantly increased apoptosis. This was associated with an increased phosphorylation of p53 and reduced Akt phosphorylation. MIF knockdown led to a changed cytokine profile in the ascitic microenvironment; tumor necrosis factor-alpha, interleukin-6 (IL-6), and IL-10 expression were all significantly decreased. Accompanying this decrease in cytokine expression was a significant decrease in macrophage infiltration into ascites. Additionally, MIF knockdown reduced the expression of proangiogenic cytokines vascular endothelial growth factor and keratinocyte chemoattractant (KC) and reduced the amount of endothelial cells in the malignant ascites. We conclude that autocrine production of MIF by ovarian cancer cells stimulates other cytokines, chemokines, and angiogenic factors that may promote colonization of the peritoneum and neovascularization of tumor deposits.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Neovascularization, Pathologic , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Animals , Ascites/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endothelial Cells/pathology , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Oncogene ; 23(41): 6954-66, 2004 Sep 09.
Article in English | MEDLINE | ID: mdl-15273742

ABSTRACT

Mice deficient in TNF-alpha (TNF-alpha(-/-) mice) are resistant to skin carcinogenesis and expression of MMP-9 is inhibited in TNF-alpha(-/-) mice during skin tumour development. In the early stages of tumour promotion, MMP-9 protein initially localized to the follicular epidermis but subsequently began to accumulate in the interfollicular epidermis of wild-type but not TNF-alpha(-/-) mice. Inhibition of TNF-alpha or MMP-9 function reduced keratinocyte migration in vitro. In addition, a deficiency of TNF-alpha delayed re-epithelialization in vivo and this correlated with reduced MMP-9 expression. Collectively, these data suggest that MMP-9 regulates keratinocyte migration in a TNF-alpha-dependent manner. Expression profiling of genes that control cell adhesion and migration revealed markedly lower levels of the integrin subunits alphav and beta6 in TNF-alpha(-/-) compared with wild-type keratinocytes in vitro. alphavbeta6 expression was upregulated by keratinocytes in vitro and during tumour promotion in vivo in a TNF-alpha-dependent manner. Furthermore, alphavbeta6 blockade significantly inhibited keratinocyte migration and TNF-alpha-stimulated MMP-9 expression in vitro. These data illustrate a novel TNF-alpha-dependent mechanism for the control of alphavbeta6 expression and suggest one pathway for TNF-alpha regulation of MMP-9. Increased MMP-9 and alphavbeta6 expression may stimulate epithelial cell migration during tumour formation and may be one mechanism whereby TNF-alpha acts as an endogenous tumour promoter.


Subject(s)
Antigens, Neoplasm/genetics , Epidermis/metabolism , Gene Expression Regulation, Neoplastic , Integrins/genetics , Keratinocytes/cytology , Matrix Metalloproteinase 9/genetics , Skin Neoplasms/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Movement , Epithelium/metabolism , Mice , RNA, Messenger/analysis , Tissue Inhibitor of Metalloproteinase-3/genetics , Wound Healing
12.
Oncogene ; 23(10): 1902-10, 2004 Mar 11.
Article in English | MEDLINE | ID: mdl-14661063

ABSTRACT

Keratinocyte-derived TNF-alpha acts as an endogenous tumour promoter and can also regulate AP-1 activity in mouse epidermis. To gain further insight into TNF-alpha signalling during skin tumour formation, mice deficient in TNFR1 (TNFR1-/- mice) or TNFR2 (TNFR2-/- mice) were subjected to chemical carcinogenesis. Tumour multiplicity was significantly reduced in TNFR1-/- and TNFR2-/- mice compared to wild-type (wt) mice, suggesting that both receptors have protumour activity. However, TNFR1-/- mice were markedly more resistant to tumour development than TNFR2-/- mice indicating that TNFR1 is the major mediator of TNF-alpha-induced tumour formation. TNFR1 and TNFR2 were both expressed in wt epidermis during tumour promotion and by primary keratinocytes in vitro. TPA-induced c-Jun expression was transient in TNFR1-/- and TNFR2-/- compared to wt epidermis and this was reflected by reduced induction of the AP-1-responsive genes granulocyte/macrophage-colony stimulating factor, matrix metalloproteinase-9 and matrix metalloproteinase-3. These genes were differentially regulated in TNFR1-/- compared to TNFR2-/- epidermis, suggesting that the TNF-alpha receptors act independently via different AP-1 complexes to transduce TNF-alpha signals during tumour promotion. In addition, TNFR2 cooperated with TNFR1 to optimise TNFR1-mediated TNF-alpha bioactivity on keratinocytes in vitro. Our data provide further insight into TNF-alpha signalling in malignancy and provide some rationale for the use of TNF-alpha antagonists in the treatment of cancer.


Subject(s)
Antigens, CD/genetics , Receptors, Tumor Necrosis Factor/genetics , Skin Neoplasms/genetics , Animals , Animals, Newborn , Antigens, CD/physiology , Cells, Cultured , Epidermis/drug effects , Epidermis/pathology , Gene Expression Regulation/drug effects , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Skin/pathology , Tetradecanoylphorbol Acetate/pharmacology
13.
Cancer Res ; 63(23): 8360-5, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678997

ABSTRACT

The leukocyte infiltrate of human and murine epithelial cancers is regulated by chemokine production in the tumor microenvironment. In this article, we tested the hypothesis that chemokine receptor antagonists may have anticancer activity by inhibiting this infiltrate. We first characterized CC chemokines, chemokine receptors, and the leukocyte infiltrate in the 410.4 murine model of breast cancer. We found that CCL5 (RANTES) was produced by the tumor cells, and its receptors, CCR1 and CCR5, were expressed by the leukocyte infiltrate. As Met-CCL5 is an antagonist of CCR1 and CCR5 with activity in models of inflammatory disease, we tested its activity against 410.4 tumors. After 5 weeks of daily treatment with Met-CCL5, the volume and weight of 410.4 tumors was significantly decreased compared with control-treated tumors. Met-CCL5 was also active against established tumors. The total cell number obtained after collagenase digestion was decreased in Met-CCL5-treated tumors as was the proportion of infiltrating macrophages. Furthermore, chemokine antagonist treatment increased stromal development and necrosis. Our results provide direct evidence that macrophages contribute to tumor development and are the first indication that chemokine receptor antagonists may provide novel strategies in cancer prevention and treatment.


Subject(s)
CCR5 Receptor Antagonists , Mammary Neoplasms, Experimental/pathology , Receptors, Chemokine/antagonists & inhibitors , Animals , Cell Division/drug effects , Chemokine CCL5/biosynthesis , Female , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/drug effects , Macrophages/immunology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, CCR1 , Receptors, CCR5/biosynthesis , Receptors, CCR5/genetics , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics
14.
Mol Cancer Ther ; 2(5): 445-51, 2003 May.
Article in English | MEDLINE | ID: mdl-12748306

ABSTRACT

The proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) was originally considered to have activity against malignant disease. However, recent studies suggest TNF-alpha may also act as an endogenous tumor promoter. In the present work, mice deficient in TNF-alpha either genetically (TNF-alpha(-/-)) or after blockade with a neutralizing antibody (cV1q) were used to investigate the role of TNF-alpha in skin tumor development. Papillomas were induced in wild-type (wt) mice after treatment of skin with the initiating agent 9,10-dimethyl-1,2-benzanthracene followed by promotion with 12-O-tetradecanoylphorbol-13-acetate (TPA) for 15 weeks. TNF-alpha(-/-) mice were resistant to papilloma development when compared with wt mice on C57Bl/6J, 129/SvEv, and BALB/c genetic backgrounds. Primary murine keratinocytes (newborn keratinocytes) and skin homogenates were used to characterize TPA-stimulated TNF-alpha expression. TPA induced TNF-alpha protein in newborn keratinocytes in vitro and epidermis in vivo. Neutralization of TNF-alpha protein with cV1q in vivo for 0-15 weeks of promotion significantly decreased skin tumor development after 9,10-dimethyl-1,2-benzanthracene/TPA treatment. cV1q treatment during the early stages of tumor promotion (0-6 weeks) was equally effective. These data suggest that early induction of TNF-alpha is critical for skin tumor promotion. cV1q also reduced TPA-stimulated expression of matrix metalloproteinase 9 and granulocyte macrophage colony-stimulating factor, proteins that are differentially regulated in wt and TNF-alpha(-/-) epidermis. Treatment of the 410.4 transplantable breast carcinoma with cV1q reduced tumor growth in vivo, illustrating that inhibition of tumor growth through neutralization of TNF-alpha is not limited to skin carcinogenesis. These results provide further evidence for procancer actions of TNF-alpha and give some rationale for use of TNF-alpha antagonists in cancer prevention and treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Papilloma/drug therapy , Skin Neoplasms/drug therapy , Transforming Growth Factor alpha/immunology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Keratinocytes/drug effects , Keratinocytes/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Papilloma/chemically induced , Papilloma/metabolism , Rats , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate/toxicity
15.
Eur J Immunol ; 33(4): 1030-40, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12672069

ABSTRACT

We have previously shown that a chimeric IgE antibody against the folic acid receptor (MOv18 IgE) inhibits tumor growth in a SCID mouse model of ovarian carcinoma. MOv18 IgE gave greater protection than the corresponding chimeric MOv18 IgG1. We have now confirmed these effects in a nude-mouse model of ovarian carcinoma and have demonstrated for the first time that human monocytes are active in IgE antibody-dependent cell-mediated cytotoxicity. Injection of tumor-bearing mice with PBMC and MOv18 IgE led to infiltration of monocytes into the tumors and prolonged survival of the mice. Incubation of PBMC or purified monocytes and MOv18 IgE with ovarian tumor cells in vitro resulted in tumor cell killing proportional to the expression of unoccupied FcepsilonRI on monocytes.We observed phagocytosis of tumor cells by the monocytes in vitro. Our results suggest that tumor-specific IgE antibodies may be exploited for immunotherapy of cancer.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Carcinoma/immunology , Immunoglobulin E/immunology , Monocytes/immunology , Ovarian Neoplasms/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Basophils/immunology , Carcinoma/pathology , Cell Movement , Female , Humans , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunologic Surveillance , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Receptors, IgE/metabolism , Receptors, IgG/metabolism , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...