Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Int J Mol Sci ; 22(18)2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34575853

ABSTRACT

Furan is a volatile compound that is formed in foods during thermal processing. It is classified as a possible human carcinogen by international authorities based on sufficient evidence of carcinogenicity from studies in experimental animals. Although a vast number of studies both in vitro and in vivo have been performed to investigate furan genotoxicity, the results are inconsistent, and its carcinogenic mode of action remains to be clarified. Here, we address the mutagenic and clastogenic activity of furan and its prime reactive metabolite cis-2 butene-1,4-dial (BDA) in mammalian cells in culture and in mouse animal models in a search for DNA lesions responsible of these effects. To this aim, Fanconi anemia-derived human cell lines defective in the repair of DNA inter-strand crosslinks (ICLs) and Ogg1-/- mice defective in the removal of 8-hydroxyguanine from DNA, were used. We show that both furan and BDA present a weak (if any) mutagenic activity but are clear inducers of clastogenic damage. ICLs are strongly indicated as key lesions for chromosomal damage whereas oxidized base lesions are unlikely to play a critical role.


Subject(s)
Chromosome Aberrations/chemically induced , Furans/adverse effects , Mutation/drug effects , Oxidative Stress/drug effects , Animals , Carcinogens , Cell Line , DNA Damage/drug effects , Dose-Response Relationship, Drug , Furans/toxicity , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Micronuclei, Chromosome-Defective/chemically induced , Mutagens , Oxidation-Reduction
2.
J Immunol ; 192(11): 5434-41, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24790144

ABSTRACT

Perforin (pfp)-mediated cytotoxicity is one of the principal immunosurveillance mechanisms involved in the fight against cancer. However, its importance in spontaneous epithelial cancer is still poorly defined. In this study, we use a realistic mouse model that displays many features that are equivalent to human pathology to evaluate the role of pfp-dependent immunosurveillance by comparing tumor progression in rat ERBB-2 (neu) transgenic, pfp-proficient (neu(+)/pfp(+)) or pfp-deficient (neu(+)/pfp(-)) BALB/c male mice. Adult neu(+)/pfp(+) males developed poorly differentiated salivary carcinomas, whereas neu(+)/pfp(-) males displayed their salivary carcinomas noticeably earlier and showed zones of more highly differentiated tumor, indicating that pfp-mediated immunosurveillance is able not only to delay the growth kinetic of an aggressive epithelial tumor, but also to shape its histology. The role of pfp-mediated immunosurveillance appeared to be of even more dramatic importance against the less aggressive male mammary carcinomas. In neu(+)/pfp(+) males, the incidence of mammary carcinomas was a sporadic and late event. In contrast, in neu(+)/pfp(-) males their incidence was four-fold higher. This higher cancer incidence was associated with a 2-fold higher occurrence of persisting mammary remnants, a major risk factor for mammary cancer in male mice, and one that would appear to be due to pfp's previously unidentified involvement in male mammary gland rejection during embryogenesis. This work thus provides further proof of the complex role that the immune system plays in the body and gives new insight into the pathogenesis of epithelial tumors, demonstrating that the penetrance and malignancy of a tumor may be dramatically affected by pfp-dependent mechanisms.


Subject(s)
Cell Transformation, Neoplastic/immunology , Neoplasms, Experimental/immunology , Pore Forming Cytotoxic Proteins/immunology , Receptor, ErbB-2/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Pore Forming Cytotoxic Proteins/genetics , Rats , Receptor, ErbB-2/genetics
3.
Cancer Cell ; 13(1): 58-68, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18167340

ABSTRACT

It is widely accepted that metastasis is a late event in cancer progression. Here, however, we show that tumor cells can disseminate systemically from earliest epithelial alterations in HER-2 and PyMT transgenic mice and from ductal carcinoma in situ in women. Wild-type mice transplanted with single premalignant HER-2 transgenic glands displayed disseminated tumor cells and micrometastasis in bone marrow and lungs. The number of disseminated cancer cells and their karyotypic abnormalities were similar for small and large tumors in patients and mouse models. When activated by bone marrow transplantation into wild-type recipients, 80 early-disseminated cancer cells sufficed to induce lethal carcinosis. Therefore, release from dormancy of early-disseminated cancer cells may frequently account for metachronous metastasis.


Subject(s)
Breast Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Precancerous Conditions/pathology , Animals , Bone Marrow Cells/pathology , Breast Neoplasms/genetics , Cell Proliferation , Cell Transformation, Neoplastic , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Karyotyping , Lung Neoplasms/secondary , Mammary Glands, Animal/transplantation , Mammary Glands, Animal/ultrastructure , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Precancerous Conditions/genetics , Siblings
4.
J Exp Med ; 203(11): 2441-50, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17043144

ABSTRACT

Inflammatory cells can either promote or inhibit tumor growth. Here we studied whether CD40, a key molecule for adaptive immune response, has any role in mammary carcinogenesis of BALB/NeuT transgenic tumor-prone mice. We transferred the HER2/neu oncogene into CD40-null background to obtain the CD40-KO/NeuT strain. CD40-KO/NeuT mice showed delayed tumor onset and reduced tumor multiplicity. BM (BM) transplantation experiments excluded a role of BM-derived cells in the reduced tumorigenicity associated with CD40 deficiency. Rather, CD40 expressed by endothelial cells (ECs) takes part to the angiogenic process. Accordingly, large vessels, well organized around the tumor lobular structures, characterize BALB/NeuT tumors, whereas tiny numerous vessels with scarce extracellular matrix are dispersed in the parenchyma of poorly organized CD40-KO/NeuT tumors. Activated platelets, which may interact with and activate ECs, are a possible source of CD40L. Their localization within tumor vessels prompted the idea of treating BALB/NeuT and CD40-KO/NeuT mice chronically with the anti-platelet drug clopidogrel, known to inhibit platelet CD40L expression. Treatment of BALB/NeuT mice reduced tumor growth to a level similar to CD40-deficient mice, whereas CD40-KO/NeuT mice treated or not showed the same attenuated tumor outgrowth, indicating that activated platelets are the likely source of CD40L in this model. Collectively, these data point to a participation of CD40/CD40L in the angiogenic processes associated with mammary carcinogenesis of BALB/NeuT mice.


Subject(s)
CD40 Antigens/physiology , Cell Proliferation , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Animals , CD40 Antigens/genetics , Endothelium, Vascular/pathology , Female , Mammary Neoplasms, Experimental/blood supply , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Rats
5.
Blood ; 115(7): 1374-84, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-19903903

ABSTRACT

Telomerase reverse transcriptase (TERT) is a good candidate for cancer immunotherapy because it is overexpressed in 85% of all human tumors and implicated in maintenance of the transformed phenotype. TERT-based cancer vaccines have been shown to be safe, not inducing any immune-related pathology, but their impact on tumor progression is modest. Here we show that adoptive cell therapy with the use of high-avidity T lymphocytes reactive against telomerase can control the growth of different established tumors. Moreover, in transgenic adenocarcinoma mouse prostate mice, which develop prostate cancer, TERT-based adoptive cell therapy halted the progression to more aggressive and poorly differentiated tumors, significantly prolonging mouse survival. We also demonstrated that human tumors, including Burkitt lymphoma, and human cancer stem cells, are targeted in vivo by TERT-specific cytotoxic T lymphocytes. Effective therapy with T cells against telomerase, different from active vaccination, however, led to autoimmunity marked by a consistent, although transient, B-cell depletion in primary and secondary lymphoid organs, associated with alteration of the spleen cytoarchitecture. These results indicate B cells as an in vivo target of TERT-specific cytotoxic T lymphocytes during successful immunotherapy.


Subject(s)
Adenocarcinoma/therapy , Adoptive Transfer/methods , Lymphopenia/pathology , Prostatic Neoplasms/therapy , T-Lymphocytes/transplantation , Telomerase/immunology , Adenocarcinoma/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow Cells/pathology , Cancer Vaccines , Cell Line, Tumor , Colonic Neoplasms , Disease Models, Animal , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Lung Neoplasms , Male , Melanoma , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Prostatic Neoplasms/immunology , Skin Neoplasms , Spleen/pathology , T-Lymphocytes/immunology
6.
J Immunol ; 184(11): 6124-32, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20435927

ABSTRACT

Rat (r)Erbb2 transgenic BALB-neuT mice genetically predestined to develop multiple invasive carcinomas allow an assessment of the potential of a vaccine against the stages of cancer progression. Because of rErbb2 expression in the thymus and its overexpression in the mammary gland, CD8(+) T cell clones reacting at high avidity with dominant rErbb2 epitopes are deleted in these mice. In BALB-neuT mice with diffuse and invasive in situ lesions and almost palpable carcinomas, a temporary regulatory T cells depletion combined with anti-rErbb2 vaccine markedly enhanced the anti-rErbb2 Ab response and allowed the expansion of latent pools of low-avidity CD8(+) T cells bearing TCRs repertoire reacting with the rErbb2 dominant peptide. This combination of a higher Ab response and activation of a low-avidity cytotoxic response persistently blocked tumor progression at stages in which the vaccine alone was ineffective. However, when diffuse and invasive microscopic cancers become almost palpable, this combination was no longer able to secure a significant extension of mice survival.


Subject(s)
Cancer Vaccines/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Vaccines, DNA/immunology , Animals , Antibodies/immunology , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Cell Separation , Electroporation , Female , Flow Cytometry , Genes, erbB-2/genetics , Genes, erbB-2/immunology , Immunohistochemistry , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/drug effects , Vaccines, DNA/pharmacology
7.
J Cell Mol Med ; 14(12): 2803-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19818098

ABSTRACT

It is unknown whether zoledronic acid (ZA) at clinically relevant doses is active against tumours not located in bone. Mice transgenic for the activated ErbB-2 oncogene were treated with a cumulative number of doses equivalent to that recommended in human beings. A significant increase in tumour-free and overall survival was observed in mice treated with ZA. At clinically compatible concentrations, ZA modulated the mevalonate pathway and affected protein prenylation in both tumour cells and macrophages. A marked reduction in the number of tumour-associated macrophages was paralleled by a significant decrease in tumour vascularization. The local production of vascular endothelial growth factor and interleukin-10 was drastically down-regulated in favour of interferon-γ production. Peritoneal macrophages and tumour-associated macrophages of ZA-treated mice recovered a full M1 antitumoral phenotype, as shown by nuclear translocation of nuclear factor kB, inducible nitric oxide synthase expression and nitric oxide production. These data indicate that clinically achievable doses of ZA inhibit spontaneous mammary cancerogenesis by targeting the local microenvironment, as shown by a decreased tumour vascularization, a reduced number of tumour-associated macrophages and their reverted polarization from M2 to M1 phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Diphosphonates/pharmacology , Imidazoles/pharmacology , Macrophages/drug effects , Mammary Neoplasms, Animal/drug therapy , Mevalonic Acid/metabolism , Animals , Antineoplastic Agents/administration & dosage , Diphosphonates/administration & dosage , Female , Genes, erbB-2 , Imidazoles/administration & dosage , Interferon-gamma/metabolism , Interleukin-10/metabolism , Macrophages/immunology , Macrophages/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Mice, Transgenic , NF-kappa B/metabolism , Neovascularization, Pathologic , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Protein Prenylation , Vascular Endothelial Growth Factor A/metabolism , Zoledronic Acid
8.
Mol Carcinog ; 49(2): 114-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20027636

ABSTRACT

The oncogenic transcription factor Stat3 is constitutively active in a high percentage of human tumors including mammary adenocarcinomas and is reported to participate in the ErbB-2 oncogene signaling. In order to assess the role of signal transducer and activator of transcription 3 (Stat3) in mammary tumorigenesis downstream of ErbB-2, we generated mice expressing the activated rat ErbB-2 (neu) but lacking Stat3 in the mammary epithelium. Stat3 is apparently not required for neu-driven mammary tumorigenesis as tumors developed similarly in both Stat3-sufficient and Stat3-deficient glands. However, short hairpin RNA (shRNA)-mediated Stat3 silencing in a neu-overexpressing tumor-derived cell line completely abolished both neu-driven anchorage-independent growth and lung metastasis. Our data suggest that Stat3 might be a useful therapeutic target in breast tumors showing amplification and/or overexpression of the ErbB-2 oncogene, which normally display aggressive, metastatic behavior.


Subject(s)
Cell Adhesion/physiology , Cell Division/physiology , Genes, erbB-2 , Mammary Neoplasms, Experimental/physiopathology , Neoplasm Metastasis/physiopathology , STAT3 Transcription Factor/physiology , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Female , Gene Silencing , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , STAT3 Transcription Factor/genetics
9.
Clin Cancer Res ; 15(9): 2979-87, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366834

ABSTRACT

PURPOSE: The human prostate is endowed with intraepithelial and stromal lymphocytes, which may develop lymphoid follicles (LF) and allow a local immune response. We sought to investigate whether interleukin (IL)-7 and BAFF/BLyS, two fundamental survival factors for T and B cells, are expressed in the normal and neoplastic prostate and affect intraprostatic lymphocyte homeostasis. EXPERIMENTAL DESIGN: We have used real-time reverse transcription-PCR of microdissected prostatic glands and confocal microscopy to detect cytokine production, combined with immunohistochemistry to characterize intraprostatic lymphocytes. RESULTS: Prostatic epithelia constitutively produce IL-7 and, to a lesser extent, BAFF/BLyS. Indeed, we show that IL-7 receptor alpha is expressed by intraepithelial T lymphocytes and parafollicular T cells, whereas BAFF-R is found on periglandular B lymphocytes and mantle zone B cells of LFs. Prostate-homing B and T lymphocytes are scarcely proliferating, whereas most of them express the antiapoptotic protein bcl-2 and reveal a low apoptotic index in the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. The transition from normal to neoplastic glands in prostate cancer (PCa) is marked by a dramatic decline of IL-7 and BAFF/BLyS production. Accordingly, PCa is characterized by a significant reduction of intraepithelial lymphocytes and loss of LFs. B-cell and T-cell expression of bcl-2 decrease, whereas the apoptotic events increase. The remaining PCa-infiltrating lymphocytes are mostly CD8(+) T cells that lack terminal differentiation and barely penetrate neoplastic glands. CONCLUSIONS: These results suggest that epithelial IL-7 and BAFF/BLyS production support intraprostatic lymphocyte survival. Its loss in PCa is associated with a severe depletion of prostate-associated lymphocytes and points to a novel tumor escape mechanism.


Subject(s)
B-Cell Activating Factor/genetics , Gene Expression Regulation, Neoplastic/physiology , Interleukin-7/genetics , Monitoring, Immunologic , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Tumor Escape , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Blotting, Western , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Immunoenzyme Techniques , Interleukin-7 Receptor alpha Subunit/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Prostate/immunology , Prostate/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
10.
Cancer Res ; 67(11): 5454-60, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545627

ABSTRACT

The concept of tumor immune surveillance has been supported by several recent studies in mice which show that immune effector mechanisms suppress hematologic malignancy. However, because the most common forms of human cancer are epithelial in origin, and comparatively very little data supports the immune surveillance of epithelial malignancies, we have chosen to evaluate the role of perforin-mediated cytotoxicity in the prevention of BALB/c Her2/neu-induced mammary cancer. Interestingly, perforin significantly delayed the onset of mammary tumorigenesis and reduced the number of mammary tumors without improving survival. Natural killer cell, but not CD8+ T cell, depletion resulted in a similar phenotype to perforin deficiency in this regard. Histologic analysis further indicated that the effect of perforin was most evident during the earliest stages of carcinogenesis rather than prior to or during the hyperplastic phase. This data suggests that perforin may mediate some suppression of epithelial carcinogenesis by intervening early in the tumor development process.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Membrane Glycoproteins/immunology , Oncogenes , Pore Forming Cytotoxic Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Female , Killer Cells, Natural/immunology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Perforin , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology
11.
Cancer Res ; 66(15): 7734-40, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885376

ABSTRACT

To assess the role of CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells in overcoming immunosurveillance of Erbb2 (HER-2/neu) mammary lesions, we studied the effects of their sustained removal in BALB/c female mice made transgenic for the rat Erbb2 (r-Erbb2) oncogene (BALB-neuT mice), which develop multiple mammary carcinomas. During the progression of these lesions, Treg cells expand in the spleen, tumor draining lymph nodes, and tumors. Repeated administration of anti-CD25 antibodies extends tumor-free survival, reduces carcinoma multiplicity, and leads to the manifestation of a natural antibody and CTL-mediated reactivity against r-Erbb2. Loss of Foxp3(+) Treg cells during anti-CD25 treatment remarkably caused the disappearance of Gr1(+) immature myeloid cells, suggesting a cross-talk between these two inhibitory immune cell types. Treg cell expansion associated with r-Erbb2 overexpression may be seen as a physiologic response to dampen the immune reaction elicited by local anomalous overexpression of a self-antigen.


Subject(s)
Cell Transformation, Neoplastic/immunology , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Transformation, Neoplastic/genetics , Female , Immunoglobulin G/immunology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , NIH 3T3 Cells , Rats , Receptor, ErbB-2/genetics , Receptors, Interleukin-2/immunology , Self Tolerance/immunology
12.
J Clin Invest ; 111(8): 1161-70, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12697735

ABSTRACT

Since the mechanisms by which specific immunity destroys Her-2/neu carcinoma cells are highly undetermined, these were assessed in BALB/c mice vaccinated with plasmids encoding extracellular and transmembrane domains of the protein product (p185(neu)) of the rat Her-2/neu oncogene shot into the skin by gene gun. Vaccinated mice rejected a lethal challenge of TUBO carcinoma cells expressing p185(neu). Depletion of CD4 T cells during immunization abolished the protection, while depletion of CD8 cells during the effector phase halved it, and depletion of polymorphonuclear granulocytes abolished all protection. By contrast, Ig mu-chain gene KO mice, as well as Fcgamma receptor I/III, beta-2 microglobulin, CD1, monocyte chemoattractant protein 1 (MCP1), IFN-gamma, and perforin gene KO mice were protected. Only mice with both IFN-gamma and perforin gene KOs were not protected. Although immunization also cured all BALB/c mice bearing established TUBO carcinomas, it did not cure any of the perforin KO or perforin and IFN-gamma KO mice. Few mice were cured that had knockouts of the gene for Ig mu-chain, Fcgamma receptor I/III, IFN-gamma, or beta-2 microglobulin. Moreover, vaccination cured half of the CD1 and the majority of the MCP1 KO mice. The eradication of established p185(neu) carcinomas involves distinct mechanisms, each endowed with a different curative potential.


Subject(s)
Antibodies, Neoplasm/physiology , Cancer Vaccines/immunology , Cytokines/physiology , Membrane Glycoproteins/physiology , Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Vaccines, DNA/immunology , Animals , Biolistics , Female , Graft Rejection , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Vaccination
13.
J Clin Invest ; 113(5): 709-17, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14991069

ABSTRACT

While much experimental data shows that vaccination efficiently inhibits a subsequent challenge by a transplantable tumor, its ability to inhibit the progress of autochthonous preneoplastic lesions is virtually unknown. In this article, we show that a combined DNA and cell vaccine persistently inhibits such lesions in a murine HER-2/neu mammary carcinogenesis model. At 10 weeks of age, all of the ten mammary gland samples from HER-2/neu-transgenic mice displayed foci of hyperplasia that progressed to invasive tumors. Vaccination with plasmids coding for the transmembrane and extracellular domain of rat p185neu followed by a boost with rp185neu+ allogeneic cells secreting IFN-gamma kept 48% of mice tumor free. At 22 weeks, their mammary glands were indistinguishable from those of 10-week-old untreated mice. Furthermore, the transcription patterns of the two sets of glands coincided. Of the 12,000 genes analyzed, 17 were differentially expressed and related to the antibody response. The use of B cell knockout mice as well as the concordance of morphologic and gene expression data demonstrated that the Ab response is the main mechanism facilitating tumor growth arrest. This finding suggests that a new way can be found to secure the immunologic control of the progression of HER-2/neu preneoplastic lesions.


Subject(s)
Cancer Vaccines , Neoplasms/prevention & control , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Animals , B-Lymphocytes/metabolism , Cell Membrane/metabolism , Cluster Analysis , Female , Gene Expression Regulation , Hyperplasia , Immunohistochemistry , Interferon-gamma/metabolism , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Models, Biological , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Plasmids/metabolism , Precancerous Conditions , Protein Structure, Tertiary , Rats , Time Factors
14.
Cancer Res ; 65(3): 1071-8, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705909

ABSTRACT

Plasmid DNA vectors encoding the full-length (VR1012/HER-2-FL) or only the extracellular and transmembrane domains (VR1012/HER-2-ECD-TM) of human (h) HER-2/neu proto-oncogene were used to vaccinate HER-2/neu transgenic mice (N202) engineered to overexpress the rat (r) neu proto-oncogene product (r-p185(neu)). Both the full-length and the deleted vaccines were significantly (P = 0.0001 and P = 0.06, respectively) more active than the empty vector (VR1012/EV) in preventing and delaying HER-2/neu-driven mammary carcinogenesis. A low-level intratumoral infiltrate of dendritic cells, macrophages, CD8 T cells and polymorphonuclear granulocytes in association with low-level cytokine production was observed, which was not detected in tumors from control mice. Morphologic analyses showed that vaccination with VR1012/HER-2-FL or ECD-TM also efficiently hampered the development of terminal ductal lobular units (TDLU). Analyses of sera from vaccinated mice revealed high titers of antihuman HER-2/neu antibodies, which correlated with the delayed time of tumor onset (P = 0.002). These antibodies did not cross-react with r-p185(neu). Nontransgenic mice treated with the vaccines produced autoreactive antibodies targeting mouse (m)-p185(neu) and showed impaired function of the lactating mammary gland and accelerated involution of the gland after weaning. Together, these data indicate that xenogeneic DNA immunization breaks tolerance against the endogenous m-p185(neu), impairing the development of mammary TDLU in which m-p185(neu) expression is concentrated. The reduction in the number of TDLU decreases the number of glandular structures available for r-p185(neu)-dependent mammary carcinogenesis, resulting in a significant inhibition of mammary carcinoma development.


Subject(s)
Cancer Vaccines/immunology , Cell Transformation, Neoplastic/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Receptor, ErbB-2/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Autoimmunity/immunology , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Cell Transformation, Neoplastic/genetics , Conserved Sequence , Female , Humans , Immunohistochemistry , Male , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plasmids/genetics , Plasmids/immunology , Pregnancy , Proto-Oncogene Mas , Receptor, ErbB-2/genetics , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
15.
Cancer Res ; 65(16): 7065-70, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103053

ABSTRACT

MicroRNAs (miRNAs) are a class of small noncoding RNAs that control gene expression by targeting mRNAs and triggering either translation repression or RNA degradation. Their aberrant expression may be involved in human diseases, including cancer. Indeed, miRNA aberrant expression has been previously found in human chronic lymphocytic leukemias, where miRNA signatures were associated with specific clinicobiological features. Here, we show that, compared with normal breast tissue, miRNAs are also aberrantly expressed in human breast cancer. The overall miRNA expression could clearly separate normal versus cancer tissues, with the most significantly deregulated miRNAs being mir-125b, mir-145, mir-21, and mir-155. Results were confirmed by microarray and Northern blot analyses. We could identify miRNAs whose expression was correlated with specific breast cancer biopathologic features, such as estrogen and progesterone receptor expression, tumor stage, vascular invasion, or proliferation index.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Blotting, Northern , Breast Neoplasms/pathology , Gene Expression Profiling , Humans
16.
Clin Cancer Res ; 11(5): 1941-52, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15756020

ABSTRACT

PURPOSE: Whereas neoadjuvant therapy is emerging as a treatment option in early primary breast cancer, no data are available on the use of antiangiogenic and immunomodulatory agents in a neoadjuvant setting. In a model of Her-2 spontaneous mammary cancer, we investigated the efficacy of neoadjuvant interleukin 12 (IL-12) followed by "immune-surgery" of the residual tumor. EXPERIMENTAL DESIGN: Female BALB/c mice transgenic for the rat Her-2 oncogene inexorably develop invasive carcinomas in all their mammary glands by the 23rd week of age. Mice with multifocal in situ carcinomas received four weekly i.p. injections of 100 ng IL-12 followed by a 3-week rest. This course was given four times. A few mice additionally received DNA plasmids encoding portions of the Her-2 receptor electroporated through transcutaneous electric pulses. RESULTS: The protection elicited by IL-12 in combination with two DNA vaccine electroporations kept 63% of mice tumor-free. Complete protection of all 1-year-old mice was achieved when IL-12-treated mice received four vaccine electroporations. Pathologic findings, in vitro tests, and the results from immunization of both IFN-gamma and immunoglobulin gene knockout transgenic mice and of adoptive transfer experiments all show that IL-12 augments the B- and T-cell response elicited by vaccination and slightly decreases the number of regulatory T cells. In addition, IL-12 strongly inhibits tumor angiogenesis. CONCLUSIONS: In Her-2 transgenic mice, IL-12 impairs tumor progression and triggers innate immunity so markedly that DNA vaccination becomes effective at late points in time when it is ineffective on its own.


Subject(s)
Cancer Vaccines , Carcinoma/genetics , Carcinoma/immunology , Electroporation , Genes, erbB-2 , Interleukin-12/pharmacology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/immunology , Animals , Female , Immune System/physiology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoadjuvant Therapy , Plasmids/genetics
17.
Cancer Res ; 64(22): 8428-34, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548714

ABSTRACT

Vaccines effectively prevent the onset of tumors in transgenic mice carrying activated oncogenes; however, human tumors are caused by combined alterations in oncogenes and oncosuppressor genes. We evaluated the impact of prophylactic vaccines in HER-2/neu transgenic, p53 wild-type/null mice that succumb to an aggressive cancer syndrome comprising mammary and salivary gland carcinomas and rhabdomyosarcoma. A vaccine made of allogeneic mammary carcinoma cells expressing HER-2/neu and interleukin 12 afforded long-term protection from tumor onset. Tumor prevention was mediated by T cell-derived cytokines, in particular gamma-interferon, and by anti-HER-2/neu antibodies. HER-2/neu expression was inhibited in target tissues of vaccinated mice, and somatic loss of the wild-type p53 allele did not occur. A highly effective vaccine against a single oncoprotein induced a powerful immune response that arrested multistep carcinogenesis in distinct target tissues.


Subject(s)
Neoplasms, Experimental/prevention & control , Animals , Cell Line, Tumor , Female , Genes, erbB-2 , Genes, p53 , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/genetics , Rats
18.
Cancer Res ; 63(10): 2518-25, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12750275

ABSTRACT

Within 33 weeks of life, all 10 mammary glands of virgin BALB/c mice transgenic for the transforming rat HER-2/neu oncogene under the mammary tumor virus promoter (BALB-neuT mice) progress from atypical hyperplasia to invasive palpable carcinoma. Repeated DNA vaccination with plasmids coding for the extracellular and transmembrane domain of the protein product of rat HER-2/neu (r-p185(neu)) delayed tumor onset and reduced tumor multiplicity, but this protection eventually declined, and few mice were tumor free at 1 year of age. Association of plasmid vaccination with administration of soluble mouse LAG-3 (lymphocyte activation gene-3/CD223) generated by fusing the extracellular domain of murine LAG-3 to a murine IgG2a Fc portion (mLAG-3Ig) elicited a stronger and sustained protection that kept 70% of 1-year-old mice tumor free. Moreover, this combined vaccination, which was performed when multiple in situ carcinomas were already evident, extended disease-free survival and reduced carcinoma multiplicity. Inhibition of carcinogenesis was associated with markedly reduced epithelial cell proliferation and r-p185(neu) expression, whereas the few remaining hyperplastic foci were heavily infiltrated by reactive leukocytes. A stronger and enduring r-p185(neu)-specific cytotoxicity, a sustained release of IFN-gamma and interleukin 4, and a marked expansion of both CD8(+)/CD11b(+)/CD28(+) effector and CD8(+)/CD11b(+)/CD28(-) memory effector T-cell populations were induced in immunized mice. This combined vaccination also elicited a quicker and higher antibody response to r-p185(neu), as well as an early antibody isotype switch. These data suggest that the appropriate costimulation provided by mLAG-3Ig enables DNA vaccination to establish an effective protection, probably by enhancing cross-presentation of the DNA coded antigen.


Subject(s)
Antigens, CD , Cancer Vaccines/therapeutic use , Genes, erbB-2/physiology , Mammary Neoplasms, Experimental/prevention & control , Membrane Proteins/pharmacology , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/blood , Antibodies, Neoplasm/immunology , Cancer Vaccines/immunology , Carcinoma in Situ/genetics , Carcinoma in Situ/immunology , Carcinoma in Situ/pathology , Carcinoma in Situ/therapy , Disease Progression , Female , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Vaccines, DNA/immunology , Lymphocyte Activation Gene 3 Protein
19.
Cancer Res ; 63(11): 2728-32, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12782574

ABSTRACT

Rhabdomyosarcomas derive from the skeletal muscle lineage and harbor a variety of genetic and molecular lesions. However, it is not clear which molecular alterations have a pathogenetic role. We show that activation of the HER-2/neu oncogene coupled with inactivation of the oncosuppressor gene p53 causes rhabdomyosarcoma in mice. At the age of 11-21 weeks, all male mice carrying both genetic lesions developed embryonal rhabdomyosarcomas expressing desmin, myosin, and insulin-like growth factor-II, in the genitourinary tract. Our findings led to the hypothesis that the interaction between HER family genes and the p53 pathway might be involved in the origin of human rhabdomyosarcoma.


Subject(s)
Genes, erbB-2/genetics , Genes, p53/genetics , Muscle Neoplasms/genetics , Mutation , Rhabdomyosarcoma/genetics , Alleles , Animals , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Muscle Neoplasms/metabolism , Muscle, Skeletal/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Rhabdomyosarcoma/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
20.
Cancer Res ; 64(8): 2858-64, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15087404

ABSTRACT

The transforming rat Her-2/neu oncogene embedded into the genome of virgin transgenic BALB/c mice (BALB-neuT) provokes the development of an invasive carcinoma in each of their 10 mammary glands. i.m. vaccination with DNA plasmids coding for the extracellular and transmembrane domains of the protein product of the Her-2/neu oncogene started when mice already display multifocal in situ carcinomas temporarily halts neoplastic progression, but all mice develop a tumor by week 43. By contrast, progressive clearance of neoplastic lesions and complete protection of all 1-year-old mice are achieved when the same plasmids are electroporated at 10-week intervals. Pathological findings, in vitro tests, and the results from the immunization of both IFN-gamma and immunoglobulin gene knockout BALB-neuT mice, and of adoptive transfer experiments, all suggest that tumor clearance rests on the combination of antibodies and IFN-gamma-releasing T cells. These findings show that an appropriate vaccine effectively inhibits the progression of multifocal preneoplastic lesions.


Subject(s)
Genes, erbB-2/genetics , Immunotherapy, Active/methods , Immunotherapy, Adoptive/methods , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Vaccines, DNA/administration & dosage , Animals , Antibody-Dependent Cell Cytotoxicity , Carcinoma in Situ/genetics , Carcinoma in Situ/immunology , Carcinoma in Situ/therapy , Electroporation/methods , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptor, ErbB-2/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL