Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 129(5): 1003-17, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26795563

ABSTRACT

The role of Ras in human skin tumorigenesis induction is still ambiguous. Overexpression of oncogenic Ras causes premature senescence in cultured human cells and hyperplasia in transgenic mice. Here, we investigated whether the oncogenic insult outcome might depend on the nature of the founding keratinocyte. We demonstrate that overexpression of the constitutively active Ras-V12 induces senescence in primary human keratinocyte cultures, but that some cells escape senescence and proliferate indefinitely. Ras overexpression in transient-amplifying- or stem-cell-enriched cultures shows that p16 (encoded by CDKN2A) levels are crucial for the final result. Indeed, transient-amplifying keratinocytes expressing high levels of p16 are sensitive to Ras-V12-induced senescence, whereas cells with high proliferative potential, but that do not display p16, are resistant. The subpopulation that sustains the indefinite culture growth exhibits stem cell features. Bypass of senescence correlates with inhibition of the pRb (also known as RB1) pathway and resumption of telomerase reverse transcriptase (TERT) activity. Immortalization is also sustained by activation of the ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1) and Akt pathways. Moreover, only transduced cultures originating from cultures bearing stem cells induce tumors in nude mice. Our findings demonstrate that the Ras overexpression outcome depends on the clonogenic potential of the recipient keratinocyte and that only the stem cell compartment is competent to initiate tumorigenesis.


Subject(s)
Keratinocytes/enzymology , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cellular Senescence , Coculture Techniques , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Transplantation , Neoplastic Stem Cells/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Skin Neoplasms/pathology
2.
J Lipid Res ; 53(8): 1654-61, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22611251

ABSTRACT

Successful development of drugs against novel targets crucially depends on reliable identification of the activity of the target gene product in vivo and a clear demonstration of its specific functional role for disease development. Here, we describe an immunological knockdown (IKD) method, a novel approach for the in vivo validation and functional study of endogenous gene products. This method relies on the ability to elicit a transient humoral response against the selected endogenous target protein. Anti-target antibodies specifically bind to the target protein and a fraction of them effectively neutralize its activity. We applied the IKD method to the in vivo validation of plasma PCSK9 as a potential target for the treatment of elevated levels of plasma LDL-cholesterol. We show that immunization with human-PCSK9 in mice is able to raise antibodies that cross-react and neutralize circulating mouse-PCSK9 protein thus resulting in increased liver LDL receptor levels and plasma cholesterol uptake. These findings closely resemble those described in PCSK9 knockout mice or in mice treated with antibodies that inhibit PCSK9 by preventing the PCSK9/LDLR interaction. Our data support the IKD approach as an effective method to the rapid validation of new target proteins.


Subject(s)
Cholesterol, LDL/blood , Immunization , Proprotein Convertases/immunology , Serine Endopeptidases/immunology , Animals , Antibodies/immunology , Female , HEK293 Cells , Humans , Liver/metabolism , Mice , Proprotein Convertase 9 , Proprotein Convertases/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism
3.
J Cell Physiol ; 227(10): 3381-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22213458

ABSTRACT

The role of the ErbB3 receptor in signal transduction is to augment the signaling repertoire of active heterodimeric ErbB receptor complexes through activating the PI3K/AKT pathway, which in turn promotes survival and proliferation. ErbB3 has recently been proposed to be involved in acquired resistance to tyrosine kinase inhibitors (TKIs), and is therefore a promising new drug cancer target. Since ErbB3 is a kinase defective receptor, it cannot be targeted by small molecule inhibitors, whereas monoclonal antibodies may offer a viable strategy for pharmacological intervention. In this study, we have utilized DNA electroporation (DNA-EP) to generate a set of novel hybridomas directed against human ErbB3, which have been characterized for their biochemical and functional properties and selected for their ability to negatively regulate the ErbB3-mediated signaling pathway. In vitro, the anti-ErbB3 antibodies modulate the growth rate of cancer cells of different origins. In vivo they show antitumoral properties in a xenograft model of human pancreatic tumor and in the ErbB2-driven carcinogenesis genetically engineered mouse model (GEMM) for mammary tumor, the BALB/neuT. Our data confirm that downregulating the ErbB3-mediated signals with the use of anti-ErbB3 monoclonal antibodies is both feasible and relevant for therapeutic purposes and provides new opportunities for novel anti-ErbB3 combinatory strategies for cancer treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Receptor, ErbB-3/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous
4.
PLoS One ; 6(7): e21320, 2011.
Article in English | MEDLINE | ID: mdl-21789168

ABSTRACT

Malignant pleural effusions (MPEs) could represent an excellent source to culture a wide variety of cancer cells from different donors. In this study, we set up culture conditions for cancer cells deriving from MPEs of several patients affected by the most frequent form of lung cancer, namely the subset of non small cell lung cancers (NSCLC) classified as Lung Adenocarcinomas (AdenoCa) which account for approximately 40% of lung cancer cases. AdenoCa malignant pleural effusions gave rise to in vitro cultures both in adherent and/or in spheroid conditions in almost all cases analyzed. We characterized in greater detail two samples which showed the most efficient propagation in vitro. In these samples we also compared gene profiles of spheroid vs adherent cultures and identified a set of differentially expressed genes. Finally we achieved efficient tumor engraftment in recipient NOD/SCID mice, also upon inoculation of small number of cells, thus suggesting indirectly the presence of tumor initiating cells.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Pleural Effusion, Malignant/genetics , Pleural Effusion, Malignant/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays , Adenocarcinoma of Lung , Aged , Aged, 80 and over , Aldehyde Dehydrogenase/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Separation , Computational Biology , Female , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , Immunohistochemistry , Immunophenotyping , Male , Mice , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
5.
Mol Ther ; 18(8): 1559-67, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20531395

ABSTRACT

Canine cancers occur with an incidence similar to that of humans and share many features with human malignancies including histological appearance, tumor genetics, biological behavior, and response to conventional therapies. As observed in humans, the telomerase reverse transcriptase (TERT) activity is largely confined to tumor tissues and absent in the majority of normal dog tissues. Therefore, dog TERT (dTERT) can constitute a valid target for translational cancer immunotherapy. We have evaluated the ability of adenovirus serotype 6 (Ad6) and DNA electroporation (DNA-EP) to induce immune responses against dTERT in dogs affected by malignant lymphoma (ML). The vaccine was combined with standard chemotherapy regimen [cyclophosphamide, vincristine, prednisone (COP)]. dTERT-specific immune response was induced in 13 out of 14 treated animals (93%) and remained detectable and long-lasting with the absence of autoimmunity or other side effects. Most interestingly, the survival time of vaccine/Chemo-treated dogs was significantly increased over historic controls of Chemo-treated animals (>97.8 versus 37 weeks, respectively, P = 0.001). Our results show that Ad6/DNA-EP-based cancer vaccine against dTERT overcomes host immune tolerance, should be combined with chemotherapy, induces long-lasting immune responses, and significantly prolongs the survival of ML canine patients. These data support further evaluation of this approach in human clinical trials.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Lymphoma, B-Cell/immunology , Telomerase/immunology , Adenoviridae/genetics , Animals , Dogs , Electroporation , Humans , Lymphoma, B-Cell/metabolism
6.
Cancer Res ; 70(6): 2476-84, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197467

ABSTRACT

ERBB2/neu and Notch signaling are known to be deregulated in many human cancers. However, pathway cross-talk and dependencies are not well understood. In this study, we use an ERBB2-transgenic mouse model of breast cancer (neuT) to show that Notch signaling plays a critical role in tumor maintenance. Inhibition of the Notch pathway with a gamma-secretase inhibitor (GSI) decreased both the Notch and the mammalian target of rapamycin/AKT pathways. Antitumor activity resulting from GSI treatment was associated with decreased cell proliferation as measured by Ki67 and decreased expression of glucose transporter Glut1. Positron emission tomography (PET) imaging showed that the functional consequences of decreased Glut1 translated to reduced glucose uptake and correlated with antitumor effects as measured by micro-computed tomography imaging. The decrease of Glut1 in neuT tumors was also observed in several human breast cancer cell lines following GSI treatment. We provide evidence that approximately 27% of ERBB2-positive human breast cancer specimens display high expression of HES1, phospho-S6RP, and GLUT1. Together, these results suggest that pathways downstream of Notch signaling are, at least in part, responsible for promoting tumor growth in neuT and also active in both neuT and a subset of human breast cancers. These findings suggest that GSI may provide therapeutic benefit to a subset of ERBB2-positive breast cancers and that [(18)F]FDG-PET imaging may be useful in monitoring clinical response.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Glucose/pharmacokinetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cyclic S-Oxides/pharmacology , Down-Regulation/drug effects , Female , Fluorodeoxyglucose F18/pharmacokinetics , Glucose Transporter Type 1/biosynthesis , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oncogene Protein v-akt/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Thiadiazoles/pharmacology
7.
Vaccine ; 28(5): 1201-8, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19944791

ABSTRACT

Pet dogs represent a valuable pre-clinical model to assess the efficacy of oncology drugs. Additionally, canine cancers occur with an incidence similar to that of humans and share many features with human malignancies including histological appearance, tumor genetics, biological behavior and response to conventional therapies. The telomerase reverse transcriptase (TERT) is reactivated in most of human and dog tumors. Similarly, HER-2/neu oncoprotein is overexpressed in a proportion of canine breast cancers. Therefore, TERT and HER-2/neu can constitute valid tumor associated antigens (TAA), suitable targets for translational cancer immunotherapy in dogs. In this study, we have evaluated the ability of DNA electroporation (DNA-EP) and Adenovirus serotype 6 (Ad6) to induce immune responses against dog TERT (dTERT) and HER-2/neu in healthy dogs. Vaccination was effective in all treated animals and the adaptive immune response remained detectable and long-lasting in the absence of autoimmunity or other side-effects. Our results show that DNA-EP/Ad6-based cancer vaccine induces adaptive immune responses against TAA in canine subjects and support further evaluation of this approach in cancer dog patients.


Subject(s)
Cancer Vaccines/immunology , Immunization , Mammary Neoplasms, Animal/prevention & control , Receptor, ErbB-2/immunology , Adaptive Immunity , Adenoviridae , Animals , Cancer Vaccines/genetics , Dog Diseases , Dogs , Electroporation , Female , Humans , Mammary Neoplasms, Animal/immunology , Mice , Mice, Inbred BALB C , Receptor, ErbB-2/genetics , Telomerase
8.
Mol Ther ; 17(10): 1804-13, 2009 10.
Article in English | MEDLINE | ID: mdl-19623161

ABSTRACT

The human telomerase reverse transcriptase (hTERT) is an attractive target for human cancer vaccination because its expression is reactivated in most human tumors. We have evaluated the ability of DNA electroporation (DNA-EP) and adenovirus serotype 6 (Ad6) to induce immune responses against hTERT in nonhuman primates (NHPs) (Macaca mulatta). Vaccination was effective in all treated animals, and the adaptive immune response remained detectable and long lasting without side effects. To further enhance the efficacy of the hTERT vaccine, we evaluated the combination of hTERT vaccine and a novel TLR9 agonist, referred to as immunomodulatory oligonucleotide (IMO). Monkeys were dosed weekly with IMO concurrently with the vaccine regimen and showed increases in cytokine secretion and activation of natural killer (NK) cells compared with the group that received vaccine alone. Using a peptide array, a specific profile of B-cell reactive epitopes was identified when hTERT vaccine was combined with IMO. The combination of IMO with hTERT genetic vaccine did not impact vaccine-induced TERT-specific cell-mediated immunity. Our results show that appropriate combination of a DNA-EP/Ad6-based cancer vaccine against hTERT with IMO induces multiple effects on innate and adaptive immune responses in NHPs.


Subject(s)
Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Telomerase/immunology , Toll-Like Receptor 9/agonists , Adenoviridae/genetics , Animals , Electroporation , Epitopes, B-Lymphocyte/immunology , Immunity, Cellular/immunology , Immunity, Innate/genetics , Interferon-alpha/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-6/metabolism , Macaca mulatta , Telomerase/genetics
9.
BMC Genomics ; 10: 135, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19327144

ABSTRACT

BACKGROUND: Spontaneous tumors in dog have been demonstrated to share many features with their human counterparts, including relevant molecular targets, histological appearance, genetics, biological behavior and response to conventional treatments. Mammary tumors in dog therefore provide an attractive alternative to more classical mouse models, such as transgenics or xenografts, where the tumour is artificially induced. To assess the extent to which dog tumors represent clinically significant human phenotypes, we performed the first genome-wide comparative analysis of transcriptional changes occurring in mammary tumors of the two species, with particular focus on the molecular pathways involved. RESULTS: We analyzed human and dog gene expression data derived from both tumor and normal mammary samples. By analyzing the expression levels of about ten thousand dog/human orthologous genes we observed a significant overlap of genes deregulated in the mammary tumor samples, as compared to their normal counterparts. Pathway analysis of gene expression data revealed a great degree of similarity in the perturbation of many cancer-related pathways, including the 'PI3K/AKT', 'KRAS', 'PTEN', 'WNT-beta catenin' and 'MAPK cascade'. Moreover, we show that the transcriptional relationships between different gene signatures observed in human breast cancer are largely maintained in the canine model, suggesting a close interspecies similarity in the network of cancer signalling circuitries. CONCLUSION: Our data confirm and further strengthen the value of the canine mammary cancer model and open up new perspectives for the evaluation of novel cancer therapeutics and the development of prognostic and diagnostic biomarkers to be used in clinical studies.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/genetics , Gene Expression Profiling , Mammary Neoplasms, Animal/genetics , Animals , Cluster Analysis , Comparative Genomic Hybridization , Computational Biology , Disease Models, Animal , Dog Diseases/genetics , Dogs , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Principal Component Analysis
10.
Hum Gene Ther ; 19(7): 670-80, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18533895

ABSTRACT

The aim of this study was to evaluate the efficacy of genetic vaccination with rat ErbB2 antigen in a therapeutic setting for the BALB-neuT mouse model of mammary carcinoma and to establish immunological correlates with vaccine efficacy. To define an early therapeutic setting we performed imaging studies of mouse mammary glands with a high-frequency ultrasound system that allowed the diagnosis of tumor lesions before they become palpable, starting from week 13 after mouse births. An intensive immunization protocol of vaccination was implemented at this stage, consisting of four weekly DNA injections with electroporation followed by two injections of adenovirus carrying the codon usage-optimized cDNA encoding the extracellular-transmembrane domain of rat ErbB2. Immunological parameters were monitored in each individual mouse by analyzing peripheral blood leukocytes. The appearance of the first palpable tumor in vaccinated mice was delayed and there was a statistically significant time gap before additional masses developed, indicating disease stabilization. As a result of the immunization, antibodies and CD8(+) T cells to rat ErbB2 were detected and the amplitude of elicited responses correlated with the efficacy of vaccination. Moreover, the vaccination regimen specifically halted the rise in circulating myeloid suppressor cells (MSCs). All three parameters, that is, CD8(+) T cells, antibodies to rat ErbB2, and circulating MSCs, measured at the end of vaccination could be used as predictive biomarkers for future tumor development. This study emphasizes the potential of genetic vaccines for the therapeutic treatment of malignancies and suggests possible predictive biomarkers to be further validated in the clinic for the follow-up of vaccinated cancer patients.


Subject(s)
Adenoviridae/genetics , Antibodies, Neoplasm/blood , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Genes, erbB-2/immunology , Genetic Vectors , Mammary Neoplasms, Experimental/therapy , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Disease-Free Survival , Female , Genes, erbB-2/genetics , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mammary Glands, Animal/diagnostic imaging , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Predictive Value of Tests , Rats , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Ultrasonography , Vaccination
11.
J Med Chem ; 51(8): 2350-3, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18370373

ABSTRACT

Histone deacetylase (HDAC) inhibitors offer a promising strategy for cancer therapy, and the first generation HDAC inhibitors are currently in the clinic. Entirely novel ketone HDAC inhibitors have been developed from the cyclic tetrapeptide apicidin. These compounds show class I subtype selectivity and levels of cellular activity comparable to clinical candidates. A representative example has demonstrated tumor growth inhibition in a human colon HCT-116 carcinoma xenograft model comparable to known inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Ketones/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Histone Deacetylases/chemistry , Humans , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...