Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
1.
Stem Cell Res ; 23: 154-157, 2017 08.
Article in English | MEDLINE | ID: mdl-28925363

ABSTRACT

Multiple Endocrine Neoplasia Type 2B (MEN2B) is a cancer-predisposing syndrome that affects patients with germline RET mutations. The clinical spectrum of the syndrome includes medullary thyroid carcinoma (MTC) and pheochromocytoma. Currently, there is no satisfactory model recapitulating all the features of the disease especially at the level of stem cells. We generated induced pluripotent stem cells (iPSCs) from a patient with RET mutation at codon 918 who developed pheochromocytoma and MTC. These iPSC had normal karyotype, harboured the RETM918T mutation and expressed pluripotency hallmarks. A comprehensive pathological assessment of teratoma was performed after injection in immunodeficient mice.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/pathology , Multiple Endocrine Neoplasia Type 2b/genetics , Multiple Endocrine Neoplasia Type 2b/pathology , Mutation/genetics , Proto-Oncogene Proteins c-ret/genetics , Cell Line , Humans , Risk Factors , Young Adult
2.
Stem Cell Res ; 17(2): 235-237, 2016 09.
Article in English | MEDLINE | ID: mdl-27879209

ABSTRACT

Chronic myeloid leukemia (CML) is a clonal malignancy initiated by the occurrence of a t (9;22) translocation, generating Ph1 chromosome and BCR-ABL oncogene in a primitive hematopoietic stem cell (HSC). The resistance of HSC to targeted therapies using tyrosine kinase inhibitors remains a major obstacle towards the cure. We have generated an iPSC line from a patient with CML using leukemic CD34+ cells cryopreserved at diagnosis. Ph1+ CML cells were reprogrammed by non-integrative viral transduction. These iPSCs harboured Ph1 chromosome and expressed pluripotency hallmarks as well as BCR-ABL. Teratoma assays revealed normal differentiation after injection in immunodeficient mice.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukocytes, Mononuclear/cytology , Adolescent , Animals , Antigens, CD34/metabolism , Cell Differentiation , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Karyotype , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred NOD , Teratoma/metabolism , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Stem Cell Res ; 17(1): 154-157, 2016 07.
Article in English | MEDLINE | ID: mdl-27558615

ABSTRACT

Multiple Endocrine Neoplasia Type 2A (MEN2A) is a cancer-predisposing syndrome that affects patients with germline RET mutations. The clinical spectrum of the syndrome includes medullary thyroid carcinoma (MTC), pheochromocytoma, hyperparathyroidism and cutaneous lichen amyloidosis (CLA) and/or Hirschsprung disease in some variants. Currently, there is no satisfactory animal model recapitulating all the features of the disease especially at the level of stem cells. We generated induced pluripotent stem cells (iPSCs) from a patient with RET mutation at codon 634 who developed pheochromocytoma and MTC. RETC634Y-mutated cells were reprogrammed by non-integrative viral transduction. These iPSCs had normal karyotype, harboured the RETC634Y mutation and expressed pluripotency hallmarks as well as RET. A comprehensive pathological assessment of teratoma was performed after injection in immunodeficient mice.

4.
Oncogene ; 34(24): 3207-13, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25065593

ABSTRACT

Osteosarcoma is the most prevalent primary pediatric cancer-related bone disease. These tumors frequently develop resistance to chemotherapy and are highly metastatic, leading to poor outcome. Thus, there is a need for new therapeutic strategies that can prevent cell dissemination. We previously showed that CYR61/CCN1 expression in osteosarcoma cells is correlated to aggressiveness both in vitro and in vivo in mouse models, as well as in patients. In this study, we found that CYR61 is a critical contributor to the vascularization of primary tumor. We demonstrate that silencing CYR61, using lentiviral transduction, leads to a significant reduction in expression level of pro-angiogenic markers such as VEGF, FGF2, PECAM and angiopoietins concomitantly to an increased expression of major anti-angiogenic markers such as thrombospondin-1 and SPARC. Matrix metalloproteinase-2 family member expression, a key pathway in osteosarcoma metastatic capacity was also downregulated when CYR61 was downregulated in osteosarcoma cells. Using a metastatic murine model, we show that CYR61 silencing in osteosarcoma cells results in reduced tumor vasculature and slows tumor growth compared with control. We also find that microvessel density correlates with lung metastasis occurrence and that CYR61 silencing in osteosarcoma cells limits the number of metastases. Taken together, our data indicate that CYR61 silencing can blunt the malignant behavior of osteosarcoma tumor cells by limiting primary tumor growth and dissemination process.


Subject(s)
Bone Neoplasms/blood supply , Cysteine-Rich Protein 61/genetics , Neovascularization, Pathologic/genetics , Osteosarcoma/blood supply , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Proliferation/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Metastasis , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA Interference , Tumor Cells, Cultured
5.
Cell Death Dis ; 5: e1423, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25232677

ABSTRACT

Radiotherapy has a critical role in the treatment of small-cell lung cancer (SCLC). The effectiveness of radiation in SCLC remains limited as resistance results from defects in apoptosis. In the current study, we investigated whether using the Bcl-2/Bcl-XL inhibitor S44563 can enhance radiosensitivity of SCLC cells in vitro and in vivo. In vitro studies confirmed that S44563 caused SCLC cells to acquire hallmarks of apoptosis. S44563 markedly enhanced the sensitivity of SCLC cells to radiation, as determined by a clonogenic assay. The combination of S44563 and cisplatin-based chemo-radiation showed a significant tumor growth delay and increased overall survival in mouse xenograft models. This positive interaction was greater when S44563 was given after the completion of the radiation, which might be explained by the radiation-induced overexpression of anti-apoptotic proteins secondary to activation of the NF-κB pathway. These data underline the possibility of combining IR and Bcl-2/Bcl-XL inhibition in the treatment of SCLC as they underscore the importance of administering conventional and targeted therapies in an optimal sequence.


Subject(s)
Heterocyclic Compounds, 3-Ring/administration & dosage , Lung Neoplasms/radiotherapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Radiation-Sensitizing Agents/administration & dosage , Small Cell Lung Carcinoma/radiotherapy , Sulfonamides/administration & dosage , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cisplatin/administration & dosage , Combined Modality Therapy , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Tolerance , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Xenograft Model Antitumor Assays , bcl-X Protein/genetics , bcl-X Protein/metabolism
7.
Cell Death Dis ; 3: e396, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23034331

ABSTRACT

The chemokine receptor CXCR4 favors the interaction of acute myeloid leukemia (AML) cells with their niche but the extent to which it participates in pathogenesis is unclear. Here, we show that CXCR4 expression at the surface of leukemic cells allowed distinguishing CXCR4 (high) from CXCR4(neg/low) AML patients. When high levels of CXCR4 are expressed at the surface of AML cells, blocking the receptor function with small molecule inhibitors could promote leukemic cell death and reduce NOD/Shi-scid/IL-2Rγ(null) (NOG) leukemia-initiating cells (LICs). Conversely, these drugs had no efficacy when AML cells do not express CXCR4 or when they do not respond to chemokine CXC motif ligand 12 (CXCL12). Functional analysis showed a greater mobilization of leukemic cells and LICs in response to drugs, suggesting that they target the interaction between leukemic cells and their supportive bone marrow microenvironment. In addition, increased apoptosis of leukemic cells in vitro and in vivo was observed. CXCR4 expression level on AML blast cells and their migratory response to CXCL12 are therefore predictive of the response to the inhibitors and could be used as biomarkers to select patients that could potentially benefit from the drugs.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Receptors, CXCR4/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Apoptosis/drug effects , Benzylamines , Chemokine CXCL12/metabolism , Child, Preschool , Cyclams , Disease Models, Animal , Female , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Middle Aged , Receptors, CXCR4/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Cancer Gene Ther ; 17(10): 700-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20559333

ABSTRACT

Vascular endothelial-specific cadherin (VE-cadherin) is an endothelial cell-specific adhesion molecule, localized at cell-cell contact sites. It is involved in physiological and pathological angiogenesis. In this study, we showed that in vitro a soluble N-terminal fragment of VE-cadherin (EC1-3) corresponding to cadherin 1-3 ectodomains inhibited vascular endothelial growth factor-stimulated endothelial cell proliferation and capillary tube structure formation in the matrigel model. In vivo, EC1-3 was tested in a murine colon cancer model. EC1-3-expressing colon cancer C51 cells were subcutaneously grafted into nude mice, and tumor growth and angiogenesis were evaluated. At day 33, the mean volume of the tumors developed was reduced (510±104 versus 990±120 mm(3) for control). Similarly, injection of EC1-3 virus-producing cells into established C51 tumors resulted in an inhibition by 33% of tumor growth. Immunohistological staining of vessels on tumor sections showed a significantly reduced intratumoral angiogenesis. Furthermore, EC1-3 did not induce vessel injury in the lung, liver, spleen, heart and brain in the mice. These results suggest that the soluble N-terminal fragment of VE-cadherin EC1-3 could exert an antitumoral effect by targeting tumor angiogenesis, which included blocking endothelial cell proliferation and capillary tube formation with no obvious toxicity on normal organs.


Subject(s)
Adenocarcinoma/metabolism , Angiogenesis Inhibitors/pharmacology , Antigens, CD/pharmacology , Cadherins/pharmacology , Colonic Neoplasms/metabolism , Endothelial Cells/metabolism , Angiogenesis Inhibitors/metabolism , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Nude , Transduction, Genetic , Vascular Endothelial Growth Factor A/metabolism
9.
Gene Ther ; 16(5): 605-19, 2009 May.
Article in English | MEDLINE | ID: mdl-19262615

ABSTRACT

The woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) is widely used in retroviral gene transfer vectors. However, this element contains an open-reading frame (ORF) encoding a truncated peptide of the woodchuck hepatitis virus X protein (WHX). Because we are developing a lentiviral vector for the gene therapy of Wiskott-Aldrich syndrome (WAS), we evaluated whether the WPRE was needed in the gene transfer cassette and tested the possibility of replacing it with a mutated derivative. The transcriptional activity of the WPRE was undetectable in the context of the lentiviral vector but the element was capable of translating a polypeptide. This capability was abrogated by mutating the WHX ORF translation start. The WPRE was required to express high levels of the transgene and for that, the native form or mutated derivatives functioned equivalently. The vector using a WAS gene promoter and the mut6 WPRE induced long-term expression of the WAS transgene in vivo, correcting cytoskeletal defects, thymocyte and B-cell numbers and improved the colitis of WAS-null mice. By providing additional evidence of efficacy of this WAS lentiviral vector with improved safety features, our results validate a mutated WPRE, which should be useful in future gene therapy applications.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hepatitis B Virus, Woodchuck/genetics , Trans-Activators/genetics , Wiskott-Aldrich Syndrome/therapy , Animals , Cell Line , Colitis/pathology , Colitis/therapy , Gene Expression Regulation , Humans , Lentivirus/genetics , Mice , Mutation , Open Reading Frames/genetics , Plasmids/genetics , Trans-Activators/biosynthesis , Transduction, Genetic , Transgenes , Wiskott-Aldrich Syndrome/pathology
10.
Cell Prolif ; 42(1): 38-48, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19143762

ABSTRACT

Angiogenesis is considered as an essential process for tumour development and invasion. Previously, we demonstrated that cyclin-dependent kinase inhibition by roscovitine induces a radiosensitization and a synergistic antitumoral effect in human carcinoma but its effect on the microenvironment and tumour angiogenesis remains unknown. Here, we investigated the effect of the combination roscovitine and ionizing radiation (IR) on normal cells in vitro and on tumour angiogenesis in MDA-MB 231 tumour xenografts. We observed that the combination roscovitine and IR induced a marked reduction of angiogenic hot spot and microvascular density in comparison with IR or roscovitine treatments alone. The Ang-2/Tie-2 ratio was increased in presence of reduced vascular endothelial growth factor level suggesting vessel destabilization. In vitro, no radiosensitization effect of roscovitine was found in endothelial, fibroblast, and keratinocyte cells. IR potentiated the antiproliferative effect of roscovitine without inducing apoptosis in endothelial cells. Roscovitine decreased IR-stimulated vascular endothelial growth factor secretion of MDA-MB 231 and endothelial cells. A reduction in the endothelial cells invasion and the capillary-like tube formation in Matrigel were observed following the combination roscovitine and IR. This combined treatment targets angiogenesis resulting in microvessel destabilization without inducing normal cell toxicity.


Subject(s)
Breast Neoplasms/blood supply , Neovascularization, Pathologic , Purines/pharmacology , Radiation, Ionizing , Apoptosis , Base Sequence , Breast Neoplasms/pathology , Cell Line, Tumor , Culture Media, Conditioned , DNA Primers , Flow Cytometry , Humans , Immunohistochemistry , Polymerase Chain Reaction , Roscovitine , Vascular Endothelial Growth Factor A/metabolism
11.
Carcinogenesis ; 29(11): 2236-42, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18753414

ABSTRACT

Plasminogen activator inhibitor-1 is known to play a paradoxical positive role in tumor angiogenesis, but its contribution to metastatic spread remains unclear. We studied the impact of plasminogen activator inhibitor (PAI)-1 deficiency in a transgenic mouse model of ocular tumors originating from retinal epithelial cells and leading to brain metastasis (TRP-1/SV40 Tag mice). PAI-1 deficiency did not affect primary tumor growth or vascularization, but was associated with a smaller number of brain metastases. Brain metastases were found to be differentially distributed between the two genotypes. PAI-1-deficient mice displayed mostly secondary foci expanding from local optic nerve infiltration, whereas wild-type animals displayed more disseminated nodules in the scissura and meningeal spaces. SuperArray GEarray analyses aimed at detecting molecules potentially compensating for PAI-1 deficiency demonstrated an increase in fibroblast growth factor-1 (FGF-1) gene expression in primary tumors, which was confirmed by reverse transcription-polymerase chain reaction and western blotting. Our data provide the first evidence of a key role for PAI-1 in a spontaneous model of metastasis and suggest that angiogenic factors, such as FGF-1, may be important for primary tumor growth and may compensate for the absence of PAI-1. They identify PAI-1 and FGF-1 as important targets for combined antitumor strategies.


Subject(s)
Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Eye Neoplasms/pathology , Plasminogen Activator Inhibitor 1/physiology , Animals , Blotting, Western , Brain Neoplasms/genetics , Eye Neoplasms/genetics , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 1/genetics , Retinal Pigment Epithelium/pathology , Reverse Transcriptase Polymerase Chain Reaction
12.
Gene Ther ; 15(21): 1436-45, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18548116

ABSTRACT

Given as a prophylactic treatment, a single muscle electrogene transfer of plasmid coding canstatin fused to human serum albumin (CanHSA), slowed down the development of two xenografted human carcinomas from mammary (MDA-MB-231) and prostate origin (PC-3) in nude mice and delayed lung metastatic spreading of B16F10 melanoma cells in syngenic mice. No effect was observed with unfused canstatin. The long lasting circulating blood level of CanHSA (20 ng ml(-1)) resulted in a profound disorganization of the tumor blood vessel network. However, when used as a curative treatment, on well-established tumors, CanHSA electrogenetherapy was ineffective in reducing tumor growth. As radiation is known to increase the alpha v beta3 and alpha v beta5 integrins, which are canstatin receptors, to extend the use of CanHSA electrogenetherapy, as a curative treatment, we explored the combination of CanHSA and ionizing radiation. We demonstrated a better efficacy (P=0.01) of the bitherapy over irradiation alone, as a result of strong vessel disorganization and dramatic increase of tumor cells apoptosis. This extremely simple virus free curative protocol could open the door to potential clinical applications, especially for prostate cancer that often develops radioresistance.


Subject(s)
Breast Neoplasms/therapy , Collagen Type IV/genetics , Genetic Therapy/methods , Peptide Fragments/genetics , Prostatic Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Apoptosis , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Collagen Type IV/blood , Combined Modality Therapy , Electroporation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Muscle, Skeletal/metabolism , Peptide Fragments/blood , Prostatic Neoplasms/radiotherapy , Radiotherapy Dosage , Recombinant Fusion Proteins/genetics , Serum Albumin/genetics , Xenograft Model Antitumor Assays
13.
Technol Cancer Res Treat ; 6(4): 301-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17668937

ABSTRACT

Immune cell recruitment during the treatment of sarcoma tumors in mice with irreversible electroporation was studied by immunohistochemistry. Irreversible electroporation is a non-thermal tissue ablation technique in which certain short duration electrical fields are used to permanently permeabilize the cell membrane, presumably through the formation of nanoscale defects in the membrane. Employing irreversible electroporation parameters known to completely ablate the tumors without thermal effects we did not find infiltration of immune cells probably because of the destruction of infiltration routes. We confirm here that immune response is not instrumental in irreversible electroporation efficacy, and we propose that irreversible electroporation may be, therefore, a treatment modality of interest to immunodepressed cancer patients.


Subject(s)
Electroporation/methods , Sarcoma, Experimental/immunology , Sarcoma, Experimental/surgery , Animals , Antigens, CD/analysis , Cell Line, Tumor , Female , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Mice , T-Lymphocyte Subsets/immunology
14.
Gene Ther ; 14(2): 108-17, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16943854

ABSTRACT

Tumor growth is dependent both on endothelial and tumor cells. The aim of this study was to investigate dynamically whether changes in tumor vasculature implicate tumor tissue degeneration during antiangiogenic therapies. In order to quantify intra-tumor vascularization and necrosis, we have used ultrasound technology. This study has identified essential parameters needed to quantify specifically and sensitively the number of microvessels and the extent of necrosis in xenografted human carcinomas during natural tumor evolution, using contrast-enhanced high-frequency ultrasonography with (HFCDUS) or without (HFUS) color Doppler. We showed that quantification of intra-tumor microvessels between HFCDUS and immunohistochemistry is correlated using an anti-CD31 antibody. Furthermore, quantification of tumor necrosis with HFUS was confirmed by histological examination of hematoxylin-eosin-saffranin-stained sections over the observation period. Subsequently, for the assessment of novel angiogenic inhibitors, HFCDUS and HFUS were used to elucidate the underlying dynamics linking vessel inhibition and tumor eradication. We describe a novel application for HFCDUS/HFUS that constitutes an effective, convenient, and non-invasive method for clinical assessment of angiogenic inhibitors. In conclusion, we showed that tumor cells abruptly became necrotic following an antivascular therapy, whereas untreated tumors were protected from degeneration by a significant blood supply.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Genetic Therapy/methods , Transduction, Genetic/methods , Adenoviridae/genetics , Angiogenesis Inhibitors/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized , Bevacizumab , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Necrosis , Neovascularization, Pathologic , Random Allocation , Ultrasonography, Doppler, Color , Ultrasonography, Interventional , Xenograft Model Antitumor Assays
15.
Cancer Gene Ther ; 14(3): 251-61, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17082795

ABSTRACT

Different antiangiogenic and antimetastatic recombinant adenoviruses were tested in a transgenic mouse model of metastatic ocular cancer (TRP1/SV40 Tag transgenic mice), which is a highly aggressive tumor, developed from the pigmented epithelium of the retina. These vectors, encoding amino-terminal fragments of urokinase plasminogen activator (ATF), angiostatin Kringles (K1-3), endostatin (ES) and canstatin (Can) coupled to human serum albumin (HSA) were injected to assess their metastatic and antiangiogenic activities in our model. Compared to AdCO1 control group, AdATF-HSA did not significantly reduce metastatic growth. In contrast, mice treated with AdK1-3-HSA, AdES-HSA and AdCan-HSA displayed significantly smaller metastases (1.19+/-1.19, 0.87+/-1.5, 0.43+/-0.56 vs controls 4.04+/-5.12 mm3). Moreover, a stronger inhibition of metastatic growth was obtained with AdCan-HSA than with AdK1-3-HSA (P=0.04). Median survival was improved by 4 weeks. A close correlation was observed between the effects of these viruses on metastatic growth and their capacity to inhibit tumor angiogenesis. Our study indicates that systemic antiangiogenic factors production by recombinant adenoviruses, particularly Can, might represent an effective way of delaying metastatic growth via inhibition of angiogenesis.


Subject(s)
Angiogenesis Inhibitors/genetics , Brain Neoplasms/therapy , Eye Neoplasms/therapy , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Serum Albumin/genetics , Activating Transcription Factors/genetics , Adenoviridae/genetics , Angiogenesis Inhibitors/therapeutic use , Angiostatins/genetics , Animals , Blood Proteins/genetics , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Collagen Type IV/genetics , Endostatins/genetics , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Humans , Mice , Mice, Nude , Mice, Transgenic , Neovascularization, Pathologic , Peptide Fragments/genetics , Survival Rate , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics
16.
Gene Ther ; 14(6): 533-44, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17109009

ABSTRACT

The immunogenicity of recombinant adenoviruses (Ad) constitutes a major concern for their use in gene therapy. Antibody- and cell-mediated immune responses triggered by adenoviral vectors hamper long-term transgene expression and efficient viral readministration. We previously reported that interleukin (IL)-6 and tumor necrosis factor (TNF)-alpha play an essential role in both the acute phase and antibody response against Ad, respectively. As TNF-alpha controls the immune response and the development of the immune system, we examined here the consequence of blockade of TNF-alpha activity through Ad-mediated gene delivery of a dimeric mouse TNFR1-IgG fusion protein on transgene expression from a second Ad. Ad encoding TNFR1-IgG (AdTNFR1-Ig) was injected intravenously along with Ad encoding beta-galactosidase or alpha1-antitrypsin transgene in wild-type (IL-6(+/+)) but also in IL-6-deficient mice (IL-6(-/-)) to analyze how TNF-alpha and IL-6 diminish liver gene transfer efficacy. Blockade of TNF-alpha leads to increased transgene expression in both wild-type and IL-6(-/-) mice due to a reduced inflammatory response and to diminished recruitment of macrophages and NK cells towards the liver. Antibody responses against adenoviral particles and expressed transgenes were only delayed in AdTNFR1-Ig-treated wild-type mice, but were markedly reduced in AdTNFR1-Ig-treated IL-6(-/-) mice. Finally, treatment of mice with etanercept, a clinically approved anti-TNF-alpha drug, confirmed the importance of controlling proinflammatory cytokines during gene therapy by adenoviral vectors.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/genetics , Genetic Therapy/methods , Interleukin-6/immunology , Transduction, Genetic/methods , Tumor Necrosis Factor-alpha/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/blood , Autoantibodies/blood , DNA, Viral/genetics , Etanercept , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hepatitis/immunology , Humans , Immunoglobulin G/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor/therapeutic use , Receptors, Tumor Necrosis Factor, Type I/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transgenes , Tumor Necrosis Factor-alpha/antagonists & inhibitors
17.
Hum Gene Ther ; 17(10): 1019-26, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17007568

ABSTRACT

Overexpression of RhoA in cancer indicates a poor prognosis, because of increased tumor cell proliferation and invasion and tumor angiogenesis. We showed previously that anti-RhoA small interfering RNA (siRNA) inhibited aggressive breast cancer more effectively than conventional blockers of Rho-mediated signaling pathways. This study reports the efficacy and lack of toxicity of intravenously administered encapsulated anti-RhoA siRNA in chitosan-coated polyisohexylcyanoacrylate (PIHCA) nanoparticles in xenografted aggressive breast cancers (MDA-MB-231). The siRNA was administered every 3 days at a dose of 150 or 1500 microg/kg body weight in nude mice. This treatment inhibited the growth of tumors by 90% in the 150-microg group and by even more in the 1500-microg group. Necrotic areas were observed in tumors from animals treated with anti-RhoA siRNA at 1500 microg/kg, resulting from angiogenesis inhibition. In addition, this therapy was found to be devoid of toxic effects, as evidenced by similarities between control and treated animals for the following parameters: body weight gain; biochemical markers of hepatic, renal, and pancreatic function; and macroscopic appearance of organs after 30 days of treatment. Because of its efficacy and the absence of toxicity, it is suggested that this strategy of anti-RhoA siRNA holds significant promise for the treatment of aggressive cancers.


Subject(s)
Breast Neoplasms/therapy , Chitosan/administration & dosage , Infusion Pumps , Neoplasm Transplantation/standards , RNA, Small Interfering/administration & dosage , rhoA GTP-Binding Protein/genetics , Animals , Breast Neoplasms/blood supply , Cell Line, Tumor , Chitosan/therapeutic use , Chitosan/toxicity , Humans , Mice , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Nanoparticles/toxicity , Neoplasm Transplantation/methods , Neoplasms/physiopathology , Neovascularization, Pathologic/therapy , RNA, Small Interfering/therapeutic use , RNA, Small Interfering/toxicity
18.
Gene Ther ; 13(23): 1619-27, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16871232

ABSTRACT

The simple injection of DNA into muscles is known to result in the expression of the injected genes, even though at low and variable levels. We report that this variability in DNA expression is partly dependent on the injection speed. The acceleration of the injection speed from values around 2 mul/s up to ones around 25 mul/s (depending on the tissue) results in a significant increase in gene expression in skeletal muscle (280 times on an average) and in liver (50 times) and a nonsignificant sevenfold increase in tumors. Heparin, which inhibits the spontaneous uptake of the injected DNA, also inhibits the increases related to the injection speed. However, at the highest injection speed, this inhibition is not total because very fast injections provoke a direct permeabilization of the cells. This "hydroporation" could be similar to the permeabilization found in the hydrodynamics method based on the fast intravascular injection of a huge volume of DNA. Neither the "hydroporation" nor the heparin-inhibitable uptake mechanism induces histologically detectable lesions. There is a limited muscle cell stress independent of the injection speed. Heterogeneity in the injection speed might thus be an explanation for the variability in DNA expression after simple injection.


Subject(s)
DNA/administration & dosage , DNA/genetics , Genetic Therapy/methods , Muscle, Skeletal/enzymology , Transfection/methods , Animals , Female , Gene Expression , Green Fluorescent Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Heparin/pharmacology , Hindlimb , Inflammation , Injections, Intravenous/methods , Liver/enzymology , Luciferases/genetics , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Time Factors
19.
Oncogene ; 25(22): 3150-9, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16501609

ABSTRACT

In vivo neuroblastoma (NB) xenograft model, resistant to the DNA-topoisomerase I inhibitor irinotecan (CPT-11), has been established to study resistance mechanisms acquired in a therapeutic setting. Common mechanisms of resistance were not involved in this resistance. Thus, we compared the gene expression profiles of sensitive, resistant, and reverted tumors using cDNA expression arrays. Expression of selected transcripts was confirmed by quantitative real-time PCR. We found that pleiotrophin (PTN), a heparin-binding growth factor, was the only gene significantly affected: PTN gene expression was downregulated in all resistant tumors (8-14-fold) as compared to sensitive tumors, and was increased (2-4-fold) in all reverted tumors as compared to resistant tumors. PTN thus appeared to be a likely candidate gene associated with resistance to CPT-11 in this in vivo model. To investigate the direct implication of PTN in NB, we transfected two NB cell lines with RNA interferences in order to silence PTN. PTN failed to demonstrate implication in resistance to CPT-11 in vitro but could influence sensitivity to CPT-11 exclusively through an in vivo mechanism. Indeed, vasculature was significantly enhanced in resistant NB xenografts compared to sensitive and reverted xenografts, and we suggest that PTN is acting in our resistant in vivo NB model as an angiostatic factor.


Subject(s)
Camptothecin/analogs & derivatives , Carrier Proteins/genetics , Cytokines/genetics , Drug Resistance, Neoplasm , Neovascularization, Pathologic/etiology , Neuroblastoma/drug therapy , Animals , Camptothecin/therapeutic use , Carrier Proteins/antagonists & inhibitors , Child, Preschool , Cytokines/antagonists & inhibitors , Gene Expression Profiling , Gene Expression Regulation , Humans , Irinotecan , Male , Mice , Neuroblastoma/blood supply , Neuroblastoma/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Topoisomerase I Inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Gene Ther ; 13(3): 235-44, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16208422

ABSTRACT

The BRCA1 (breast cancer 1) breast cancer susceptibility gene is recognized as responsible for most familial breast and ovarian cancers and is suggested to be a tissue-specific tumor suppressor gene. In this report, we investigated the tissue specificity of tumor inhibitory activities induced by a recombinant adenovirus coding for wild-type BRCA1 (wtAdBRCA1). We demonstrated a pronounced in vitro antiproliferative effect on H1299 lung and HT29 colon cells upon infection with AdBRCA1. We describe a prolonged G1 cell cycle arrest associated with a decrease in the hyperphosphorylated form of Rb, suggesting that the Rb/E2F pathway is implicated in BRCA1-induced cell growth arrest. We also observed a significant antitumor effect in these pre-established subcutaneous tumors after in situ delivery of AdBRCA1, although these two tumors do not express wt p53, and also estrogen alpha and beta, progesterone and androgen receptors. Moreover, BRCA1 can induce a strong prolonged cell cycle arrest and apoptotic cell death but no significant antiangiogenic effect in H1299 tumors. Finally, our data indicate that intratumor administration of wtAdBRCA1 significantly inhibits growth of lung and colon steroid hormone-independent tumors.


Subject(s)
Adenoviridae/genetics , Colonic Neoplasms/therapy , Genes, BRCA1 , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Lung Neoplasms/therapy , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Female , G1 Phase , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Genetic Vectors/genetics , HT29 Cells , Humans , Injections, Intralesional , Male , Mice , Neoplasm Transplantation , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL