Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38464085

ABSTRACT

Chimeric antigen receptor (CAR)-engineered T and NK cells can cause durable remission of B-cell malignancies; however, limited persistence restrains the full potential of these therapies in many patients. The FAS ligand (FAS-L)/FAS pathway governs naturally-occurring lymphocyte homeostasis, yet knowledge of which cells express FAS-L in patients and whether these sources compromise CAR persistence remains incomplete. Here, we constructed a single-cell atlas of diverse cancer types to identify cellular subsets expressing FASLG, the gene encoding FAS-L. We discovered that FASLG is limited primarily to endogenous T cells, NK cells, and CAR-T cells while tumor and stromal cells express minimal FASLG. To establish whether CAR-T/NK cell survival is regulated through FAS-L, we performed competitive fitness assays using lymphocytes modified with or without a FAS dominant negative receptor (ΔFAS). Following adoptive transfer, ΔFAS-expressing CAR-T and CAR-NK cells became enriched across multiple tissues, a phenomenon that mechanistically was reverted through FASLG knockout. By contrast, FASLG was dispensable for CAR-mediated tumor killing. In multiple models, ΔFAS co-expression by CAR-T and CAR-NK enhanced antitumor efficacy compared with CAR cells alone. Together, these findings reveal that CAR-engineered lymphocyte persistence is governed by a FAS-L/FAS auto-regulatory circuit.

2.
Cancer Res ; 82(22): 4261-4273, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36112789

ABSTRACT

Mutationally activated BRAF is detected in approximately 7% of human lung adenocarcinomas, with BRAFT1799A serving as a predictive biomarker for treatment of patients with FDA-approved inhibitors of BRAFV600E oncoprotein signaling. In genetically engineered mouse (GEM) models, expression of BRAFV600E in the lung epithelium initiates growth of benign lung tumors that, without additional genetic alterations, rarely progress to malignant lung adenocarcinoma. To identify genes that cooperate with BRAFV600E for malignant progression, we used Sleeping Beauty-mediated transposon mutagenesis, which dramatically accelerated the emergence of lethal lung cancers. Among the genes identified was Rbms3, which encodes an RNA-binding protein previously implicated as a putative tumor suppressor. Silencing of RBMS3 via CRISPR/Cas9 gene editing promoted growth of BRAFV600E lung organoids and promoted development of malignant lung cancers with a distinct micropapillary architecture in BRAFV600E and EGFRL858R GEM models. BRAFV600E/RBMS3Null lung tumors displayed elevated expression of Ctnnb1, Ccnd1, Axin2, Lgr5, and c-Myc mRNAs, suggesting that RBMS3 silencing elevates signaling through the WNT/ß-catenin signaling axis. Although RBMS3 silencing rendered BRAFV600E-driven lung tumors resistant to the effects of dabrafenib plus trametinib, the tumors were sensitive to inhibition of porcupine, an acyltransferase of WNT ligands necessary for their secretion. Analysis of The Cancer Genome Atlas patient samples revealed that chromosome 3p24, which encompasses RBMS3, is frequently lost in non-small cell lung cancer and correlates with poor prognosis. Collectively, these data reveal the role of RBMS3 as a lung cancer suppressor and suggest that RBMS3 silencing may contribute to malignant NSCLC progression. SIGNIFICANCE: Loss of RBMS3 cooperates with BRAFV600E to induce lung tumorigenesis, providing a deeper understanding of the molecular mechanisms underlying mutant BRAF-driven lung cancer and potential strategies to more effectively target this disease.


Subject(s)
Adenocarcinoma of Lung , Carcinogenesis , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Proto-Oncogene Proteins B-raf , RNA-Binding Proteins , Trans-Activators , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation , Lung/pathology , Lung Neoplasms/genetics , Mutagenesis , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/genetics , Trans-Activators/metabolism , Wnt Signaling Pathway , Carcinogenesis/genetics
3.
Mol Ther ; 30(4): 1381-1395, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35151842

ABSTRACT

T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αß T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Genetic Engineering , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
4.
Nat Rev Clin Oncol ; 17(3): 147-167, 2020 03.
Article in English | MEDLINE | ID: mdl-31848460

ABSTRACT

T cells genetically engineered to express chimeric antigen receptors (CARs) have proven - and impressive - therapeutic activity in patients with certain subtypes of B cell leukaemia or lymphoma, with promising efficacy also demonstrated in patients with multiple myeloma. Nevertheless, various barriers restrict the efficacy and/or prevent the widespread use of CAR T cell therapies in these patients as well as in those with other cancers, particularly solid tumours. Key challenges relating to CAR T cells include severe toxicities, restricted trafficking to, infiltration into and activation within tumours, suboptimal persistence in vivo, antigen escape and heterogeneity, and manufacturing issues. The evolution of CAR designs beyond the conventional structures will be necessary to address these limitations and to expand the use of CAR T cells to a wider range of malignancies. Investigators are addressing the current obstacles with a wide range of engineering strategies in order to improve the safety, efficacy and applicability of this therapeutic modality. In this Review, we discuss the innovative designs of novel CAR T cell products that are being developed to increase and expand the clinical benefits of these treatments in patients with diverse cancers.


Subject(s)
Cell- and Tissue-Based Therapy , Neoplasms/drug therapy , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Cell Engineering , Humans , Lymphoma/drug therapy , Lymphoma/immunology , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology
5.
Cancer Discov ; 5(4): 380-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637275

ABSTRACT

UNLABELLED: Regulation of mRNA splicing, a critical and tightly regulated cellular function, underlies the majority of proteomic diversity and is frequently disrupted in disease. Using an integrative genomics approach, we combined both genomic data and exon-level transcriptome data in two somatic tissues (cerebella and peripheral ganglia) from a transgenic mouse model of neuroblastoma, a tumor that arises from the peripheral neural crest. Here, we describe splicing quantitative trait loci associated with differential splicing across the genome that we use to identify genes with previously unknown functions within the splicing pathway and to define de novo intronic splicing motifs that influence splicing from hundreds of bases away. Our results show that these splicing motifs represent sites for functional recurrent mutations and highlight novel candidate genes in human cancers, including childhood neuroblastoma. SIGNIFICANCE: Somatic mutations with predictable downstream effects are largely relegated to coding regions, which comprise less than 2% of the human genome. Using an unbiased in vivo analysis of a mouse model of neuroblastoma, we have identified intronic splicing motifs that translate into sites for recurrent somatic mutations in human cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , RNA Splicing , Alternative Splicing , Animals , Cerebellum/metabolism , Disease Models, Animal , Epistasis, Genetic , Exons , Gene Regulatory Networks , Genome-Wide Association Study , Genomics , Introns , Mice , Mutation , Neuroblastoma/metabolism , Nucleotide Motifs , Quantitative Trait Loci , RNA Isoforms , Species Specificity , Superior Cervical Ganglion/metabolism
6.
Cell Rep ; 9(3): 1034-46, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25437558

ABSTRACT

The development of targeted therapeutics for neuroblastoma, the third most common tumor in children, has been limited by a poor understanding of growth signaling mechanisms unique to the peripheral nerve precursors from which tumors arise. In this study, we combined genetics with gene-expression analysis in the peripheral sympathetic nervous system to implicate arginase 1 and GABA signaling in tumor formation in vivo. In human neuroblastoma cells, either blockade of ARG1 or benzodiazepine-mediated activation of GABA-A receptors induced apoptosis and inhibited mitogenic signaling through AKT and MAPK. These results suggest that ARG1 and GABA influence both neural development and neuroblastoma and that benzodiazepines in clinical use may have potential applications for neuroblastoma therapy.


Subject(s)
Arginase/genetics , Brain Neoplasms/genetics , Molecular Targeted Therapy , Neuroblastoma/genetics , Quantitative Trait Loci/genetics , Receptors, GABA-A/genetics , Animals , Apoptosis , Arginase/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Chromosomes, Mammalian/genetics , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Humans , Mice , Survival Analysis , gamma-Aminobutyric Acid/metabolism
7.
Epigenetics ; 8(12): 1254-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24107773

ABSTRACT

Genetic and epigenetic alterations are essential for the initiation and progression of human cancer. We previously reported that primary human medulloblastomas showed extensive cancer-specific CpG island DNA hypermethylation in critical developmental pathways. To determine whether genetically engineered mouse models (GEMMs) of medulloblastoma have comparable epigenetic changes, we assessed genome-wide DNA methylation in three mouse models of medulloblastoma. In contrast to human samples, very few loci with cancer-specific DNA hypermethylation were detected, and in almost all cases the degree of methylation was relatively modest compared with the dense hypermethylation in the human cancers. To determine if this finding was common to other GEMMs, we examined a Burkitt lymphoma and breast cancer model and did not detect promoter CpG island DNA hypermethylation, suggesting that human cancers and at least some GEMMs are fundamentally different with respect to this epigenetic modification. These findings provide an opportunity to both better understand the mechanism of aberrant DNA methylation in human cancer and construct better GEMMs to serve as preclinical platforms for therapy development.


Subject(s)
CpG Islands , DNA Methylation/genetics , Neoplasms, Experimental/genetics , Neoplasms/genetics , Promoter Regions, Genetic , Animals , Breast Neoplasms/genetics , Burkitt Lymphoma/genetics , Cerebellar Neoplasms/genetics , Female , Humans , Medulloblastoma/genetics , Mice , Mice, Transgenic
8.
Genes Dev ; 26(16): 1780-96, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22855790

ABSTRACT

Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.


Subject(s)
Cell Size , Ether-A-Go-Go Potassium Channels/metabolism , Medulloblastoma/physiopathology , Mitosis , Animals , COS Cells , Cell Cycle Checkpoints/genetics , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation/genetics , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Signaling System , Medulloblastoma/mortality , Mice , Survival Analysis
9.
Cancer Cell ; 21(5): 601-613, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22624711

ABSTRACT

The proto-oncogene MYCN is mis-expressed in various types of human brain tumors. To clarify how developmental and regional differences influence transformation, we transduced wild-type or mutationally stabilized murine N-myc(T58A) into neural stem cells (NSCs) from perinatal murine cerebellum, brain stem, and forebrain. Transplantation of N-myc(WT) NSCs was insufficient for tumor formation. N-myc(T58A) cerebellar and brain stem NSCs generated medulloblastoma/primitive neuroectodermal tumors, whereas forebrain NSCs developed diffuse glioma. Expression analyses distinguished tumors generated from these different regions, with tumors from embryonic versus postnatal cerebellar NSCs demonstrating Sonic Hedgehog (SHH) dependence and SHH independence, respectively. These differences were regulated in part by the transcription factor SOX9, activated in the SHH subclass of human medulloblastoma. Our results demonstrate context-dependent transformation of NSCs in response to a common oncogenic signal.


Subject(s)
Brain Neoplasms/metabolism , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Biomarkers/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Stem/embryology , Brain Stem/metabolism , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Cerebellum/metabolism , Female , Gestational Age , Glioma/metabolism , Glioma/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Mutation , N-Myc Proto-Oncogene Protein , Neural Stem Cells/pathology , Neuroectodermal Tumors, Primitive/metabolism , Neuroectodermal Tumors, Primitive/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prosencephalon/embryology , Prosencephalon/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Spheroids, Cellular , Time Factors , Transduction, Genetic , Zinc Finger Protein Gli2
10.
Sci Transl Med ; 4(115): 115ra3, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22218692

ABSTRACT

Neuroblastoma, a tumor of peripheral neural crest origin, numbers among the most common childhood cancers. Both amplification of the proto-oncogene MYCN and increased neoangiogenesis mark high-risk disease. Because angiogenesis is regulated by phosphatidylinositol 3-kinase (PI3K), we tested a clinical PI3K inhibitor, NVP-BEZ235, in MYCN-dependent neuroblastoma. NVP-BEZ235 decreased angiogenesis and improved survival in both primary human (highly pretreated recurrent MYCN-amplified orthotopic xenograft) and transgenic mouse models for MYCN-driven neuroblastoma. Using both gain- and loss-of-function approaches, we demonstrated that the antiangiogenic efficacy of NVP-BEZ235 depended critically on MYCN in vitro and in vivo. Thus, clinical PI3K/mammalian target of rapamycin inhibitors drove degradation of MYCN in tumor cells, with secondary paracrine blockade of angiogenesis. Our data demonstrated significantly improved survival in treated animals and suggest that NVP-BEZ235 should be tested in children with high-risk, MYCN-amplified neuroblastoma.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neuroblastoma/metabolism , Nuclear Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neovascularization, Pathologic , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Mas , Quinolines/pharmacology , Signal Transduction
11.
Nat Med ; 16(10): 1134-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20871609

ABSTRACT

Inactivation of the p53 tumor suppressor pathway allows cell survival in times of stress and occurs in many human cancers; however, normal embryonic stem cells and some cancers such as neuroblastoma maintain wild-type human TP53 and mouse Trp53 (referred to collectively as p53 herein). Here we describe a miRNA, miR-380-5p, that represses p53 expression via a conserved sequence in the p53 3' untranslated region (UTR). miR-380-5p is highly expressed in mouse embryonic stem cells and neuroblastomas, and high expression correlates with poor outcome in neuroblastomas with neuroblastoma derived v-myc myelocytomatosis viral-related oncogene (MYCN) amplification. miR-380 overexpression cooperates with activated HRAS oncoprotein to transform primary cells, block oncogene-induced senescence and form tumors in mice. Conversely, inhibition of endogenous miR-380-5p in embryonic stem or neuroblastoma cells results in induction of p53, and extensive apoptotic cell death. In vivo delivery of a miR-380-5p antagonist decreases tumor size in an orthotopic mouse model of neuroblastoma. We demonstrate a new mechanism of p53 regulation in cancer and stem cells and uncover a potential therapeutic target for neuroblastoma.


Subject(s)
Gene Amplification , MicroRNAs/physiology , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , 3' Untranslated Regions , Animals , Apoptosis , Binding Sites , DNA Damage , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Oncogenes , Tumor Suppressor Protein p53/physiology
12.
Genes Dev ; 24(10): 1059-72, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20478998

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Medulloblastoma/physiopathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cell Cycle/physiology , Cellular Senescence/physiology , Cerebellum/metabolism , Down-Regulation , Gene Expression Profiling , Genomic Instability , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/pathology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics
14.
Cancer Res ; 69(21): 8429-37, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843846

ABSTRACT

The Sleeping Beauty (SB) transposon system has been used as a somatic mutagen to identify candidate cancer genes. In previous studies, efficient leukemia/lymphoma formation on an otherwise wild-type genetic background occurred in mice undergoing whole-body mobilization of transposons, but was accompanied by high levels of embryonic lethality. To explore the utility of SB for large-scale cancer gene discovery projects, we have generated mice that carry combinations of different transposon and transposase transgenes. We have identified a transposon/transposase combination that promotes highly penetrant leukemia/lymphoma formation on an otherwise wild-type genetic background, yet does not cause embryonic lethality. Infiltrating gliomas also occurred at lower penetrance in these mice. SB-induced or accelerated tumors do not harbor large numbers of chromosomal amplifications or deletions, indicating that transposon mobilization likely promotes tumor formation by insertional mutagenesis of cancer genes, and not by promoting wide-scale genomic instability. Cloning of transposon insertions from lymphomas/leukemias identified common insertion sites at known and candidate novel cancer genes. These data indicate that a high mutagenesis rate can be achieved using SB without high levels of embryonic lethality or genomic instability. Furthermore, the SB system could be used to identify new genes involved in lymphomagenesis/leukemogenesis.


Subject(s)
DNA Transposable Elements/genetics , Embryo, Mammalian/cytology , Genes, Lethal , Glioma/genetics , Leukemia/genetics , Lymphoma/genetics , Transposases/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Comparative Genomic Hybridization , Embryo, Mammalian/metabolism , Flow Cytometry , Gene Expression Profiling , Genomic Instability , Glioma/pathology , Immunoenzyme Techniques , Leukemia/pathology , Lymphoma/pathology , Mice , Mice, Transgenic , Mutagenesis , Oligonucleotide Array Sequence Analysis , Survival Rate
15.
Genome Biol ; 8 Suppl 1: S12, 2007.
Article in English | MEDLINE | ID: mdl-18047689

ABSTRACT

Viral and transposon vectors have been employed in gene therapy as well as functional genomics studies. However, the goals of gene therapy and functional genomics are entirely different; gene therapists hope to avoid altering endogenous gene expression (especially the activation of oncogenes), whereas geneticists do want to alter expression of chromosomal genes. The odds of either outcome depend on a vector's preference to integrate into genes or control regions, and these preferences vary between vectors. Here we discuss the relative strengths of DNA vectors over viral vectors, and review methods to overcome barriers to delivery inherent to DNA vectors. We also review the tendencies of several classes of retroviral and transposon vectors to target DNA sequences, genes, and genetic elements with respect to the balance between insertion preferences and oncogenic selection. Theoretically, knowing the variables that affect integration for various vectors will allow researchers to choose the vector with the most utility for their specific purposes. The three principle benefits from elucidating factors that affect preferences in integration are as follows: in gene therapy, it allows assessment of the overall risks for activating an oncogene or inactivating a tumor suppressor gene that could lead to severe adverse effects years after treatment; in genomic studies, it allows one to discern random from selected integration events; and in gene therapy as well as functional genomics, it facilitates design of vectors that are better targeted to specific sequences, which would be a significant advance in the art of transgenesis.


Subject(s)
DNA/genetics , Genetic Therapy , Genetic Vectors/genetics , Genomics , Mutagenesis, Insertional , Animals , Base Sequence , DNA/metabolism , Genetic Therapy/adverse effects , Genome , Humans
16.
Cancer Res ; 67(17): 7960-5, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804702

ABSTRACT

We have shown previously that blockade of epidermal growth factor receptor (EGFR) cooperates with a pan-selective inhibitor of phosphoinositide-3-kinase (PI3K) in EGFR-driven glioma. In this communication, we tested EGFR-driven glioma differing in PTEN status, treating with the EGFR inhibitor erlotinib and a novel dual inhibitor of PI3Kalpha and mTOR (PI-103). Erlotinib blocked proliferation only in PTEN(wt) cells expressing EGFR. Although erlotinib monotherapy showed little effect in PTEN(mt) glioma, PI-103 greatly augmented the antiproliferative efficacy of erlotinib in this setting. To address the importance of PI3K blockade, we showed in PTEN(mt) glioma that combining PI-103 and erlotinib was superior to either monotherapy or to therapy combining erlotinib with either rapamycin (an inhibitor of mTOR) or PIK-90 (an inhibitor of PI3Kalpha). These experiments show that a dual inhibitor of PI3Kalpha and mTOR augments the activity of EGFR blockade, offering a mechanistic rationale for targeting EGFR, PI3Kalpha, and mTOR in the treatment of EGFR-driven, PTEN-mutant glioma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , ErbB Receptors/antagonists & inhibitors , Furans/pharmacology , Glioma/drug therapy , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Cell Cycle/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Erlotinib Hydrochloride , Furans/administration & dosage , Glioma/genetics , Glioma/pathology , Humans , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Quinazolines/administration & dosage , TOR Serine-Threonine Kinases , Treatment Outcome , Tumor Cells, Cultured
17.
Eur J Cancer ; 43(9): 1467-75, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17449239

ABSTRACT

Overexpression of the human MYCN oncogene driven by a tyrosine hydroxylase promoter causes tumours in transgenic mice that recapitulate the childhood cancer neuroblastoma. To establish an in vitro model to study this process, a series of isogenic cell lines were developed from these MYCN-driven murine tumours. Lines were established from tumours arising in homozygous and hemizygous MYCN transgenic mice. Hemizygous tumours gave rise to cell lines growing only in suspension. Homozygous tumours gave rise to similar suspension lines as well as morphologically distinct substrate-adherent lines characteristic of human S-type neuroblastoma cells. FISH analysis demonstrated selective MYCN transgene amplification in cell lines derived from hemizygous mice. Comparative genomic hybridisation (CGH) and fluorescence in situ hybridisation (FISH) analysis confirmed a range of neuroblastoma-associated genetic changes in the various lines, in particular, gain of regions syntenic with human 17q. These isogenic lines together with the transgenic mice thus represent valuable models for investigating the biological characteristics of aggressive neuroblastoma.


Subject(s)
Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Chromosome Aberrations , Gene Expression , Heterozygote , Homozygote , Humans , Karyotyping , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Ploidies
18.
Nucleic Acids Res ; 34(9): 2803-11, 2006.
Article in English | MEDLINE | ID: mdl-16717285

ABSTRACT

Mobile genetic elements with the ability to integrate genetic information into chromosomes can cause disease over short periods of time and shape genomes over eons. These elements can be used for functional genomics, gene transfer and human gene therapy. However, their integration-site preferences, which are critically important for these uses, are poorly understood. We analyzed the insertion sites of several transposons and retroviruses to detect patterns of integration that might be useful for prediction of preferred integration sites. Initially we found that a mathematical description of DNA-deformability, called V(step), could be used to distinguish preferential integration sites for Sleeping Beauty (SB) transposons into a particular 100 bp region of a plasmid [G. Liu, A. M. Geurts, K. Yae, A. R. Srinivassan, S. C. Fahrenkrug, D. A. Largaespada,J. Takeda, K. Horie, W. K. Olson and P. B. Hackett (2005) J. Mol. Biol., 346, 161-173 ]. Based on these findings, we extended our examination of integration of SB transposons into whole plasmids and chromosomal DNA. To accommodate sequences up to 3 Mb for these analyses, we developed an automated method, ProTIS, that can generate profiles of predicted integration events. However, a similar approach did not reveal any structural pattern of DNA that could be used to predict favored integration sites for other transposons as well as retroviruses and lentiviruses due to a limitation of available data sets. Nonetheless, ProTIS has the utility for predicting likely SB transposon integration sites in investigator-selected regions of genomes and our general strategy may be useful for other mobile elements once a sufficiently high density of sites in a single region are obtained. ProTIS analysis can be useful for functional genomic, gene transfer and human gene therapy applications using the SB system.


Subject(s)
Algorithms , Chromosomes, Mammalian/chemistry , DNA Transposable Elements , Genomics/methods , Animals , Gene Targeting , Genetic Vectors , Introns , Mice , Proto-Oncogene Proteins B-raf/genetics , Retroviridae/genetics
19.
Cancer Res ; 63(17): 5266-73, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500357

ABSTRACT

Neuroblastoma, the third most common tumor of childhood, is a complex disease in which few genetic mutations have been identified.Mice expressing a human MYCN oncogene driven by the rat tyrosine hydroxylase promoter (TH-MYCN) represent an animal model for this disorder. We performed microarray-based comparative genomic hybridization analysis on murine tumors, identifying gains on chromosomes 1, 3, 11, 14, 17, and 18 and losses on chromosomes 5, 9, and 16. Fluorescence in situ hybridization analysis confirmed an amplicon on chromosome 18 as the site of TH-MYCN transgene integration. Selected tumors with localized gains of chromosome 11 delineate a 15-Mb region orthologous to human chromosome 17q and help to narrow the minimal region gained in human tumors. We observed clustered loss of chromosomes 5, 9, and 16, orthologous to a similar pattern of combined loss of chromosomes 3p, 4p, and 11q in human tumors. These data demonstrate conservation of many genetic changes in murine and human neuroblastoma and suggest that further delineation of genetic abnormalities in murine tumors may identify genes important in human disease.


Subject(s)
Chromosome Aberrations , Neuroblastoma/genetics , Allelic Imbalance , Animals , Cluster Analysis , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Nuclear Proteins/genetics , Nucleic Acid Hybridization , Oncogene Proteins/genetics , Tyrosine 3-Monooxygenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...