Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 171(6): 1284-1300.e21, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195073

ABSTRACT

Combining DNA-demethylating agents (DNA methyltransferase inhibitors [DNMTis]) with histone deacetylase inhibitors (HDACis) holds promise for enhancing cancer immune therapy. Herein, pharmacologic and isoform specificity of HDACis are investigated to guide their addition to a DNMTi, thus devising a new, low-dose, sequential regimen that imparts a robust anti-tumor effect for non-small-cell lung cancer (NSCLC). Using in-vitro-treated NSCLC cell lines, we elucidate an interferon α/ß-based transcriptional program with accompanying upregulation of antigen presentation machinery, mediated in part through double-stranded RNA (dsRNA) induction. This is accompanied by suppression of MYC signaling and an increase in the T cell chemoattractant CCL5. Use of this combination treatment schema in mouse models of NSCLC reverses tumor immune evasion and modulates T cell exhaustion state towards memory and effector T cell phenotypes. Key correlative science metrics emerge for an upcoming clinical trial, testing enhancement of immune checkpoint therapy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Drug Therapy, Combination , Lung Neoplasms/therapy , Tumor Escape/drug effects , Animals , Antigen Presentation/drug effects , Antineoplastic Agents/therapeutic use , Azacitidine/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , T-Lymphocytes/immunology , Transcriptome , Tumor Microenvironment
2.
Nature ; 579(7798): 284-290, 2020 03.
Article in English | MEDLINE | ID: mdl-32103175

ABSTRACT

Cancer recurrence after surgery remains an unresolved clinical problem1-3. Myeloid cells derived from bone marrow contribute to the formation of the premetastatic microenvironment, which is required for disseminating tumour cells to engraft distant sites4-6. There are currently no effective interventions that prevent the formation of the premetastatic microenvironment6,7. Here we show that, after surgical removal of primary lung, breast and oesophageal cancers, low-dose adjuvant epigenetic therapy disrupts the premetastatic microenvironment and inhibits both the formation and growth of lung metastases through its selective effect on myeloid-derived suppressor cells (MDSCs). In mouse models of pulmonary metastases, MDSCs are key factors in the formation of the premetastatic microenvironment after resection of primary tumours. Adjuvant epigenetic therapy that uses low-dose DNA methyltransferase and histone deacetylase inhibitors, 5-azacytidine and entinostat, disrupts the premetastatic niche by inhibiting the trafficking of MDSCs through the downregulation of CCR2 and CXCR2, and by promoting MDSC differentiation into a more-interstitial macrophage-like phenotype. A decreased accumulation of MDSCs in the premetastatic lung produces longer periods of disease-free survival and increased overall survival, compared with chemotherapy. Our data demonstrate that, even after removal of the primary tumour, MDSCs contribute to the development of premetastatic niches and settlement of residual tumour cells. A combination of low-dose adjuvant epigenetic modifiers that disrupts this premetastatic microenvironment and inhibits metastases may permit an adjuvant approach to cancer therapy.


Subject(s)
Epigenesis, Genetic , Genetic Therapy , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/therapy , Tumor Microenvironment , Animals , Azacitidine/pharmacology , Benzamides/pharmacology , Cell Differentiation , Cell Movement/drug effects , Chemotherapy, Adjuvant , Disease Models, Animal , Down-Regulation/drug effects , Mice , Myeloid-Derived Suppressor Cells/cytology , Neoplasm Metastasis/therapy , Neoplasms/surgery , Pyridines/pharmacology , Receptors, CCR2/genetics , Receptors, Interleukin-8B/genetics , Tumor Microenvironment/drug effects
3.
Mol Cell ; 65(2): 323-335, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28107650

ABSTRACT

TET proteins, by converting 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), are hypothesized, but not directly shown, to protect promoter CpG islands (CGIs) against abnormal DNA methylation (DNAm) in cancer. We define such a protective role linked to DNA damage from oxidative stress (OS) known to induce this abnormality. TET2 removes aberrant DNAm during OS through interacting with DNA methyltransferases (DNMTs) in a "Yin-Yang" complex targeted to chromatin and enhanced by p300 mediated TET2 acetylation. Abnormal gains of DNAm and 5hmC occur simultaneously in OS, and knocking down TET2 dynamically alters this balance by enhancing 5mC and reducing 5hmC. TET2 reduction results in hypermethylation of promoter CGIs and enhancers in loci largely overlapping with those induced by OS. Thus, TET2 indeed may protect against abnormal, cancer DNAm in a manner linked to DNA damage.


Subject(s)
Chromatin/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Oxidative Stress , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Acetylation , Chromatin/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Dioxygenases , E1A-Associated p300 Protein/metabolism , HCT116 Cells , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Neoplasms/genetics , Protein Binding , Protein Stability , Proto-Oncogene Proteins/genetics , RNA Interference , Time Factors , Transfection , Ubiquitination
4.
Proc Natl Acad Sci U S A ; 117(30): 17785-17795, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32651270

ABSTRACT

Poly(ADP ribose) polymerase inhibitors (PARPi) have efficacy in triple negative breast (TNBC) and ovarian cancers (OCs) harboring BRCA mutations, generating homologous recombination deficiencies (HRDs). DNA methyltransferase inhibitors (DNMTi) increase PARP trapping and reprogram the DNA damage response to generate HRD, sensitizing BRCA-proficient cancers to PARPi. We now define the mechanisms through which HRD is induced in BRCA-proficient TNBC and OC. DNMTi in combination with PARPi up-regulate broad innate immune and inflammasome-like signaling events, driven in part by stimulator of interferon genes (STING), to unexpectedly directly generate HRD. This inverse relationship between inflammation and DNA repair is critical, not only for the induced phenotype, but also appears as a widespread occurrence in The Cancer Genome Atlas datasets and cancer subtypes. These discerned interactions between inflammation signaling and DNA repair mechanisms now elucidate how epigenetic therapy enhances PARPi efficacy in the setting of BRCA-proficient cancer. This paradigm will be tested in a phase I/II TNBC clinical trial.


Subject(s)
Homologous Recombination/drug effects , Immunity, Innate/drug effects , Signal Transduction/drug effects , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cell Line, Tumor , Computational Biology , DNA Modification Methylases/antagonists & inhibitors , DNA Repair/drug effects , Fanconi Anemia/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Interferons/metabolism , Membrane Proteins/metabolism , Models, Biological , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism
5.
Genome Res ; 27(4): 533-544, 2017 04.
Article in English | MEDLINE | ID: mdl-28232479

ABSTRACT

Reversing DNA methylation abnormalities and associated gene silencing, through inhibiting DNA methyltransferases (DNMTs) is an important potential cancer therapy paradigm. Maximizing this potential requires defining precisely how these enzymes maintain genome-wide, cancer-specific DNA methylation. To date, there is incomplete understanding of precisely how the three DNMTs, 1, 3A, and 3B, interact for maintaining DNA methylation abnormalities in cancer. By combining genetic and shRNA depletion strategies, we define not only a dominant role for DNA methyltransferase 1 (DNMT1) but also distinct roles of 3A and 3B in genome-wide DNA methylation maintenance. Lowering DNMT1 below a threshold level is required for maximal loss of DNA methylation at all genomic regions, including gene body and enhancer regions, and for maximally reversing abnormal promoter DNA hypermethylation and associated gene silencing to reexpress key genes. It is difficult to reach this threshold with patient-tolerable doses of current DNMT inhibitors (DNMTIs). We show that new approaches, like decreasing the DNMT targeting protein, UHRF1, can augment the DNA demethylation capacities of existing DNA methylation inhibitors for fully realizing their therapeutic potential.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Genome, Human , HCT116 Cells , Humans , Promoter Regions, Genetic , Ubiquitin-Protein Ligases
6.
Nat Commun ; 15(1): 1384, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360902

ABSTRACT

Cancers of the same tissue-type but in anatomically distinct locations exhibit different molecular dependencies for tumorigenesis. Proximal and distal colon cancers exemplify such characteristics, with BRAFV600E predominantly occurring in proximal colon cancers along with increased DNA methylation phenotype. Using mouse colon organoids, here we show that proximal and distal colon stem cells have distinct transcriptional programs that regulate stemness and differentiation. We identify that the homeobox transcription factor, CDX2, which is silenced by DNA methylation in proximal colon cancers, is a key mediator of the differential transcriptional programs. Cdx2-mediated proximal colon-specific transcriptional program concurrently is tumor suppressive, and Cdx2 loss sufficiently creates permissive state for BRAFV600E-driven transformation. Human proximal colon cancers with CDX2 downregulation showed similar transcriptional program as in mouse proximal organoids with Cdx2 loss. Developmental transcription factors, such as CDX2, are thus critical in maintaining tissue-location specific transcriptional programs that create tissue-type origin specific dependencies for tumor development.


Subject(s)
Colonic Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Mice , Animals , Proto-Oncogene Proteins B-raf/genetics , CDX2 Transcription Factor/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins , Transcription Factors/genetics , Homeodomain Proteins/genetics
7.
Nat Genet ; 36(6): 582-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15156141

ABSTRACT

The role of the primary mammalian DNA methyltransferase, DNMT1, in maintaining CpG island methylation in human colon cancer cells has recently been questioned. This controversy has arisen from discrepancies between genetic knockout and RNA interference-mediated knockdown studies. Here, we re-examined the RNA interference-based approach and found that hypermethylation of single-copy genes is maintained in cells transiently and stably depleted of DNMT1.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Base Sequence , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Silencing , Genes, Tumor Suppressor , HCT116 Cells , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics
8.
Cancer Res ; 66(2): 682-92, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16423997

ABSTRACT

A major obstacle toward understanding how patterns of abnormal mammalian cytosine DNA methylation are established is the difficulty in quantitating the de novo methylation activities of DNA methyltransferases (DNMT) thought to catalyze these reactions. Here, we describe a novel method, using native human CpG island substrates from genes that frequently become hypermethylated in cancer, which generates robust activity for measuring de novo CpG methylation. We then survey colon cancer cells with genetically engineered deficiencies in different DNMTs and find that the major activity against these substrates in extracts of these cells is DNMT1, with minor contribution from DNMT 3b and none from DNMT3a, the only known bona fide de novo methyltransferases. The activity of DNMT1 against unmethylated CpG rich DNA was further tested by introducing CpG island substrates and DNMT1 into Drosophila melanogaster cells. The exogenous DNMT1 methylates the integrated mammalian CpG islands but not the Drosophila DNA. Additionally, in human cancer cells lacking DNMT1 and DNMT3b and having nearly absent genomic methylation, gene-specific de novo methylation can be initiated by reintroduction of DNMT1. Our studies provide a new assay for de novo activity of DNMTs and data suggesting a potential role for DNMT1 in the initiation of promoter CpG island hypermethylation in human cancer cells.


Subject(s)
Colonic Neoplasms/genetics , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Animals , Colonic Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1 , Drosophila/genetics , Genetic Engineering , Humans , Tumor Cells, Cultured
9.
Oncotarget ; 5(3): 587-98, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24583822

ABSTRACT

Epigenetic therapy is emerging as a potential therapy for solid tumors. To investigate its mechanism of action, we performed integrative expression and methylation analysis of 63 cancer cell lines (breast, colorectal, and ovarian) after treatment with the DNA methyltransferase inhibitor 5-azacitidine (AZA). Gene Set Enrichment Analysis demonstrated significant enrichment for immunomodulatory pathways in all three cancers (14.4-31.3%) including interferon signaling, antigen processing and presentation, and cytokines/chemokines. Strong upregulation of cancer testis antigens was also observed. An AZA IMmune gene set (AIMs) derived from the union of these immunomodulatory pathway genes classified primary tumors from all three types, into "high" and "low" AIM gene expression subsets in tumor expression data from both TCGA and GEO. Samples from selected patient biopsies showed upregulation of AIM genes after treatment with epigenetic therapy. These results point to a broad immune stimulatory role for DNA demethylating drugs in multiple cancers.


Subject(s)
Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Epigenomics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/enzymology , Neoplasms/genetics
10.
Cancer Cell ; 21(3): 430-46, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22439938

ABSTRACT

Reversal of promoter DNA hypermethylation and associated gene silencing is an attractive cancer therapy approach. The DNA methylation inhibitors decitabine and azacitidine are efficacious for hematological neoplasms at lower, less toxic, doses. Experimentally, high doses induce rapid DNA damage and cytotoxicity, which do not explain the prolonged time to response observed in patients. We show that transient exposure of cultured and primary leukemic and epithelial tumor cells to clinically relevant nanomolar doses, without causing immediate cytotoxicity, produce an antitumor "memory" response, including inhibition of subpopulations of cancer stem-like cells. These effects are accompanied by sustained decreases in genomewide promoter DNA methylation, gene reexpression, and antitumor changes in key cellular regulatory pathways. Low-dose decitabine and azacitidine may have broad applicability for cancer management.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Leukemia/genetics , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Damage , Decitabine , Gene Silencing , Humans , Leukemia/drug therapy , Leukemia/pathology , Mice , Molecular Sequence Data , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Signal Transduction , Tumor Cells, Cultured
11.
Cell Cycle ; 3(8): 1024-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15280656

ABSTRACT

The role of DNA methyltransferase 1, DNMT1, in human cancer cells has recently been contested. In this setting, DNMT1's function as the sole maintenance methyltransferase was based on the assumption that its biological activity is identical to the mouse homologue. However, the application of recent technological advances, including gene targeting and siRNA mediated ablation studies, has cast doubt on this presumed role. Here, we attempt to shed light on these new data within the context of previous experiments.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , Humans , Mammals , Neoplasms/enzymology
12.
Nature ; 416(6880): 552-6, 2002 Apr 04.
Article in English | MEDLINE | ID: mdl-11932749

ABSTRACT

Inactivation of tumour suppressor genes is central to the development of all common forms of human cancer. This inactivation often results from epigenetic silencing associated with hypermethylation rather than intragenic mutations. In human cells, the mechanisms underlying locus-specific or global methylation patterns remain unclear. The prototypic DNA methyltransferase, Dnmt1, accounts for most methylation in mouse cells, but human cancer cells lacking DNMT1 retain significant genomic methylation and associated gene silencing. We disrupted the human DNMT3b gene in a colorectal cancer cell line. This deletion reduced global DNA methylation by less than 3%. Surprisingly, however, genetic disruption of both DNMT1 and DNMT3b nearly eliminated methyltransferase activity, and reduced genomic DNA methylation by greater than 95%. These marked changes resulted in demethylation of repeated sequences, loss of insulin-like growth factor II (IGF2) imprinting, abrogation of silencing of the tumour suppressor gene p16INK4a, and growth suppression. Here we demonstrate that two enzymes cooperatively maintain DNA methylation and gene silencing in human cancer cells, and provide compelling evidence that such methylation is essential for optimal neoplastic proliferation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Neoplasms/genetics , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , DNA, Neoplasm/metabolism , Gene Targeting , Humans , Insulin-Like Growth Factor II/genetics , Molecular Sequence Data , Neoplasms/enzymology , Promoter Regions, Genetic , Tissue Inhibitor of Metalloproteinase-3/genetics , Tumor Cells, Cultured , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL