Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nucleic Acids Res ; 52(10): 5698-5719, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38587186

ABSTRACT

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Subject(s)
Chromatin , DNA Repair , DNA-Binding Proteins , Immunity , Transcription Factors , Humans , Cell Line, Tumor , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Homologous Recombination/genetics , Immunity/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/immunology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Trans-Activators , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Int J Cancer ; 154(12): 2106-2120, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38353495

ABSTRACT

Mutations in histone H3.3-encoding genes causing mutant histone tails are associated with specific cancers such as pediatric glioblastomas (H3.3-G34R/V) and giant cell tumor of the bone (H3.3-G34W). The mechanisms by which these mutations promote malignancy are not completely understood. Here we show that cells expressing H3.3-G34W exhibit DNA double-strand breaks (DSBs) repair defects and increased cellular sensitivity to ionizing radiation (IR). Mechanistically, H3.3-G34W can be deposited to damaged chromatin, but in contrast to wild-type H3.3, does not interact with non-homologous end-joining (NHEJ) key effectors KU70/80 and XRCC4 leading to NHEJ deficiency. Together with defective cell cycle checkpoints reported previously, this DNA repair deficiency in H3.3-G34W cells led to accumulation of micronuclei and cytosolic DNA following IR, which subsequently led to activation of the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) pathway, thereby inducing release of immune-stimulatory cytokines. These findings suggest a potential for radiotherapy for tumors expressing H3.3-G34W, which can be further improved by combination with STING agonists to induce immune-mediated therapeutic efficacy.


Subject(s)
DNA Repair-Deficiency Disorders , Histones , Child , Humans , Histones/genetics , Nucleotidyltransferases/genetics , Immunity , DNA
3.
Int J Cancer ; 151(2): 275-286, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35239184

ABSTRACT

Radiotherapy can induce various adverse effects including fibrosis in cancer patients. Radiation-induced aberrant expression of profibrotic genes has been associated with dysregulated epigenetic mechanisms. Pan-BET (bromodomain and extraterminal domain) inhibitors, such as JQ1 and I-BET151, have been reported to attenuate the profibrotic response after irradiation. Despite their profound preclinical efficacy, the clinical utility of pan-inhibitors is limited due to observed cytotoxicicities. Recently, inhibitors were developed that selectively target the first (BD1) and second (BD2) bromodomain of the BET proteins (iBET-BD1 [GSK778] and iBET-BD2 [GSK046]). Here, their potential to attenuate radiation-induced fibroblast activation with low-toxicity was investigated. Our results indicated that cell proliferation and cell cycle progression in fibroblasts from BJ cells and six donors were reduced when treated with I-BET151 and iBET-BD1, but not with iBET-BD2. After irradiation, induction of DGKA and profibrotic markers, especially COL1A1 and ACTA2, was attenuated with all BET inhibitors. H3K27ac enrichment was similar at the DGKA enhancer region after I-BET151 treatment and irradiation, but was reduced at the COL1A1 transcription start site and the ACTA2 enhancer site. iBET-BD2 did not change H3K27ac levels in these regions. BRD4 occupancy at these regions was not altered by any of the compounds. Cell migration activity was measured as a characteristic independent of extracellular matrix production and was unchanged in fibroblasts after irradiation and BET inhibitor-treatment. In conclusion, iBET-BD2 efficiently suppressed radiation-induced expression of DGKA and profibrotic markers without showing cytotoxicity. Thus BD2-selective targeting is a promising new therapeutic avenue for further investigations to prevent or attenuate radiotherapy-induced fibrosis.


Subject(s)
Antineoplastic Agents , Nuclear Proteins , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Nuclear Proteins/metabolism , Protein Domains , Transcription Factors/metabolism
4.
Nucleic Acids Res ; 49(20): 11666-11689, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718742

ABSTRACT

The inhibitor of DNA-binding 3 (ID3) is a transcriptional regulator that limits interaction of basic helix-loop-helix transcription factors with their target DNA sequences. We previously reported that ID3 loss is associated with mutational signatures linked to DNA repair defects. Here we demonstrate that ID3 exhibits a dual role to promote DNA double-strand break (DSB) repair, particularly homologous recombination (HR). ID3 interacts with the MRN complex and RECQL helicase to activate DSB repair and it facilitates RAD51 loading and downstream steps of HR. In addition, ID3 promotes the expression of HR genes in response to ionizing radiation by regulating both chromatin accessibility and activity of the transcription factor E2F1. Consistently, analyses of TCGA cancer patient data demonstrate that low ID3 expression is associated with impaired HR. The loss of ID3 leads to sensitivity of tumor cells to PARP inhibition, offering new therapeutic opportunities in ID3-deficient tumors.


Subject(s)
Homologous Recombination , Inhibitor of Differentiation Proteins/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , Drug Resistance, Neoplasm , E2F1 Transcription Factor/metabolism , HEK293 Cells , Humans , Inhibitor of Differentiation Proteins/chemistry , Male , Neoplasm Proteins/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism
5.
Cancers (Basel) ; 13(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070078

ABSTRACT

Radiotherapy, a common component in cancer treatment, can induce adverse effects including fibrosis in co-irradiated tissues. We previously showed that differential DNA methylation at an enhancer of diacylglycerol kinase alpha (DGKA) in normal dermal fibroblasts is associated with radiation-induced fibrosis. After irradiation, the transcription factor EGR1 is induced and binds to the hypomethylated enhancer, leading to increased DGKA and pro-fibrotic marker expression. We now modulated this DGKA induction by targeted epigenomic and genomic editing of the DGKA enhancer and administering epigenetic drugs. Targeted DNA demethylation of the DGKA enhancer in HEK293T cells resulted in enrichment of enhancer-related histone activation marks and radiation-induced DGKA expression. Mutations of the EGR1-binding motifs decreased radiation-induced DGKA expression in BJ fibroblasts and caused dysregulation of multiple fibrosis-related pathways. EZH2 inhibitors (GSK126, EPZ6438) did not change radiation-induced DGKA increase. Bromodomain inhibitors (CBP30, JQ1) suppressed radiation-induced DGKA and pro-fibrotic marker expression. Similar drug effects were observed in donor-derived fibroblasts with low DNA methylation. Overall, epigenomic manipulation of DGKA expression may offer novel options for a personalized treatment to prevent or attenuate radiotherapy-induced fibrosis.

6.
Hepatology ; 73(6): 2293-2310, 2021 06.
Article in English | MEDLINE | ID: mdl-33020926

ABSTRACT

BACKGROUND AND AIMS: Gallbladder cancer (GBC) is a highly aggressive malignancy of the biliary tract. Most cases of GBC are diagnosed in low-income and middle-income countries, and research into this disease has long been limited. In this study we therefore investigate the epigenetic changes along the model of GBC carcinogenesis represented by the sequence gallstone disease → dysplasia → GBC in Chile, the country with the highest incidence of GBC worldwide. APPROACH AND RESULTS: To perform epigenome-wide methylation profiling, genomic DNA extracted from sections of formalin-fixed, paraffin-embedded gallbladder tissue was analyzed using Illumina Infinium MethylationEPIC BeadChips. Preprocessed, quality-controlled data from 82 samples (gallstones n = 32, low-grade dysplasia n = 13, high-grade dysplasia n = 9, GBC n = 28) were available to identify differentially methylated markers, regions, and pathways as well as changes in copy number variations (CNVs). The number and magnitude of epigenetic changes increased with disease development and predominantly involved the hypermethylation of cytosine-guanine dinucleotide islands and gene promoter regions. The methylation of genes implicated in Wnt signaling, Hedgehog signaling, and tumor suppression increased with tumor grade. CNVs also increased with GBC development and affected cyclin-dependent kinase inhibitor 2A, MDM2 proto-oncogene, tumor protein P53, and cyclin D1 genes. Gains in the targetable Erb-B2 receptor tyrosine kinase 2 gene were detected in 14% of GBC samples. CONCLUSIONS: Our results indicate that GBC carcinogenesis comprises three main methylation stages: early (gallstone disease and low-grade dysplasia), intermediate (high-grade dysplasia), and late (GBC). The identified gradual changes in methylation and CNVs may help to enhance our understanding of the mechanisms underlying this aggressive disease and eventually lead to improved treatment and early diagnosis of GBC.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gallbladder Neoplasms/genetics , Gallstones/genetics , Hyperplasia/genetics , Carcinogenesis , Cell Line, Tumor , DNA Copy Number Variations , Female , Genes, Neoplasm/genetics , Humans , Male
7.
Front Oncol ; 10: 737, 2020.
Article in English | MEDLINE | ID: mdl-32477950

ABSTRACT

Radiotherapy is an efficient tool in cancer treatment, but it brings along the risk of side effects such as fibrosis in the irradiated healthy tissue thus limiting tumor control and impairing quality of life of cancer survivors. Knowledge on radiation-related fibrosis risk and therapeutic options is still limited and requires further research. Recent studies demonstrated that epigenetic regulation of diacylglycerol kinase alpha (DGKA) is associated with radiation-induced fibrosis. However, the specific mechanisms are still unknown. In this review, we scrutinized the role of DGKA in the radiation response and in further cellular functions to show the potential of DGKA as a predictive marker or a novel target in fibrosis treatment. DGKA was reported to participate in immune response, lipid signaling, exosome production, and migration as well as cell proliferation, all processes which are suggested to be critical steps in fibrogenesis. Most of these functions are based on the conversion of diacylglycerol (DAG) to phosphatidic acid (PA) at plasma membranes, but DGKA might have also other, yet not well-known functions in the nucleus. Current evidence summarized here underlines that DGKA activation may play a central role in fibrosis formation post-irradiation and shows a potential of direct DGKA inhibitors or epigenetic modulators to attenuate pro-fibrotic reactions, thus providing novel therapeutic choices.

8.
Epigenomics ; 11(16): 1765-1778, 2019 12.
Article in English | MEDLINE | ID: mdl-31755748

ABSTRACT

Aim: Use of menopausal hormone therapy (MHT) has been associated with a reduced risk for colorectal cancer, but mechanisms underlying this relationship are not well understood. In the colon, MHT appears to act through estrogen receptor ß (ERß) which may influence DNA methylation by binding to DNA. Using genome-wide methylation profiling data, we aimed to identify genes that may be differentially methylated according to MHT use. Materials & methods: DNA methylation was measured using Illumina HumanMethylation450k arrays in two independent tumor sample sets of colorectal cancer patients. Differential methylation was determined using R/limma. Results: In the discovery analysis, two CpG sites showed differential DNA methylation according to MHT use, both were not replicated. In stratified analyses, 342 CpG sites were associated with current MHT use only in ERß-positive tumors. Conclusion: The suggestive findings of differential methylation according to current MHT use in ERß-positive tumors warrant further investigation.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Estrogen Replacement Therapy , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , CpG Islands , Estrogen Receptor beta/metabolism , Female , Humans , Menopause , Middle Aged
9.
Clin Epigenetics ; 11(1): 67, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053176

ABSTRACT

BACKGROUND: Genetic aberrations in DNA repair genes are linked to cancer, but less is reported about epigenetic regulation of DNA repair and functional consequences. We investigated the intragenic methylation loss at the three prime repair exonuclease 2 (TREX2) locus in laryngeal (n = 256) and colorectal cancer cases (n = 95) and in pan-cancer data from The Cancer Genome Atlas (TCGA). RESULTS: Significant methylation loss at an intragenic site of TREX2 was a frequent trait in both patient cohorts (p = 0.016 and < 0.001, respectively) and in 15 out of 22 TCGA studies. Methylation loss correlated with immunohistochemically staining for TREX2 (p < 0.0001) in laryngeal tumors and improved overall survival of laryngeal cancer patients (p = 0.045). Chromatin immunoprecipitation, demethylation experiments, and reporter gene assays revealed that the region of methylation loss can function as a CCAAT/enhancer binding protein alpha (CEBPA)-responsive enhancer element regulating TREX2 expression. CONCLUSIONS: The data highlight a regulatory role of TREX2 DNA methylation for gene expression which might affect incidence and survival of laryngeal cancer. Altered TREX2 protein levels in tumors may affect drug-induced DNA damage repair and provide new tailored therapies.


Subject(s)
DNA Methylation , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Laryngeal Neoplasms/mortality , Phosphoproteins/genetics , Phosphoproteins/metabolism , Up-Regulation , Aged , Cell Line, Tumor , DNA Repair , Epigenesis, Genetic , Exodeoxyribonucleases/chemistry , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/metabolism , Male , Middle Aged , Phosphoproteins/chemistry , Protein Domains , Survival Analysis
10.
Epigenetics ; 14(5): 477-493, 2019 05.
Article in English | MEDLINE | ID: mdl-30931802

ABSTRACT

Involvement of sex hormones in colorectal cancer (CRC) development has been linked to oestrogen receptor ß (ERß). Expression of ERß is found reduced in tumour tissue and inversely related to mortality. However, mechanisms are not well understood. Our study aimed to detect differentially methylated genes associated with ERß expression, which could point to mechanisms by which ERß could influence risk and prognosis of CRC. Epigenome-wide DNA methylation profiling was performed using Illumina HumanMethylation450k BeadChip arrays in two independent tumour sample sets of CRC patients recruited in 2003-2010 by the German DACHS study (discovery cohort n = 917, replication cohort n = 907). ERß expression was measured using immunohistochemistry and scored as negative, moderate and high. Differentially methylated CpG sites and genomic regions were determined using limma in the R-package RnBeads. For the comparison of tumours with moderate/high ERß versus negative expression, differentially methylated CpG sites were identified but not confirmed by replication. Comparing tumours of high with tumours of negative ERß expression revealed 2,904 differentially methylated CpG sites of which 403 were replicated (FDR adjusted p-value<0.05). Replicated CpGs were annotated to genes such as CD36, HK1 or LRP5. A survival analysis indicates that 30 of the replicated CpGs are also associated with overall survival (FDR-adjusted p-value<0.05). The regional analysis identified 60 differentially methylated promotor regions. The epigenome-wide analysis identified both novel genes as well as genes already implicated in CRC. Follow-up mechanistic studies to better understand the regulatory role of ERß could inform potential targets for improving treatment or prevention of CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Adult , Aged , Aged, 80 and over , Case-Control Studies , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Genome-Wide Association Study , Humans , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Survival Rate
11.
Nat Commun ; 8(1): 1323, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29109526

ABSTRACT

Pancreatic acinar cell carcinoma (ACC) is an aggressive exocrine tumor with largely unknown biology. Here, to identify potential targets for personalized treatment, we perform integrative genome-wide and epigenome-wide analyses. The results show frequently aberrant DNA methylation, abundant chromosomal amplifications and deletions, and mutational signatures suggesting defective DNA repair. In contrast to pancreatic ductal adenocarcinoma, no recurrent point mutations are detected. The tumor suppressors ID3, ARID1A, APC, and CDKN2A are frequently impaired also on the protein level and thus potentially affect ACC tumorigenesis. Consequently, this work identifies promising therapeutic targets in ACC for drugs recently approved for precision cancer therapy.


Subject(s)
Carcinoma, Acinar Cell/genetics , Epigenesis, Genetic , Genomic Instability , Pancreatic Neoplasms/genetics , Carcinoma, Acinar Cell/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Checkpoints/genetics , Chromosome Aberrations , Gene Dosage , Genes, Tumor Suppressor , Humans , Mutation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms
12.
Radiother Oncol ; 125(1): 168-174, 2017 10.
Article in English | MEDLINE | ID: mdl-28916223

ABSTRACT

BACKGROUND AND PURPOSE: Fibrosis is a frequent adverse effect of radiotherapy and no effective treatments are currently available to prevent or reverse fibrotic disease. We have previously identified altered epigenetic patterns at a gene enhancer of the diacylglycerol kinase alpha (DGKA) locus in normal skin fibroblasts derived from fibrosis patients. An open chromatin pattern related to radiation-inducibility of DGKA is associated with onset of radiation-induced fibrosis. Here, we explore epigenetic modulation of DGKA as a way to mitigate predisposition to fibrosis. MATERIAL AND METHODS: We studied the effect of the BET-bromodomain inhibitors (JQ1, PFI-1) on DGKA inducibility in primary fibroblasts. Hence, DGKA transcription was additionally induced by the radiomimetic drug bleomycin, and DGKA mRNA expression, histone H3K27 acetylation and downstream markers of profibrotic fibroblast activation after BET-bromodomain inhibition were determined. RESULTS: BET-bromodomain inhibition suppressed induction of DGKA in bleomycin-treated fibroblasts, reduced H3K27ac at the DGKA enhancer and repressed collagen marker gene expression. Alterations in fibroblast morphology and reduction of collagen deposition were observed. CONCLUSION: For the DGKA enhancer, we show that BET-bromodomain inhibitors can alter the epigenetic landscape of fibroblasts, thus counteracting profibrotic transcriptional events. Interference with epigenetic patterns of fibrosis predisposition may provide novel preventive therapies that improve radiotherapy.


Subject(s)
Diacylglycerol Kinase/genetics , Fibrosis/etiology , Proteins/antagonists & inhibitors , Radiation Injuries/etiology , Acetylation , Aged , Azepines/pharmacology , Bleomycin/pharmacology , Diacylglycerol Kinase/biosynthesis , Enhancer Elements, Genetic/drug effects , Enhancer Elements, Genetic/radiation effects , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/radiation effects , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/radiation effects , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Humans , Middle Aged , Radiation Injuries/genetics , Radiation Injuries/metabolism , Radiation Injuries/pathology , Skin/drug effects , Skin/pathology , Skin/radiation effects , Triazoles/pharmacology
13.
Oncotarget ; 7(41): 66344-66359, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27572323

ABSTRACT

The systemic and resistant nature of metastatic neuroblastoma renders it largely incurable with current multimodal treatment. Clinical progression stems mainly from the increasing burden of metastatic colonization. Therapeutically inhibiting the migration-invasion-metastasis cascade would be of great benefit, but the mechanisms driving this cycle are as yet poorly understood. In-depth transcriptome analyses and ChIP-qPCR identified the cell surface glycoprotein, CD9, as a major downstream player and direct target of the recently described GRHL1 tumor suppressor. CD9 is known to block or facilitate cancer cell motility and metastasis dependent upon entity. High-level CD9 expression in primary neuroblastomas correlated with patient survival and established markers for favorable disease. Low-level CD9 expression was an independent risk factor for adverse outcome. MYCN and HDAC5 colocalized to the CD9 promoter and repressed transcription. CD9 expression diminished with progressive tumor development in the TH-MYCN transgenic mouse model for neuroblastoma, and CD9 expression in neuroblastic tumors was far below that in ganglia from wildtype mice. Primary neuroblastomas lacking MYCN amplifications displayed differential CD9 promoter methylation in methyl-CpG-binding domain sequencing analyses, and high-level methylation was associated with advanced stage disease, supporting epigenetic regulation. Inducing CD9 expression in a SH-EP cell model inhibited migration and invasion in Boyden chamber assays. Enforced CD9 expression in neuroblastoma cells transplanted onto chicken chorioallantoic membranes strongly reduced metastasis to embryonic bone marrow. Combined treatment of neuroblastoma cells with HDAC/DNA methyltransferase inhibitors synergistically induced CD9 expression despite hypoxic, metabolic or cytotoxic stress. Our results show CD9 is a critical and indirectly druggable suppressor of the invasion-metastasis cycle in neuroblastoma.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Histone Deacetylases/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/pathology , Tetraspanin 29/biosynthesis , Animals , Histone Deacetylases/genetics , Humans , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Invasiveness/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Tetraspanin 29/genetics
14.
Radiother Oncol ; 121(3): 431-439, 2016 12.
Article in English | MEDLINE | ID: mdl-27443449

ABSTRACT

PURPOSE: Several small studies have indicated that the ATM rs1801516 SNP is associated with risk of normal tissue toxicity after radiotherapy. However, the findings have not been consistent. In order to test this SNP in a well-powered study, an individual patient data meta-analysis was carried out by the International Radiogenomics Consortium. MATERIALS AND METHODS: The analysis included 5456 patients from 17 different cohorts. 2759 patients were given radiotherapy for breast cancer and 2697 for prostate cancer. Eight toxicity scores (overall toxicity, acute toxicity, late toxicity, acute skin toxicity, acute rectal toxicity, telangiectasia, fibrosis and late rectal toxicity) were analyzed. Adjustments were made for treatment and patient related factors with potential impact on the risk of toxicity. RESULTS: For all endpoints except late rectal toxicity, a significantly increased risk of toxicity was found for carriers of the minor (Asn) allele with odds ratios of approximately 1.5 for acute toxicity and 1.2 for late toxicity. The results were consistent with a co-dominant pattern of inheritance. CONCLUSION: This study convincingly showed a significant association between the ATM rs1801516 Asn allele and increased risk of radiation-induced normal tissue toxicity.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Breast Neoplasms/radiotherapy , Prostatic Neoplasms/radiotherapy , Radiation Injuries/genetics , Alleles , Breast Neoplasms/genetics , Female , Genetic Predisposition to Disease , Genotype , Heterozygote , Humans , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Radiation Injuries/etiology , Radiation Tolerance/genetics , Radiotherapy/adverse effects , Risk Factors
15.
Pharmacogenomics ; 17(7): 755-94, 2016 05.
Article in English | MEDLINE | ID: mdl-27183147

ABSTRACT

Several new chemotherapeutic agents have become available for the treatment of colorectal cancer, which has led to increased complexity in treatment planning. Treatment decision making for individual patients could be facilitated if guided by predictive and prognostic markers. As most cytotoxic drugs induce DNA damage, the DNA damage repair pathways hold potential for yielding such biomarkers. Here, we review the current evidence of a possible involvement of the nucleotide excision repair pathway in the efficacy of chemotherapeutic agents used in the treatment of colorectal cancer. Although a large number of studies have been conducted, they are generally of moderate size and heterogeneous in design. Up to date no firm conclusions can be drawn to translate these results into the clinic. We recommend further comprehensive investigations of the nucleotide excision repair pathway in large patient studies that include both discovery and validation cohorts.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , DNA Repair/drug effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Colorectal Neoplasms/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Fluorouracil/pharmacology , Genetic Markers , Humans , Irinotecan , Organoplatinum Compounds/pharmacology , Oxaliplatin , Pharmacogenomic Testing , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
16.
Int J Cancer ; 139(2): 424-32, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26934681

ABSTRACT

MGMT promoter methylation status is currently the only established molecular prognosticator in IDH wild-type glioblastoma multiforme (GBM). Therefore, we aimed to discover novel therapy-associated epigenetic biomarkers. After enrichment for hypermethylated fractions using methyl-CpG-immunoprecipitation (MCIp), we performed global DNA methylation profiling for 14 long-term (LTS; >36 months) and 15 short-term (STS; 6-10 months) surviving GBM patients. Even after exclusion of the G-CIMP phenotype, we observed marked differences between the LTS and STS methylome. A total of 1,247 probes in 706 genes were hypermethylated in LTS and 463 probes in 305 genes were found to be hypermethylated in STS patients (p values < 0.05, log2 fold change ± 0.5). We identified 13 differentially methylated regions (DMRs) with a minimum of four differentially methylated probes per gene. Indeed, we were able to validate a subset of these DMRs through a second, independent method (MassARRAY) in our LTS/STS training set (ADCY1, GPC3, LOC283731/ISLR2). These DMRs were further assessed for their prognostic capability in an independent validation cohort (n = 62) of non-G-CIMP GBMs from the TCGA. Hypermethylation of multiple CpGs mapping to the promoter region of LOC283731 correlated with improved patient outcome (p = 0.03). The prognostic performance of LOC283731 promoter hypermethylation was confirmed in a third independent study cohort (n = 89), and was independent of gender, performance (KPS) and MGMT status (p = 0.0485, HR = 0.63). Intriguingly, the prediction was most pronounced in younger GBM patients (<60 years). In conclusion, we provide compelling evidence that promoter methylation status of this novel gene is a prognostic biomarker in IDH1 wild-type/non-G-CIMP GBMs.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/mortality , DNA Methylation , Glioblastoma/genetics , Glioblastoma/mortality , Isocitrate Dehydrogenase/genetics , Promoter Regions, Genetic , Adult , Aged , Aged, 80 and over , Brain Neoplasms/therapy , Chemoradiotherapy , CpG Islands , Female , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Glioblastoma/therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Reproducibility of Results , Young Adult
17.
Nat Commun ; 7: 10893, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964756

ABSTRACT

Radiotherapy is a fundamental part of cancer treatment but its use is limited by the onset of late adverse effects in the normal tissue, especially radiation-induced fibrosis. Since the molecular causes for fibrosis are largely unknown, we analyse if epigenetic regulation might explain inter-individual differences in fibrosis risk. DNA methylation profiling of dermal fibroblasts obtained from breast cancer patients prior to irradiation identifies differences associated with fibrosis. One region is characterized as a differentially methylated enhancer of diacylglycerol kinase alpha (DGKA). Decreased DNA methylation at this enhancer enables recruitment of the profibrotic transcription factor early growth response 1 (EGR1) and facilitates radiation-induced DGKA transcription in cells from patients later developing fibrosis. Conversely, inhibition of DGKA has pronounced effects on diacylglycerol-mediated lipid homeostasis and reduces profibrotic fibroblast activation. Collectively, DGKA is an epigenetically deregulated kinase involved in radiation response and may serve as a marker and therapeutic target for personalized radiotherapy.


Subject(s)
Breast Neoplasms/radiotherapy , Breast/pathology , DNA Methylation/genetics , Diacylglycerol Kinase/genetics , Epigenesis, Genetic/genetics , Fibroblasts/pathology , RNA, Messenger/metabolism , Radiation Injuries/genetics , Adult , Aged , Blotting, Western , Case-Control Studies , Chromatin Immunoprecipitation , Chromatography, Liquid , Diacylglycerol Kinase/antagonists & inhibitors , Early Growth Response Protein 1/metabolism , Female , Fibrosis/etiology , Fibrosis/genetics , HCT116 Cells , HEK293 Cells , Humans , Mass Spectrometry , Middle Aged , Radiotherapy/adverse effects , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology , Transcriptome
18.
Cancer Epidemiol Biomarkers Prev ; 25(1): 180-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26585118

ABSTRACT

BACKGROUND: Aspirin use is an effective strategy for the chemoprevention of colorectal cancer, even at low doses. However, in order to implement aspirin interventions, risk-benefit balances and biologic mechanisms need to be better defined; to further this aim, we used a metabolomics approach. METHODS: We metabolically profiled 40 healthy, nonsmoking men and women ages 20 to 45 years enrolled in a randomized, double-blind, crossover trial of 325 mg aspirin/day over a period of 60 days. Gas and liquid chromatography-mass spectrometry were used to comprehensively profile participants' plasma samples after aspirin and placebo interventions. RESULTS: A total of 363 metabolites, covering most human biochemical pathways, were measured. Compared with placebo-treated participants, plasma concentrations of the oncometabolite 2-hydroxyglutarate (R+S) decreased after aspirin treatment in both men and women (P = 0.005). This signal proved robust during 20-fold random splitting of the data using 80% of the samples in each split. We subsequently performed functional follow-up studies using targeted, enantiospecific detection in human colorectal cancer cell lines and observed an aspirin-induced reduction of (R)-2-hydroxyglutarate. We further showed that salicylate, the primary aspirin metabolite, inhibits the hydroxyacid-oxoacid transhydrogenase mediated production of (R)-2-hydroxyglutarate, thereby providing mechanistic evidence for the clinically observed effects of aspirin on total-2-hydroxyglutarate. CONCLUSIONS: Using a metabolomics approach with functional follow-up, we propose that a decrease in the oncometabolite (R)-2-hydroxyglutarate may identify an additional mechanism for aspirin or its metabolites in cancer prevention. IMPACT: Reduction of the oncometabolite (R)-2-hydroxyglutarate identifies a novel, non-COX-inhibition-mediated mechanism of aspirin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Colorectal Neoplasms/drug therapy , Glutarates/blood , Metabolomics/methods , Adult , Alcohol Oxidoreductases/antagonists & inhibitors , Colorectal Neoplasms/metabolism , Cross-Over Studies , Double-Blind Method , Enzyme Inhibitors/pharmacology , Female , Follow-Up Studies , Healthy Volunteers , Humans , Male , Middle Aged , Mitochondrial Proteins/antagonists & inhibitors , Prognosis , Risk Assessment , Tumor Cells, Cultured , Young Adult
19.
BMC Cancer ; 15: 978, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674097

ABSTRACT

BACKGROUND: Personalized therapy considering clinical and genetic patient characteristics will further improve breast cancer survival. Two widely used treatments, chemotherapy and radiotherapy, can induce oxidative DNA damage and, if not repaired, cell death. Since base excision repair (BER) activity is specific for oxidative DNA damage, we hypothesized that germline genetic variation in this pathway will affect breast cancer-specific survival depending on treatment. METHODS: We assessed in 1,408 postmenopausal breast cancer patients from the German MARIE study whether cancer specific survival after adjuvant chemotherapy, anthracycline chemotherapy, and radiotherapy is modulated by 127 Single Nucleotide Polymorphisms (SNPs) in 21 BER genes. For SNPs with interaction terms showing p<0.1 (likelihood ratio test) using multivariable Cox proportional hazard analyses, replication in 6,392 patients from nine studies of the Breast Cancer Association Consortium (BCAC) was performed. RESULTS: rs878156 in PARP2 showed a differential effect by chemotherapy (p=0.093) and was replicated in BCAC studies (p=0.009; combined analysis p=0.002). Compared to non-carriers, carriers of the variant G allele (minor allele frequency=0.07) showed better survival after chemotherapy (combined allelic hazard ratio (HR)=0.75, 95% 0.53-1.07) and poorer survival when not treated with chemotherapy (HR=1.42, 95% 1.08-1.85). A similar effect modification by rs878156 was observed for anthracycline-based chemotherapy in both MARIE and BCAC, with improved survival in carriers (combined allelic HR=0.73, 95% CI 0.40-1.32). None of the SNPs showed significant differential effects by radiotherapy. CONCLUSIONS: Our data suggest for the first time that a SNP in PARP2, rs878156, may together with other genetic variants modulate cancer specific survival in breast cancer patients depending on chemotherapy. These germline SNPs could contribute towards the design of predictive tests for breast cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Poly(ADP-ribose) Polymerases/genetics , Aged , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Female , Genotype , Humans , Middle Aged , Polymorphism, Single Nucleotide , Postmenopause , Prognosis , Proportional Hazards Models , Radiotherapy
20.
Int J Radiat Oncol Biol Phys ; 92(5): 1084-1092, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26072091

ABSTRACT

PURPOSE: To identify single-nucleotide polymorphisms (SNPs) in oxidative stress-related genes associated with risk of late toxicities in breast cancer patients receiving radiation therapy. METHODS AND MATERIALS: Using a 2-stage design, 305 SNPs in 59 candidate genes were investigated in the discovery phase in 753 breast cancer patients from 2 prospective cohorts from Germany. The 10 most promising SNPs in 4 genes were evaluated in the replication phase in up to 1883 breast cancer patients from 6 cohorts identified through the Radiogenomics Consortium. Outcomes of interest were late skin toxicity and fibrosis of the breast, as well as an overall toxicity score (Standardized Total Average Toxicity). Multivariable logistic and linear regression models were used to assess associations between SNPs and late toxicity. A meta-analysis approach was used to summarize evidence. RESULTS: The association of a genetic variant in the base excision repair gene XRCC1, rs2682585, with normal tissue late radiation toxicity was replicated in all tested studies. In the combined analysis of discovery and replication cohorts, carrying the rare allele was associated with a significantly lower risk of skin toxicities (multivariate odds ratio 0.77, 95% confidence interval 0.61-0.96, P=.02) and a decrease in Standardized Total Average Toxicity scores (-0.08, 95% confidence interval -0.15 to -0.02, P=.016). CONCLUSIONS: Using a stage design with replication, we identified a variant allele in the base excision repair gene XRCC1 that could be used in combination with additional variants for developing a test to predict late toxicities after radiation therapy in breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Breast/radiation effects , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Radiation Injuries/genetics , Adult , Aged , Aged, 80 and over , Alleles , Breast/pathology , Cohort Studies , Female , Fibrosis/genetics , Genome-Wide Association Study , Germany , Humans , Middle Aged , Odds Ratio , Oxidative Stress/genetics , Phenotype , Predictive Value of Tests , Radiation Injuries/pathology , Radiation Tolerance/genetics , X-ray Repair Cross Complementing Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...