Skip to main content
Erschienen in: Molecular Diagnosis & Therapy 1/2019

Open Access 01.02.2019 | Review Article

Prognostic and Predictive Epigenetic Biomarkers in Oncology

verfasst von: Katarzyna Kamińska, Ewelina Nalejska, Marta Kubiak, Joanna Wojtysiak, Łukasz Żołna, Janusz Kowalewski, Marzena Anna Lewandowska

Erschienen in: Molecular Diagnosis & Therapy | Ausgabe 1/2019

Abstract

Epigenetic patterns, such as DNA methylation, histone modifications, and non-coding RNAs, can be both driver factors and characteristic features of certain malignancies. Aberrant DNA methylation can lead to silencing of crucial tumor suppressor genes or upregulation of oncogene expression. Histone modifications and chromatin spatial organization, which affect transcription, regulation of gene expression, DNA repair, and replication, have been associated with multiple tumors. Certain microRNAs (miRNAs), mainly those that silence tumor suppressor genes and occur in a greater number of copies, have also been shown to promote oncogenesis. Multiple patterns of these epigenetic factors occur specifically in certain malignancies, which allows their potential use as biomarkers. This review presents examples of tests for each group of epigenetic factors that are currently available or in development for use in early cancer detection, prediction, prognosis, and response to treatment. The availability of blood-based biomarkers is noted, as they allow sampling invasiveness to be reduced and the sampling procedure to be simplified. The article stresses the role of epigenetics as a crucial element of future cancer diagnostics and therapy.
Key Points
There is a broad spectrum of epigenetic biomarkers in oncology, but those related to methylation and microRNA (miRNA) copy number variation or changed expression seem to be the most promising.
There are a few diagnostics assays that provide prognostic and predictive information.
Epigenetic alterations have been identified in many solid tumors in formalin-fixed paraffin-embedded material; however, it seems that miRNAs and circulating tumor DNA are the most promising biological material to work with in personalized oncology.

1 Introduction

Genetic information is provided by the order of nucleotides arranged in DNA, but it is the pattern of epigenetic factors that dictates how this information should be expressed. Epigenetics answer the question of how the same genotype can be translated into different phenotypes [1]. Methylation of DNA, heritable modifications of chromatin (histone and nucleosome modifications), and regulation by non-coding RNAs are essential for normal development and maintenance of tissue-specific gene expression patterns in mammals [2, 3].
Epimutation is defined as a heritable abnormal transcriptional repression of gene activity that is not associated with a DNA sequence. Epimutation generally occurs in somatic cells and manifests as tumor progression but may also arise in the germline. Several diseases have been linked to epigenetic alterations. Epigenetic mutations and secondary genetic mutations in both cis and trans, including copy number variation (CNV), chromosomal rearrangements, and uniparental disomy [4], lead to eight classic imprinting disorders: (1) Angelman syndrome; (2) Prader–Willi syndrome [5]; (3) Beckwit–Wiedemann syndrome; (4) Silver–Russell syndrome; (5) Temple syndrome; (6) Wang syndrome [4]; (7) transient neonatal type 1 diabetes mellitus [6]; and (8) pseudohypoparathyroidism type 1. Cancers associated with changes in the epigenetic landscape include Lynch syndrome (hereditary non-polyposis colorectal cancer [HNPCC]) (Table 1), chronic lymphocytic leukemia (CLL), breast cancer, and ovarian cancer. Many tumor suppressor genes (i.e., RB1, VHL, MLH1, APC, and BRCA1) are involved in epigenetic abnormalities [3].
Table 1
Methylation: prognostic and predictive biomarkers with diagnostic utility
Methylation
Prognostic
Predictive
Invasive/non-invasive diagnostics
Biological material
Target cancer
MLH1
Hypomethylation
+
Invasive
FFPE
Colorectal cancer [16, 17]
MGMT
Hypermethylation
+
+
Invasive
FFPE
Glioblastoma [24, 25]
IDH1 p.R132H mutation and MGMT hypermethylation
+
Invasive
FFPE
Glioblastoma [30]
RB1 hypermethylation
+
Invasive
FFPE
Retinoblastoma [33, 34]
GSTP1, RASSF1, APC methylation status
+
Invasive
FFPE
Prostate cancer [35, 36]
SEPT9
+
Non-invasive
Blood
Colorectal cancer [39, 40]
Lung cancer [41]
MGMT-STP27
+
Invasive
FFPE
Oligodendrogliomas and oligoastrocytomas [124]
ESR1
+
Non-invasive
Blood
Breast cancer [125]
ZNF331
+
Invasive
FFPE
Colorectal cancer [126]
SALL1
+
Invasive
FFPE
Head and neck cancer [127]
FFPE formalin-fixed paraffin-embedded cancer tissue
DNA methylation, histone modifications, and non-coding RNAs are present ubiquitously in all types of human malignancies and can appear at early stages of cancer, and therefore constitute particularly attractive markers with a wide range of applications in diagnostics [7].
The first epigenetic data were available in ENCODE (Encyclopedia of DNA Elements) for different cell lines. The first map of epigenetic changes in cancer tissue was included in The Cancer Genome Atlas (TCGA). Finally, the map of human epigenome has been developed under the Human Epigenome Project (HEP). Over the years, ENCODE has become a large database including information about the epigenome of 212 cell cultures, as well as that of many types of human cancer tissues. The HEP has expanded and united many international members, creating the International Human Epigenome Consortium (IHEC) [8]. Thus, we have gained a better understanding of the role of epigenetic changes in normal development, aging, imprinting disorders, and abnormal gene control in cancer, as well as the role of the environment in shaping phenotype in humans [9].

2 Aberrant DNA Methylation in Cancer

Aberrant DNA methylation is the main studied epigenetic alteration in cancer [10]. Aberrant hypermethylation of promoters in eukaryotic cells can lead to silencing of important genes, such as tumor suppressor genes, and ultimately result in the development of disease. Cancer development can also be affected by the opposite process. Hypomethylation of genes, e.g., oncogenes, which are normally methylated, can upregulate their expression [11]. Interestingly, DNA hypomethylation was the first reported DNA methylation abnormality in human cancer [12]. However, despite the initial evidence provided in 1983 and by later work, studies of the molecular mechanisms leading to cancer have not focused on epigenetics [12, 13]. Instead, the emphasis was put on DNA mutations and loss of heterozygosity (LOH) [11]. In 1999, Toyota et al. [14] proposed the CpG Island Methylator Phenotype (CIMP) as another pathway of tumorigenesis. They used CIMP to describe the clinical and pathological features of colorectal cancer (CRC) for the first time. This pioneering study consisted of methylation profiling of the CDKN2A (p16), MINT1, MINT2, MINT12, MINT17, MINT25, MINT27, MINT31, MLH1, and THBS1 genes in tumor tissue [15]. Almost 20 years after the publication by Toyota et al. [14], we have now developed methylation in vitro diagnostic (IVD) assays for blood and tissue [13]. Moreover, they have been successfully introduced in the clinical setting for cancer screening, prognosis, and prediction [15].

2.1 DNA Methylation Biomarkers in Tissue

One of the genes included in the first methylation profiling in 1999 was MLH1, a DNA mismatch repair (MMR) gene (Table 1). Epigenetic silencing of the MLH1 gene via hypermethylation of its promoter results in microsatellite instability (MSI). It has been found that 13% of sporadic CRCs show MLH1 hypermethylation, and a BRAF c.1799T>A, p.Val600Glu mutation has often also been identified in tumor DNA [16, 17]. MSI and loss of MLS1 also occurs in Lynch syndrome (the most common cause of hereditary CRC) [18], but it is caused by mutations in one of the DNA MMR genes. In order to fully diagnose Lynch syndrome, genetic analysis of constitutional mutations in the MMR genes is performed. Differentiation of non-heritable CRC and Lynch syndrome includes a two-level screening test. The first tier includes analysis of expression of MMR genes and MSI testing. In the case of loss of MMR expression and a positive result for MSI, constitutional mutations in MLH1, MSH2, MSH6, PMS2, or EPCAM are analyzed. Alternatively, determination of the MLH1 methylation level and the BRAF V600E mutation is conducted. Constitutional MLH1 epimutations testing is recommended to confirm Lynch syndrome [19]. Methylation analysis of MLH1 can improve the selection of patients for Lynch syndrome genetic testing and thus reduce the cost of detecting a mutation by almost half [20]. MLH1 methylation can be assessed by methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) [21, 22] and some laboratories use pyrosequencing [23].
Clinical trials have provided evidence that O6-methylguanine (O6-meG)–DNA methyltransferase (MGMT) is useful as a prognostic and predictive marker in glioblastoma (Table 1) [24, 25]. MGMT is a DNA repair gene participating in the removal of mutagenic and cytotoxic alkyl groups from O6-meG [28]. DNA alkylation leads to the formation of mutations, and therefore MGMT protect cells against damage [26, 27]. Temozolomide causes alkyl DNA damage and thus leads to cell death. Its cytotoxic effect is more potent against the rapidly dividing cancer cells than against normal cells, as the DNA repair mechanisms in cancer cells are impaired [25, 28]. Therefore, cells with MGMT silenced by hypermethylation show a better response to temozolomide therapy [25]. It has been found that in glioma and CRC, methylation of MGMT occurs in 40% of tumors, while in non-small cell lung carcinomas (NSCLCs), lymphomas, and head and neck carcinomas, methylation of MGMT occurs in 25% of tumors [27]. Diagnostic recommendations for glioma include analysis of MGMT methylation, which is the key point in the therapeutic algorithm and provides predictive information about the response to temozolomide [29]. Furthermore, the MGMT methylation status in combination with IDH1 mutations plays the role of a prognostic biomarker. Glioma patients with the IDH1 p.R132H mutation and hypermethylated MGMT have a better prognosis (Table 1) [30].
There are a number of commercial tests available to evaluate the MGMT methylation level by (1) methylation-specific polymerase chain reaction (PCR): PredictMDx Glioblastoma (MDx Health); (2) real-time PCR: MGMT Methylation Detection Kit (EntroGen); (3) MS-MLPA: SALSA MS-MLPA probe mix ME011 MMR genes (MRC-Holland); and (4) pyrosequencing technology: PyroMark MGMT Kit (Qiagen).
The RB1 gene is primarily associated with retinoblastoma caused by the loss of the RB1 function (Table 1). The lack of expression of this gene in retinoblastoma, as well as in other tumors, including bladder carcinomas and malignant neuroendocrine lung carcinomas, is associated with an LOH or RB1 mutations. Nevertheless, in some cases, the silencing of RB1 expression is caused by its methylation [31, 32]. It has been reported that for full molecular diagnostics of retinoblastoma, it is necessary to evaluate RB1 methylation beyond the LOH and mutations. Ohtani-Fujita et al. [33] suggested that hypermethylation in the RB1 gene is always acquired and causes approximately 9% of sporadic unilateral tumors [33]. Currently, there are tests available on the market based on the MS-MLPA methodology for the evaluation of the methylation level of the RB1 promoter [34].
Tumor suppressor genes GSTP1, RASSF1, and APC are commonly methylated in prostate tumors and, therefore, are considered as cancer biomarkers (Table 1). A set of these genes has been used in a commercially available assay—ConfirmMDx® (MDxHealth). This test allows a better stratification of patients with a negative prostate biopsy result. It takes advantage of the epigenetic field effect, based on the principle that normal cells surrounding the foci of cancer can contain DNA methylation changes. Two independent studies, MATLOC (Methylation Analysis to Locate Occult Cancer) and DOCUMENT (Detection of Cancer Using Methylated Events in Negative Tissue), confirmed the predictive value of ConfirmMDx® and showed a sensitivity of 68%, a specificity of 64%, and a negative predictive value of 90% [35, 36]. Moreover, it was found that use of the methylation-based biomarkers GSTP1, RASSF1, and APC resulted in a reduction of the number of unnecessary repeated biopsies by up to 64% [35].

2.2 DNA Methylation Biomarkers in Biofluids

Measurement of DNA methylation can be performed in various types of biological material—not only solid tissues, but also plasma, serum, sputum, urine, and cerebrospinal fluid (CSF). One of the examples of a plasma epigenetic biomarker for CRC screening is circulating methylated SEPT9 DNA (Table 1). SEPT9 regulates cell growth and prevents uncontrolled cell division and, therefore, it is considered to be a tumor suppressor [37]. It has been demonstrated that methylation of SEPT9 is associated with the pathogenesis of CRC, and a decrease in its expression is correlated with progression of neoplastic disease [38]. The first commercial diagnostic test based on the SEPT9 biomarker was developed by Epigenomics AG 10 years ago. It involves evaluation of the SEPT9 promoter methylation in plasma using real-time PCR [39]. Currently, two generations of these CE-marked IVD assays called Epi proColon® are available [39]. Other commercially available SEPT9 methylation tests for CRC diagnostics are ColoVantage® (Quest Diagnostics) and RealTime mS9 (Abbott) [40]. Apart from CRC, the usefulness of SEPT9 methylation has been evaluated in the early diagnostics of others cancers, including lung cancer [41]. However, there are more specific tests for lung cancer diagnostics, such as Epi proLung® (Epigenomics AG), in which methylation of SHOX2 and PTGER4 is evaluated using real-time PCR [15, 42]. SHOX2 hypermethylation has been noticed in the bronchial aspirates [43], pleural effusions [44], and blood plasma of patients with lung cancer [15]. DNA methylation analysis of SHOX2 combined with PTGER4 in blood plasma allows detection of lung cancer and differentiation of non-malignant diseases [15]. Additionally, prognostic application of an assay based on SHOX2 methylation has been demonstrated [45]. The advantage of SHOX2 as a methylation biomarker is its high specificity (> 95% in bronchial aspirates) [43, 45].
In the case of CRC, in addition to the previously mentioned biomarkers, it is also possible to use a stool-based methylated biomarker test, i.e., the Cologuard® kit (Exact Sciences) [40, 46]. This PCR-based assay is used to assess the level of vimentin gene (VIM) methylation and DNA integrity for the early detection of CRC. The sensitivity and specificity of the Cologuard® assay are 83% and 82%, respectively, and the specificity is almost at the same level in patients with CRC at stages I–III [47, 48].

3 Histone Modification in Cancer

Chromatin can generally be distinguished into heterochromatin, which is highly condensed and contains inactive genes, and euchromatin, which is more relaxed and contains actively transcribed genes. Covalent modification of the nucleosome components—histones—alters the organization and function of chromatin, which affects gene regulation and expression [49]. Histone modification plays an important role in the determination of the role of chromatin, which can be divided into six broad functions: promoter, enhancer, insulator, transcribed, repressed, and inactive chromatin [50]. Modification of histone structure includes phosphorylation, acetylation, methylation (mainly of lysine and arginine residues), ubiquitylation, glycosylation, SUMOylation, ADP (adenosine diphosphate)-ribosylation, and carbonylation [49, 51]. Histone acetylation and methylation are often associated with a more relaxed chromatin conformation and are the main markers of active chromatin. On the other hand, histone deacetylation and phosphorylation are often associated with chromatin condensation and are markers of inactive chromatin [49].
Over the last 15 years, global histone modifications have been analyzed along with single acetylation and methylation of lysine and arginine residues in coding regions and non-promoter sequences. Importantly, global histone H3 and H4 modification patterns are potential markers of tumor recurrence and disease-free survival. NSCLC patients with global histone acetylation have shown a better prognosis in survival analysis than patients with globally methylated or non-modified histones, which were associated with poor prognosis [52]. The profiling of histone modifications allows patients to be classified into two subtypes, with distinct risks of tumor recurrence in patients with low-grade prostate cancer [53] and has also identified bladder cancer patients at risk of early cancer recurrence [54]. Since global profiling is more costly, single modifications are often evaluated as possible prognostic tools. For example, Fraga et al. [55] found that a global loss of H4 histone Lys16 monoacetylation and Lys20 trimethylation is associated with hypomethylation of DNA and is generally characteristic of human tumor cells. In breast cancer, a reduced status of H4Lys16 acetylation and H4Lys20 methylation is correlated with a worse prognosis [56]. However, the role of H4Lys16 acetylation and H4Lys20 methylation in cancer pathogenesis requires further investigation.
Not only covalent modifications but also various isoforms of histone proteins present in the nucleosome can increase the predisposition to cancer. For example, overexpression of an H2A histone isoform, H2A.Z, has been observed in undifferentiated cancers and genitourinary cancers (prostate and bladder cancers). Furthermore, H2A.Z can play a role in endocrine resistance in breast cancer patients. It seems that H2A.Z can be useful as a tumor progression biomarker because of the correlation between the levels of H2A.Z and short overall patient survival [57].
Analysis of histones, as well as of their covalent modifications, have also been identified in blood derived from cancer patients. Circulating histones reflect changes in tumor cells, and therefore are promising non-invasive biomarkers originating from blood (Table 2). The presence of histones in blood is a result of tumor cell death (apoptosis and necrosis) or active release from living cells. Increasing the level of circulating nucleosomes/histones has recently been identified in the blood of patients in a wide range of cancers, e.g., breast, colorectal, and prostate cancers [58, 59]. However, a comparison of the levels of circulating histones in cancer patients to results obtained in patients with numerous benign diseases has shown that the difference is not statistically significant, which reduces their clinical usefulness for detecting cancer [60]. Moreover, it has been found that the level of circulating nucleosomes can predict tumor responses to chemotherapeutic agents in various cancer types [61]. Increased levels of circulating nucleosomes have been associated with a worse response to chemotherapy in patients with advanced lung cancer [62], as well as to selective internal radiation therapy (SIRT) in metastasizing CRC [63].
Table 2
Histone modifications: prognostic and predictive biomarkers with diagnostic utility
Histone modifications
Prognostic
Predictive
Invasive/non-invasive diagnostics
Biological material
Target cancer
H3Cit
+
Non-invasive
Blood
Advanced cancers [66]
cf-nucleosome epitope combination
+
Non-invasive
Blood
Colorectal cancer [128]
H3K4me3 and Wdr82 expression
+
Non-invasive
Blood
Colorectal cancer [129]
FFPE formalin-fixed paraffin-embedded cancer tissue, H3Cit citrullinated histone 3
Regarding post-translational histone modifications, the levels of H3K9me3 and H4K20me3 have been downregulated in the blood of CRC patients in comparison with cancer-free individuals [64, 65]. In addition, a combination of H4K20me3 with H3K27me3 has shown an area under the curve (AUC) of 0.769 and a sensitivity of 49.2% at a 90% specificity for CRC patients [65]. Citrullinated histone 3 (H3Cit) has also recently been proposed as a prognostic biomarker in advanced cancers (Table 2). Thålin et al. [66] demonstrated a statistically significant correlation between the upregulation of circulating H3Cit histone levels and short-term mortality.

4 Mutations Affecting the SWitch/Sucrose Non-Fermentable (SWI/SNF) Chromatin Remodeling Complex

Not only epigenetic modifications of chromatin, such as histone modifications, but also the spatial organization of chromatin plays a critical role in the regulation of all principal biological processes, such as transcription, regulation of gene expression, and DNA repair and replication.
The SWitch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex is known to be mutated in up to 20% of human malignancies [49], including ovarian clear-cell carcinoma, clear-cell renal cell carcinoma, hepatocellular carcinoma, gastric cancer, melanoma, and pancreatic cancer [67].
One of the SWI/SNF family members is the SMARCB1 gene (hSNF5/INI1), which has been identified as a tumor suppressor gene [68]. Germline loss of function mutations in SMARCB1 predispose to schwannomatosis and malignant rhabdoid tumor [69]. Biallelic inactivation has been observed in nearly all cases of childhood rhabdoid sarcoma (somatic alteration of SMARCB1 locus that encompasses a complete loss of one allele with nonsense mutations or silencing by methylation in the other allele) [68, 70]. Identification of aberration of the SMARCB1 gene is generally more focused on genetic (point mutations, deletion, and amplifications) than epigenetic (methylation) changes. A broad range of diagnostic methods have been implemented to evaluate this marker in familial screening and rhabdoid tumor monitoring: PCR with bidirectional sequencing and microarray-based comparative genomic hybridization encompassing SMARCB1 [69], as well as MLPA assays together with fluorescence in situ hybridization (FISH) and single nucleotide polymorphism (SNP) array analysis (to identify the second inactivating event in rhabdoid tumors without homozygous SMARCB1 deletions) [71]. Finally, high-throughput technologies have been implemented: whole-genome (WGS) and whole-exome (WES) sequencing analysis, gene expression analysis using RNA sequencing and Illumina gene expression array, and, last but not least, epigenomic analysis using Illumina 450K DNA Methylation Array, Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq), and H3K27ac chromatin immunoprecipitation (ChIP) sequencing [71].
Another component of SWI/SNF chromatin remodeling is ARID1A, in which frequent aberrations have been shown to be associated with different cancer types (Table 3), such as uterine carcinomas [72], pancreatic ductal adenocarcinomas [73], breast cancers [74], lung cancer [75], hepatocellular carcinomas [76], and gastric cancers [77]. Moreover, intragenic deletions of ARID1 (ARID1A and ARID1B) (Table 3) in neuroblastoma tumors [78] and breast cancer [79] correlate with a more aggressive phenotype and poor prognosis for patients. These studies suggest that ARID1 may be a valuable prognostic factor in a broad range of tumors, e.g., invasive breast cancer [80], gastric cancer [81], and ovarian cancer [82].
Table 3
Chromatin conformation: prognostic and predictive biomarkers with diagnostic utility
Chromatin conformation
Prognostic
Predictive
Invasive/non-invasive diagnostics
Biological material
Target cancer
ARID1 (ARID1A and ARIDB)
+
Non-invasive
Blood
Neuroblastoma tumors [78]
Methylation status of CTCF locus
+
Invasive
FFPE
Colorectal cancer [88]
SMARCA4/BRG1
+
Invasive
Frozen tissue
Non-small cell lung cancer [130]
FFPE formalin-fixed paraffin-embedded cancer tissue

5 Chromatin Conformation in Cancer

Spatial organization of chromatin plays an important role in many malignant tumors, and chromosomal conformation changes inside the nucleus can be detected effectively using a broad spectrum of chromosome conformation capture (3C)-based methods [83]. CTCF (CCCTC-binding factor) is a transcription factor that plays a crucial role in the formation of the three-dimensional chromatin architecture and epigenetic homeostasis and is present in over 86% of loops [84]. Disruption of the genomic neighborhood through abnormal CTCF binding to the imprinting control region can result in Beckwith–Wiedemann or Silver–Russell syndromes [85], and can be involved in various cancers (Table 3) [86].
Aberrations of CTCF bindings depend on many mechanisms, including DNA methylation at the imprinting control region observed in testicular germ-cell tumors (TGCTs), CRC, bladder cancer, and ovarian cancer [87]. Liu et al. [88] indicated a set of five biomarkers (CTCF_13 [chr10: 15761963-15762083], CTCF_33 [chr13: 61564369-61564467], CTCF_55 [chr17: 27940559-27940669], CTCF_94 [chr4: 165304966-165305120], and CTCF_113 [chr8: 69243102-69243188]) based on the methylation status of CTCF binding sites that were considered as a diagnostic marker panel in CRC (Table 3) [88]. This panel achieves a sensitivity of 91.67% for adenomas, 97.44% for stage I CRC, 94.06% for stage II CRC, 93.62% for stage III CRC, and 93.54% for total colorectal tumors with a specificity of 94.05%. As the CTCF gene plays a critical role in gene silencing, in addition to abnormal CTCF binding, loss of this gene is a known genetic event in an invasive and in situ carcinoma. Deletion of the 16q22.1 region in which the CTCF gene is located has been proposed as a novel biomarker for lobular carcinoma in situ (LCIS) [89].

6 Aberrant MicroRNA (miRNA) in Cancer

The microRNA (miRNA)–messenger RNA (mRNA) interactions are the second, after methylation, intensively studied epigenetic mechanism of gene regulation. miRNAs (miRs) were discovered in 1993 by Lee et al. [90] in Caenorhabditis elegans. They are small, non-coding RNA molecules of 18–25 nucleotides, which play a key role in the regulation of gene expression at the post-transcriptional level. miRNAs function by binding to the target mRNA molecule, thus inhibiting translation. It is estimated that up to 60% of genes encoding proteins can be controlled by miRNA.
The first evidence that miRNAs may be involved in the pathogenesis of human cancers was found in 2002 in a study of CLL [91]. This study showed that the deletion of miR-15a and miR-16-1 genes located on chromosome 13q14 is associated with the development of CLL [91]. Moreover, it has been found that downregulation of these two miRNAs correlates with Bcl2 (antiapoptotic B cell lymphoma 2 protein) overexpression [92]. Bcl2 is involved in inhibiting cell death. Malignant B cells are immortalized due to the absence of miR15-a and miR16-1 that are able to repress Bcl2 at the post-transcriptional level [92]. Deletions of miR15 and miR16 located in cluster 13q14.3 were found in ~ 65% of patients with CLL [92]. The simplest and most common diagnostic method for the detection of chromosomal rearrangements or deletion in locus 13q14.3 is to use FISH and evaluate the status of chromosomal changes in peripheral blood [93].
Dysregulation of miRNA expression significantly affects the regulation of many genes involved in cancer development. Overexpression of miRNA involved in the negative regulation of the suppressor gene can lead to the silencing of this suppressor. On the other hand, deletion of the chromosomal loci encoding the miRNA that silences the oncogene results in overexpression of this oncogene. Therefore, miRNAs themselves can act as suppressors, as well as oncogenes (oncomiRs). It is also important to remember that one miRNA molecule can regulate many genes, but also one mRNA can be targeted by several miRNAs.
Due to the changes in miRNA function and expression in cancer development, miRNAs can act as biomarkers and can be evaluated in both tumor tissue and blood derived from cancer patients.

6.1 miRNA as a Cancer Biomarker in Solid Tissues

Both fresh tissue and formalin-fixed paraffin-embedded (FFPE) tissue can be used for miRNA evaluation. It is well-known that miRNA expression profiling or single miRNA evaluation performed on FFPE tissue corresponds well to fresh frozen (FF) tissue evaluation, as miRNA molecules are more resistant to formalin than mRNA [94]. When looking for miRNA biomarkers, it should be taken into account that changes in their expression can result from CNV. Therefore, miRNA analysis can be based on both expression and/or CNV evaluation.
One of the potential prognostic biomarkers in NSCLC is the set of four miRNAs (miR-30d, miR-21, miR-17, and miR-155) and two miRNA biogenesis genes (DICER1 and DROSHA) (Table 4) [95]. In particular, miR-30d has been found to act as an oncomiR in cancer [96] and an increased copy number of miR-30d in cancer tissue (gains or amplifications vs. others) is correlated with significantly reduced survival [95] (Table 4). In contrast, it seems that miR-200b acts as a tumor suppressor: deletion or homozygous deletion of miR-200b correlates with decreased survival of patients [95]. Finally, it has been found that an increased expression of DROSHA significantly correlates with a decreased survival, and DICER1 amplification results in overexpression in cancer tissue [95]. This study suggested that CNV can be an important regulation mechanism of miRNA expression in cancer and shows a potential oncogenic role for DROSHA [95]. As was indicated by the authors, CNV (especially amplification, deletion, etc.) seems to be a very promising biomarker as DNA is more stable than RNA and is required in lower amounts to start a series. Finally, MLPA seems to be a cost effective and reliable method for copy number analysis [93].
Table 4
MicroRNA: prognostic and predictive biomarkers with diagnostic utility
miRNA
Prognostic
Predictive
Invasive/non-invasive diagnostics
Biological material
Target cancer
miR-21
+
+
Invasive/non-invasive
FFPE/blood
Multiple types of cancers [104108]
miR-30d, miR-21
+
Invasive
FFPE
Non-small cell lung cancer [95]
miR-31-3p
+
+
Invasive
FFPE
Colorectal cancer [97]
miR-106a, miR125a-5p, miR-129-3p, miR-205, miR-21, miR-29b, miR-375, miR-7
+
Invasive
FFPE
Non-small cell lung cancer [98]
miR-29a, miR-92a
+
Non-invasive
Blood
Colorectal cancer [131]
miR-506, miR-4316
+
Non-invasive
Blood
Colorectal cancer [132]
miR-126, miR-145, miR-210, miR-205-5p
+
Non-invasive
Blood
Non-small cell lung cancer [133]
miR-149-3p, miR-150-5p, miR-193a-3p
+
Non-invasive
Blood
Melanoma [134]
miR-200 family, miR-17 family
+
Non-invasive
Blood
Prostate cancer [135]
miR-17, miR-155
+
Invasive
FFPE
Non-small cell lung cancer [95]
FFPE formalin-fixed paraffin-embedded cancer tissue, miRNA microRNA
Another interesting prognostic and predictive biomarker in metastatic CRC is miR-31-3p (Table 4) [97]. The expression level of this miRNA in FF or FFPE samples has been associated with progression-free survival during anti-epidermal growth factor receptor (EGFR) antibody therapy in KRAS wild-type patients [97]. Following these data, a novel assay (MIRPREDX 31-3p) was launched in 2017 to measure the miR-31-3p expression in CRC. This test predicts which first-line chemotherapy is likely to be more beneficial to a patient with RAS wild-type metastatic CRC: anti-EGFR or anti-vascular endothelial growth factor (VEGF) treatment. The test can also be used to establish when second or further lines of anti-EGFR therapy will be beneficial versus chemotherapy alone for patients with RAS wild-type metastatic CRC [136].
Another example of harnessing translational medicine biomarkers into the clinical setting is the miRview® lung assay (Rosetta Genomics Ltd.). This miRNA signature evaluates the expression of hsa-miR-106a, hsa-miR-125a-5p, hsa-miR-129-3p, hsa-miR-205, hsa-miR-21, hsa-miR-29b, hsa-miR-375, and hsa-miR-7 in histopathology and cytology samples using real-time PCR (Table 4) [98]. Assessment of these miRNAs distinguishes squamous from non-squamous NSCLC (with 97% sensitivity and 91% specificity) and is a novel tool for differentiating primary lung cancer into four subgroups with an overall accuracy of 94%: squamous cell lung carcinoma, non-squamous cell lung carcinoma, carcinoid, and small-cell lung carcinoma [98].
Tissue miRNA profiles seem to be very valuable diagnostic biomarkers; however, one of their limitations is the invasiveness of the method used to obtain tissue samples for testing.

6.2 miRNA as a Cancer Biomarker in Biofluids

Tumor cells release miRNAs into body fluids, such as plasma, serum, urine, and saliva. Therefore, analysis of circulating miRNAs in samples of liquid biopsy provides promising biomarkers for non-invasive diagnostics in many human cancers, including colorectal, lung, breast, prostate, gastric, pancreatic, esophageal, liver, thyroid, kidney, ovarian, endometrial, and cervical cancers, as well as melanoma and rhabdomyosarcoma [99]. However, although many potential cell-free circulating miRNA biomarkers have been identified in several types of cancer, there is no proportional reflection in the number of diagnostic tests developed.
There are many miRNAs dysregulated in cancers whose changes are reflected in blood. Among the well-known miRNAs with altered expression in tumors is the let-7 family: let-7a-1, 7a-2, 7a-3, 7b, 7c, 7d, 7e, 7f-1, 7f-2, 7g, 7i, miR-98, and miR-202 [100]. The let-7 family are considered to be tumor suppressors, because selected members of this family are downregulated in melanoma, pancreatic cancer, prostate cancer, and sarcoma [101]. However, they can also be upregulated in lymphoma, mesothelioma, and breast cancer [101]. Circulating let-7 has been described as serum/plasma-based diagnostic tools in those cancers. However, those specific miRNAs that allow metastatic and non-metastatic patients to be distinguished, help to discriminate the different histological subtypes, or provide predictive information are the most diagnostically valuable.
It was found that miR-21 silences the expression of many tumor suppressors, so it functions as an oncogene (‘oncomiR’) (Table 4) [102]. This miRNA targets, for example, PDCD4, which is associated with inhibition of neoplastic transformation, cancer promotion, progression, and invasion [101105]. Among the other targets of miR-21 are BCL2, PTEN, RECK, RHOB, and TPM1 [106]. The diagnostic, predictive, and/or prognostic role of this miRNA has been described in hematological cancers, breast cancer, gastric cancer, ovarian cancer, pancreatic cancer, CRC, lung cancer, and liver cancer [104108]. Abnormal expression of miR-21 has been demonstrated in many cancers, and it has been used in commercial tests not only as a single miRNA but also in small panels.
miR-21, along with many other miRNAs, is included in the Lumira test, which is under development by Microlin Bio, Inc. Lumira is intended to help diagnose and determine the risk of developing lung cancer. Microlin Bio, Inc. is also working on the development of other tests: Omira, employing miR-484, for the screening and analysis of the chemosensitivity of ovarian cancer; Colomira, employing a miR-17 cluster for the diagnosis and prognosis of colon cancer, as well as determination of its sensitivity to therapies; and Promira, employing miR-34a, for the diagnosis and prognosis of prostate cancer.
The expression of miR-21 has also been evaluated in plasma derived from gastric cancer patients with a sensitivity of 66.5%, a specificity of 83.1%, and an AUC of 0.8 [109]. Interestingly, a combination of miR-21 with miR-106b has demonstrated a sensitivity of 69%, a specificity of 69.4%, and an AUC of 0.7 [110], while the combination of miR-21 with miR-223 and miR-218 has shown a sensitivity of 84.29%, a specificity of 92.86%, and an AUC of 0.9531 [96]. In gastric cancer, measuring the expression level of different miRNAs, including hsa-miR-21-3p, was patented in 2014 [137], and a CE IVD test—MIRXES gastric cancer kit (MiRXES, ID3AL)—has been introduced into the market for the miRNA expression profile in blood.

7 Influence of Epigenetics on Standard Therapeutic Options in Cancer

The main way to prevent deaths from CRC and to improve the cure rate is regular screening. The US Preventive Services Task Force (USPSTF) has shown that approximately 60% of CRC deaths could be avoided with periodic screening, and the average 5-year survival rate could be increased from 46% to 73% [111]. According to the guidelines of the American Cancer Society, CRC screening in people at average risk should begin at the age of 45 years. Colonoscopy is one of the visual examinations of CRC and should be performed every 10 years. For people at increased risk, screening should start earlier and be performed more often [112]. Unfortunately, colonoscopy is associated with significant discomfort and thus patients tend to avoid it. Statistics show that less than 60% of eligible adults adhere to screening recommendations [113]. It is believed that more patient-friendly approaches, such as analysis of the SEPT9 methylation level in blood, might encourage people who did not follow the screening recommendations due to the fear of this invasive examination [111]. A meta-analysis performed by Nian et al. [113] based on 25 studies assessing the diagnostic utility of the SEPT9 assay showed that pooled sensitivity was 0.71, specificity was 0.92, and the AUC was 0.88. Among the various methods and assays, Epipro Colon® 2.0 with 2/3 algorithm was the most effective in CRC detection [113].
Methylation of MGMT occurs in 35–45% of malignant gliomas (World Health Organization [WHO] grades III and IV) and in about 80% of WHO grade II gliomas. At present, radiotherapy with concomitant and adjuvant temozolomide chemotherapy is the standard treatment for glioblastoma. The presence of the methylated MGMT promoter has been seen to improve median overall survival (21.7 vs. 12.7 months) and progression-free survival compared with patients without methylation [114]. Thus, the MGMT methylation status is important when making clinical decisions in an older glioblastoma patient. In the elderly subpopulation, MGMT promoter methylation is an important biomarker for personalized treatment strategies. It should be taken into account that chemoradiotherapy including temozolomide might be too toxic for the elderly. Overall survival in methylated patients was better when temozolomide treatment was used, whereas radiotherapy alone was more effective in unmethylated patients [114, 115].
Due to the high risk of a standard brain biopsy, the assessment of MGMT methylation in a non-invasive liquid biopsy seems to be an attractive alternative. Unfortunately, the use of serum circulating tumor DNA (ctDNA) analysis for tumors of the nervous system is difficult due to the impermeability of the blood–brain barrier. Studies show that the sensitivity of detection of the MGMT promoter methylation in blood serum, expressed as the percentage of positive serum samples among patients with MGMT methylation detected in tissue samples, is low (37.3%), but the use of CSF can increase the sensitivity of detection to 65.0%. However, it seems that the overall sensitivity of liquid biopsy (including CSF) is significantly lower than that of tissue samples [116]. Therefore, detection of MGMT promoter methylation in tissue is more reasonable for diagnostic purposes.

8 Epigenetic Therapy: Clinical Trials

Targeting enzymatic regulators responsible for epigenetic events holds promise for modulating the transcriptional regulation of genes involved in cancer [117]. Additionally, reversal of epigenetic changes represents a potential target for novel preventive and therapeutic strategies, as well as medication design [118]. Epigenetic therapy primarily involves DNA methylation inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) [119]. To date, five epidrugs have been approved by the US Food and Drug Administration (FDA) (two DNMTi and three HDACi) and are described as the first generation of epigenetic inhibitors in cancer treatment. At present, 21 clinical trials using epigenetic drugs as epigenetic cancer therapy are registered at ClinicalTrials.gov. Most of them involve lung cancer, cervical carcinoma, CRC, and pancreatic adenocarcinoma.
DNMTi entered clinical trials as classical anticancer agents more than 40 years ago. The role of DNMTi is to remove the hypermethylation of tumor suppressor genes [120]. DNMTi are classified into two categories: nucleoside and non-nucleoside inhibitors. The cytosine analogs, azacytidine (5-azacytidine) and decitabine (5-aza-2ʹ-deoxycytidine), belong to the first-generation epigenetic nucleoside inhibitors that intercalate with the DNA. These epidrugs are approved by the FDA and used in the treatment for myeloid leukemias [120]. In recent years second-generation nucleoside DNMTi have been developed and preclinical and clinical studies are ongoing for their use as therapy against advanced solid tumors and leukemias [121].
HDACi reduce oncogene transcription and angiogenesis, induce cancer cell cycle arrest, and modulate immune response [122]. The use of HDACi has been clinically validated in cancer treatment and four drugs have been approved by the FDA: vorinostat, romidepsin, panobinostat, and belinostat [120]. Belinostat has been approved as an epigenetic compound for the treatment of peripheral T cell lymphoma, vorinostat and romidepsin have been approved for the treatment of cutaneous manifestations in patients with cutaneous T cell lymphoma (CTCL), and panobinostat in combination with the proteasome inhibitor bortezomib has been approved for use in patients with multiple myeloma [120].
Epigenetic drugs have shown very modest antitumor efficacy, with some responses seen in a small percentage of cancer patients (3% of patients responded to epidrugs in a single therapy, 20% of patients responded to epidrugs in combination with other chemotherapies) [123]. Additionally, epidrugs used in cancer treatment will potentially have genome-wide effects, such as uncontrolled upregulation of unwanted genes, e.g., oncogenes [123]. Therefore, development of therapies targeting specific epigenetic defects in cancer can provide a novel approach to prevent such unwanted events.
Therapeutic strategies for cancer will exhibit the greatest effectiveness with treatment approaches based not on single therapies but on juxtaposition of mutational events and epigenetic alterations [120].

9 Summary

Epigenetic modifications of DNA and gene regulation by miRNA are the hope and promise of novel biomarkers for early cancer detection, prediction, prognosis, and response to treatment. The most commonly used epigenetic biomarkers were first investigated in solid tissues, but acquiring this material using invasive techniques limits the potential of early cancer detection and screening programs. The ideal sampling source is blood in which circulating free DNA and miRNAs can be measured using minimally invasive techniques, and one which can be used in all patients—those at both minimal and high risk [118]. Obviously, the phases of development of biomarkers, from quality control, analytical and clinical validation to the approved biomarker ready for clinical use, are expensive and time-consuming, but this work can be conducted through interdisciplinary collaboration between researchers, diagnosticians, and diagnostics companies.
Alteration of DNA methylation, histone modifications, and miRNAs often helps to better differentiate tumor subtypes and bring new prognostic information related to patient survival in relation to age, sex, etc. On the other hand, there are also a few epigenetic biomarkers that predict response to chemotherapeutic agents, the most common being MGMT promoter methylation, which predicts response to temozolomide. International epigenetic projects have allowed not only better characterization of novel biomarkers, but have also opened a new strategy in pharmacology. Reversal of epigenetic changes represents a potential target for novel preventive and therapeutic strategies, as well as medication design [118]. Epigenetic therapy primarily involves DNMTi and HDACi that have been clinically validated in cancer treatment [119]. Therapeutic strategies for cancer will exhibit the greatest efficiency with treatment approaches based not only on single therapies but on juxtaposition of mutational events and epigenetic alterations [120].

Compliance with Ethical Standards

Funding

This study was supported by grants from the International Centre for Genetic Engineering and Biotechnology—POL 13-01 contract CRP 13/024.

Conflict of interest

Katarzyna Kamińska, Ewelina Nalejska, Marta Kubiak, Joanna Wojtysiak, Łukasz Żołna, Janusz Kowalewski, and Marzena Anna Lewandowska have no conflicts of interest to declare.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Esteller M. The necessity of a human epigenome project. Carcinogenesis. 2006;27(6):1121–5. Esteller M. The necessity of a human epigenome project. Carcinogenesis. 2006;27(6):1121–5.
2.
3.
Zurück zum Zitat Banno K, Kisu I, Yanokura M, Tsuji K, Masuda K, Ueki A, et al. Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review). Int J Oncol. 2012;41(3):793–7.CrossRefPubMedPubMedCentral Banno K, Kisu I, Yanokura M, Tsuji K, Masuda K, Ueki A, et al. Epimutation and cancer: a new carcinogenic mechanism of Lynch syndrome (Review). Int J Oncol. 2012;41(3):793–7.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Poole RL, Docherty LE, Al Sayegh A, Caliebe A, Turner C, Baple E, et al. Targeted methylation testing of a patient cohort broadens the epigenetic and clinical description of imprinting disorders. Am J Med Genet A. 2013;161A(9):2174–82.CrossRefPubMed Poole RL, Docherty LE, Al Sayegh A, Caliebe A, Turner C, Baple E, et al. Targeted methylation testing of a patient cohort broadens the epigenetic and clinical description of imprinting disorders. Am J Med Genet A. 2013;161A(9):2174–82.CrossRefPubMed
5.
Zurück zum Zitat Ehrhart F, Janssen KJM, Coort SL, Evelo CT, Curfs LMG. Prader-Willi syndrome and Angelman syndrome: visualisation of the molecular pathways for two chromosomal disorders. World J Biol Psychiatry. 2018;1:1–13.CrossRef Ehrhart F, Janssen KJM, Coort SL, Evelo CT, Curfs LMG. Prader-Willi syndrome and Angelman syndrome: visualisation of the molecular pathways for two chromosomal disorders. World J Biol Psychiatry. 2018;1:1–13.CrossRef
6.
Zurück zum Zitat Gardner RJ, Mackay DJ, Mungall AJ, Polychronakos C, Siebert R, Shield JP, et al. An imprinted locus associated with transient neonatal diabetes mellitus. Hum Mol Genet. 2000;9(4):589–96.CrossRefPubMed Gardner RJ, Mackay DJ, Mungall AJ, Polychronakos C, Siebert R, Shield JP, et al. An imprinted locus associated with transient neonatal diabetes mellitus. Hum Mol Genet. 2000;9(4):589–96.CrossRefPubMed
7.
Zurück zum Zitat Costa FF. Epigenomics in cancer management. Cancer Manag Res. 2010;27(2):255–65.CrossRef Costa FF. Epigenomics in cancer management. Cancer Manag Res. 2010;27(2):255–65.CrossRef
8.
Zurück zum Zitat Bae J-B. Perspectives of international human epigenome consortium. Genom Inform. 2013;11(1):7–14.CrossRef Bae J-B. Perspectives of international human epigenome consortium. Genom Inform. 2013;11(1):7–14.CrossRef
9.
Zurück zum Zitat Jones PA, Martienssen R. A blueprint for a human epigenome project: the AACR human epigenome workshop. Cancer Res. 2005;65(24):11241–6.CrossRefPubMed Jones PA, Martienssen R. A blueprint for a human epigenome project: the AACR human epigenome workshop. Cancer Res. 2005;65(24):11241–6.CrossRefPubMed
10.
Zurück zum Zitat Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005;2(Suppl 1):S4–11.CrossRefPubMed Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005;2(Suppl 1):S4–11.CrossRefPubMed
11.
Zurück zum Zitat Søes S, Daugaard IL, Sørensen BS, Carus A, Mattheisen M, Alsner J, et al. Hypomethylation and increased expression of the putative oncogene ELMO3 are associated with lung cancer development and metastases formation. Oncoscience. 2014;1(5):367–74.CrossRefPubMedPubMedCentral Søes S, Daugaard IL, Sørensen BS, Carus A, Mattheisen M, Alsner J, et al. Hypomethylation and increased expression of the putative oncogene ELMO3 are associated with lung cancer development and metastases formation. Oncoscience. 2014;1(5):367–74.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301(5895):89–92.CrossRefPubMed Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301(5895):89–92.CrossRefPubMed
13.
Zurück zum Zitat Curtin K, Slattery ML, Samowitz WS. CpG island methylation in colorectal cancer: past, present and future. Patholog Res Int. 2011;12(2011):902674. Curtin K, Slattery ML, Samowitz WS. CpG island methylation in colorectal cancer: past, present and future. Patholog Res Int. 2011;12(2011):902674.
14.
Zurück zum Zitat Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999;96(15):8681–6.CrossRefPubMed Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999;96(15):8681–6.CrossRefPubMed
15.
Zurück zum Zitat Weiss G, Schlegel A, Kottwitz D, König T, Tetzner R. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J Thorac Oncol. 2017;12(1):77–84.CrossRefPubMedPubMedCentral Weiss G, Schlegel A, Kottwitz D, König T, Tetzner R. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J Thorac Oncol. 2017;12(1):77–84.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38(7):787–93.CrossRefPubMed Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38(7):787–93.CrossRefPubMed
17.
Zurück zum Zitat Jass JR. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology. 2007;50(1):113–30.CrossRefPubMed Jass JR. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology. 2007;50(1):113–30.CrossRefPubMed
18.
19.
Zurück zum Zitat Giardiello FM, Allen JI, Axilbund JE, Boland CR, Burke CA, Burt RW, et al. Guidelines on genetic evaluation and management of Lynch syndrome: a consensus statement by the U.S. Multi-Society Task Force on Colorectal Cancer. Gastrointest Endosc. 2014;80(2):197–220.CrossRefPubMed Giardiello FM, Allen JI, Axilbund JE, Boland CR, Burke CA, Burt RW, et al. Guidelines on genetic evaluation and management of Lynch syndrome: a consensus statement by the U.S. Multi-Society Task Force on Colorectal Cancer. Gastrointest Endosc. 2014;80(2):197–220.CrossRefPubMed
20.
Zurück zum Zitat Pérez-Carbonell L, Alenda C, Payá A, Castillejo A, Barberá VM, Guillén C, et al. Methylation analysis of MLH1 improves the selection of patients for genetic testing in Lynch syndrome. J Mol Diagn. 2010;12(4):498–504.CrossRefPubMedPubMedCentral Pérez-Carbonell L, Alenda C, Payá A, Castillejo A, Barberá VM, Guillén C, et al. Methylation analysis of MLH1 improves the selection of patients for genetic testing in Lynch syndrome. J Mol Diagn. 2010;12(4):498–504.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Castillejo A, Hernández-Illán E, Rodriguez-Soler M, Pérez-Carbonell L, Egoavil C, Barberá VM, et al. Prevalence of MLH1 constitutional epimutations as a cause of Lynch syndrome in unselected versus selected consecutive series of patients with colorectal cancer. J Med Genet. 2015;52(7):498–502.CrossRefPubMed Castillejo A, Hernández-Illán E, Rodriguez-Soler M, Pérez-Carbonell L, Egoavil C, Barberá VM, et al. Prevalence of MLH1 constitutional epimutations as a cause of Lynch syndrome in unselected versus selected consecutive series of patients with colorectal cancer. J Med Genet. 2015;52(7):498–502.CrossRefPubMed
22.
Zurück zum Zitat Ozer O, Bilezikci B, Aktas S, Sahin FI. Methylation profile analysis of DNA repair genes in hepatocellular carcinoma with MS-MLPA. Diagn Mol Pathol. 2013;22(4):222–7.CrossRefPubMed Ozer O, Bilezikci B, Aktas S, Sahin FI. Methylation profile analysis of DNA repair genes in hepatocellular carcinoma with MS-MLPA. Diagn Mol Pathol. 2013;22(4):222–7.CrossRefPubMed
23.
Zurück zum Zitat Newton K, Jorgensen NM, Wallace AJ, Buchanan DD, Lalloo F, McMahon RF, et al. Tumour MLH1 promoter region methylation testing is an effective prescreen for Lynch Syndrome (HNPCC). J Med Genet. 2014;51(12):789–96.CrossRefPubMedPubMedCentral Newton K, Jorgensen NM, Wallace AJ, Buchanan DD, Lalloo F, McMahon RF, et al. Tumour MLH1 promoter region methylation testing is an effective prescreen for Lynch Syndrome (HNPCC). J Med Genet. 2014;51(12):789–96.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13(7):707–15.CrossRefPubMed Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13(7):707–15.CrossRefPubMed
25.
Zurück zum Zitat Cabrini G, Fabbri E, Lo Nigro C, Dechecchi MC, Gambari R. Regulation of expression of O6-methylguanine-DNA methyltransferase and the treatment of glioblastoma (Review). Int J Oncol. 2015;47(2):417–28.CrossRefPubMedPubMedCentral Cabrini G, Fabbri E, Lo Nigro C, Dechecchi MC, Gambari R. Regulation of expression of O6-methylguanine-DNA methyltransferase and the treatment of glioblastoma (Review). Int J Oncol. 2015;47(2):417–28.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Coulondre C, Miller JH. Genetic studies of the lac repressor. IV. Mutagenic specificity in the lacI gene of Escherichia coli. J Mol Biol. 1977;117(3):577–606.CrossRefPubMed Coulondre C, Miller JH. Genetic studies of the lac repressor. IV. Mutagenic specificity in the lacI gene of Escherichia coli. J Mol Biol. 1977;117(3):577–606.CrossRefPubMed
27.
Zurück zum Zitat Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999;59(4):793–7.PubMed Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 1999;59(4):793–7.PubMed
28.
Zurück zum Zitat Chakravarti A, Erkkinen MG, Nestler U, Stupp R, Mehta M, Aldape K, et al. Temozolomide-mediated radiation enhancement in glioblastoma: a report on underlying mechanisms. Clin Cancer Res. 2006;12(15):4738–46.CrossRefPubMed Chakravarti A, Erkkinen MG, Nestler U, Stupp R, Mehta M, Aldape K, et al. Temozolomide-mediated radiation enhancement in glioblastoma: a report on underlying mechanisms. Clin Cancer Res. 2006;12(15):4738–46.CrossRefPubMed
29.
Zurück zum Zitat Weller M, Pfister SM, Wick W, Hegi ME, Reifenberger G, Stupp R. Molecular neuro-oncology in clinical practice: a new horizon. Lancet Oncol. 2013;14(9):e370–9.CrossRefPubMed Weller M, Pfister SM, Wick W, Hegi ME, Reifenberger G, Stupp R. Molecular neuro-oncology in clinical practice: a new horizon. Lancet Oncol. 2013;14(9):e370–9.CrossRefPubMed
30.
Zurück zum Zitat Roszkowski K, Furtak J, Zurawski B, Szylberg T, Lewandowska MA. Potential role of methylation marker in glioma supporting clinical decisions. Int J Mol Sci. 2016;17(11):1876.CrossRefPubMedCentral Roszkowski K, Furtak J, Zurawski B, Szylberg T, Lewandowska MA. Potential role of methylation marker in glioma supporting clinical decisions. Int J Mol Sci. 2016;17(11):1876.CrossRefPubMedCentral
31.
Zurück zum Zitat Greger V, Passarge E, Höpping W, Messmer E, Horsthemke B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Hum Genet. 1989;83(2):155–8.CrossRefPubMed Greger V, Passarge E, Höpping W, Messmer E, Horsthemke B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Hum Genet. 1989;83(2):155–8.CrossRefPubMed
32.
Zurück zum Zitat Simpson DJ, Hibberts NA, McNicol AM, Clayton RN, Farrell WE. Loss of pRb expression in pituitary adenomas is associated with methylation of the RB1 CpG island. Cancer Res. 2000;60(5):1211–6.PubMed Simpson DJ, Hibberts NA, McNicol AM, Clayton RN, Farrell WE. Loss of pRb expression in pituitary adenomas is associated with methylation of the RB1 CpG island. Cancer Res. 2000;60(5):1211–6.PubMed
33.
Zurück zum Zitat Ohtani-Fujita N, Dryja TP, Rapaport JM, Fujita T, Matsumura S, Ozasa K, et al. Hypermethylation in the retinoblastoma gene is associated with unilateral, sporadic retinoblastoma. Cancer Genet Cytogenet. 1997;98(1):43–9.CrossRefPubMed Ohtani-Fujita N, Dryja TP, Rapaport JM, Fujita T, Matsumura S, Ozasa K, et al. Hypermethylation in the retinoblastoma gene is associated with unilateral, sporadic retinoblastoma. Cancer Genet Cytogenet. 1997;98(1):43–9.CrossRefPubMed
34.
Zurück zum Zitat Livide G, Epistolato MC, Amenduni M, Disciglio V, Marozza A, Mencarelli MA, et al. Epigenetic and copy number variation analysis in retinoblastoma by MS-MLPA. Pathol Oncol Res. 2012;18(3):703–12.CrossRefPubMed Livide G, Epistolato MC, Amenduni M, Disciglio V, Marozza A, Mencarelli MA, et al. Epigenetic and copy number variation analysis in retinoblastoma by MS-MLPA. Pathol Oncol Res. 2012;18(3):703–12.CrossRefPubMed
35.
Zurück zum Zitat Stewart GD, Van Neste L, Delvenne P, Delrée P, Delga A, McNeill SA, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol. 2013;189(3):1110–6.CrossRefPubMed Stewart GD, Van Neste L, Delvenne P, Delrée P, Delga A, McNeill SA, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol. 2013;189(3):1110–6.CrossRefPubMed
36.
Zurück zum Zitat Partin AW, Van Neste L, Klein EA, Marks LS, Gee JR, Troyer DA, et al. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol. 2014;192(4):1081–7.CrossRefPubMedPubMedCentral Partin AW, Van Neste L, Klein EA, Marks LS, Gee JR, Troyer DA, et al. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol. 2014;192(4):1081–7.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Burrows JF, Chanduloy S, McIlhatton MA, Nagar H, Yeates K, Donaghy P, et al. Altered expression of the septin gene, SEPT9, in ovarian neoplasia. J Pathol. 2003;201(4):581–8.CrossRefPubMed Burrows JF, Chanduloy S, McIlhatton MA, Nagar H, Yeates K, Donaghy P, et al. Altered expression of the septin gene, SEPT9, in ovarian neoplasia. J Pathol. 2003;201(4):581–8.CrossRefPubMed
38.
Zurück zum Zitat Tóth K, Galamb O, Spisák S, Wichmann B, Sipos F, Valcz G, et al. The influence of methylated septin 9 gene on RNA and protein level in colorectal cancer. Pathol Oncol Res. 2011;17(3):503–9. Tóth K, Galamb O, Spisák S, Wichmann B, Sipos F, Valcz G, et al. The influence of methylated septin 9 gene on RNA and protein level in colorectal cancer. Pathol Oncol Res. 2011;17(3):503–9.
39.
Zurück zum Zitat Wang Y, Chen P-M, Liu R-B. Advance in plasma SEPT9 gene methylation assay for colorectal cancer early detection. World J Gastrointest Oncol. 2018;10(1):15–22.CrossRefPubMedPubMedCentral Wang Y, Chen P-M, Liu R-B. Advance in plasma SEPT9 gene methylation assay for colorectal cancer early detection. World J Gastrointest Oncol. 2018;10(1):15–22.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Powrózek T, Krawczyk P, Kucharczyk T, Milanowski J. Septin 9 promoter region methylation in free circulating DNA-potential role in noninvasive diagnosis of lung cancer: preliminary report. Med Oncol. 2014;31(4):917.CrossRefPubMedPubMedCentral Powrózek T, Krawczyk P, Kucharczyk T, Milanowski J. Septin 9 promoter region methylation in free circulating DNA-potential role in noninvasive diagnosis of lung cancer: preliminary report. Med Oncol. 2014;31(4):917.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Nikolaidis G, Raji OY, Markopoulou S, Gosney JR, Bryan J, Warburton C, et al. DNA methylation biomarkers offer improved diagnostic efficiency in lung cancer. Cancer Res. 2012;72(22):5692–701.CrossRefPubMedPubMedCentral Nikolaidis G, Raji OY, Markopoulou S, Gosney JR, Bryan J, Warburton C, et al. DNA methylation biomarkers offer improved diagnostic efficiency in lung cancer. Cancer Res. 2012;72(22):5692–701.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Schmidt B, Liebenberg V, Dietrich D, Schlegel T, Kneip C, Seegebarth A, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer based on bronchial aspirates. BMC Cancer. 2010;3(10):600.CrossRef Schmidt B, Liebenberg V, Dietrich D, Schlegel T, Kneip C, Seegebarth A, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer based on bronchial aspirates. BMC Cancer. 2010;3(10):600.CrossRef
44.
Zurück zum Zitat Ilse P, Biesterfeld S, Pomjanski N, Fink C, Schramm M. SHOX2 DNA methylation is a tumour marker in pleural effusions. Cancer Genomics Proteomics. 2013;10(5):217-23. Ilse P, Biesterfeld S, Pomjanski N, Fink C, Schramm M. SHOX2 DNA methylation is a tumour marker in pleural effusions. Cancer Genomics Proteomics. 2013;10(5):217-23.
45.
Zurück zum Zitat Dietrich D, Jung M, Puetzer S, Leisse A, Holmes EE, Meller S, et al. Diagnostic and prognostic value of SHOX2 and SEPT9 DNA methylation and cytology in benign, paramalignant and malignant pleural effusions. PloS One. 2013;8(12):e84225.CrossRefPubMedPubMedCentral Dietrich D, Jung M, Puetzer S, Leisse A, Holmes EE, Meller S, et al. Diagnostic and prognostic value of SHOX2 and SEPT9 DNA methylation and cytology in benign, paramalignant and malignant pleural effusions. PloS One. 2013;8(12):e84225.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Li Y-W, Kong F-M, Zhou J-P, Dong M. Aberrant promoter methylation of the vimentin gene may contribute to colorectal carcinogenesis: a meta-analysis. Tumour Biol. 2014;35(7):6783–90.CrossRefPubMed Li Y-W, Kong F-M, Zhou J-P, Dong M. Aberrant promoter methylation of the vimentin gene may contribute to colorectal carcinogenesis: a meta-analysis. Tumour Biol. 2014;35(7):6783–90.CrossRefPubMed
47.
Zurück zum Zitat Itzkowitz S, Brand R, Jandorf L, Durkee K, Millholland J, Rabeneck L, et al. A simplified, noninvasive stool DNA test for colorectal cancer detection. Am J Gastroenterol. 2008;103(11):2862–70.CrossRefPubMed Itzkowitz S, Brand R, Jandorf L, Durkee K, Millholland J, Rabeneck L, et al. A simplified, noninvasive stool DNA test for colorectal cancer detection. Am J Gastroenterol. 2008;103(11):2862–70.CrossRefPubMed
49.
Zurück zum Zitat Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12–27.CrossRefPubMed Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12–27.CrossRefPubMed
50.
Zurück zum Zitat Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature. 2011;473(7345):43–9.CrossRefPubMedPubMedCentral Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature. 2011;473(7345):43–9.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Song JS, Kim YS, Kim DK, Park SI, Jang SJ. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol Int. 2012;62(3):182–90.CrossRefPubMed Song JS, Kim YS, Kim DK, Park SI, Jang SJ. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol Int. 2012;62(3):182–90.CrossRefPubMed
53.
Zurück zum Zitat Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature. 2005;435(7046):1262–6.CrossRefPubMed Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature. 2005;435(7046):1262–6.CrossRefPubMed
54.
Zurück zum Zitat Ellinger J, Schneider A-C, Bachmann A, Kristiansen G, Müller SC, Rogenhofer S. Evaluation of global histone acetylation levels in bladder cancer patients. Anticancer Res. 2016;36(8):3961–4.PubMed Ellinger J, Schneider A-C, Bachmann A, Kristiansen G, Müller SC, Rogenhofer S. Evaluation of global histone acetylation levels in bladder cancer patients. Anticancer Res. 2016;36(8):3961–4.PubMed
55.
Zurück zum Zitat Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;102(30):10604–9.CrossRefPubMed Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;102(30):10604–9.CrossRefPubMed
56.
Zurück zum Zitat Elsheikh SE, Green AR, Rakha EA, Powe DG, Ahmed RA, Collins HM, et al. Global histone modifications in breast cancer correlate with tumor phenotypes, prognostic factors, and patient outcome. Cancer Res. 2009;69(9):3802–9.CrossRefPubMed Elsheikh SE, Green AR, Rakha EA, Powe DG, Ahmed RA, Collins HM, et al. Global histone modifications in breast cancer correlate with tumor phenotypes, prognostic factors, and patient outcome. Cancer Res. 2009;69(9):3802–9.CrossRefPubMed
57.
Zurück zum Zitat Monteiro FL, Baptista T, Amado F, Vitorino R, Jerónimo C, Helguero LA. Expression and functionality of histone H2A variants in cancer. Oncotarget. 2014;5(11):3428–43.CrossRefPubMedPubMedCentral Monteiro FL, Baptista T, Amado F, Vitorino R, Jerónimo C, Helguero LA. Expression and functionality of histone H2A variants in cancer. Oncotarget. 2014;5(11):3428–43.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Yörüker EE, Holdenrieder S, Gezer U. Potential of circulating nucleosome-associated histone modifications in cancer. Transl Cancer Res. 2017;7(2):S185–91. Yörüker EE, Holdenrieder S, Gezer U. Potential of circulating nucleosome-associated histone modifications in cancer. Transl Cancer Res. 2017;7(2):S185–91.
59.
Zurück zum Zitat McAnena P, Brown JAL, Kerin MJ. Circulating nucleosomes and nucleosome modifications as biomarkers in cancer. Cancers (Basel). 2017;9(1):5.CrossRefPubMedCentral McAnena P, Brown JAL, Kerin MJ. Circulating nucleosomes and nucleosome modifications as biomarkers in cancer. Cancers (Basel). 2017;9(1):5.CrossRefPubMedCentral
60.
Zurück zum Zitat Holdenrieder S, Stieber P, Bodenmüller H, Busch M, Fertig G, Fürst H, et al. Nucleosomes in serum of patients with benign and malignant diseases. Int J Cancer. 2001;95(2):114–20.CrossRefPubMed Holdenrieder S, Stieber P, Bodenmüller H, Busch M, Fertig G, Fürst H, et al. Nucleosomes in serum of patients with benign and malignant diseases. Int J Cancer. 2001;95(2):114–20.CrossRefPubMed
61.
Zurück zum Zitat Stoetzer OJ, Fersching DMI, Salat C, Steinkohl O, Gabka CJ, Hamann U, et al. Prediction of response to neoadjuvant chemotherapy in breast cancer patients by circulating apoptotic biomarkers nucleosomes, DNAse, cytokeratin-18 fragments and survivin. Cancer Lett. 2013;336(1):140–8.CrossRefPubMed Stoetzer OJ, Fersching DMI, Salat C, Steinkohl O, Gabka CJ, Hamann U, et al. Prediction of response to neoadjuvant chemotherapy in breast cancer patients by circulating apoptotic biomarkers nucleosomes, DNAse, cytokeratin-18 fragments and survivin. Cancer Lett. 2013;336(1):140–8.CrossRefPubMed
62.
Zurück zum Zitat Holdenrieder S, Stieber P, von Pawel J, Raith H, Nagel D, Feldmann K, et al. Circulating nucleosomes predict the response to chemotherapy in patients with advanced non-small cell lung cancer. Clin Cancer Res. 2004;10(18 Pt 1):5981–7.CrossRefPubMed Holdenrieder S, Stieber P, von Pawel J, Raith H, Nagel D, Feldmann K, et al. Circulating nucleosomes predict the response to chemotherapy in patients with advanced non-small cell lung cancer. Clin Cancer Res. 2004;10(18 Pt 1):5981–7.CrossRefPubMed
63.
Zurück zum Zitat Fahmueller YN, Nagel D, Hoffmann R-T, Tatsch K, Jakobs T, Stieber P, et al. Predictive and prognostic value of circulating nucleosomes and serum biomarkers in patients with metastasized colorectal cancer undergoing selective internal radiation therapy. BMC Cancer. 2012;4(12):5.CrossRef Fahmueller YN, Nagel D, Hoffmann R-T, Tatsch K, Jakobs T, Stieber P, et al. Predictive and prognostic value of circulating nucleosomes and serum biomarkers in patients with metastasized colorectal cancer undergoing selective internal radiation therapy. BMC Cancer. 2012;4(12):5.CrossRef
64.
Zurück zum Zitat Gezer U, Holdenrieder S. Post-translational histone modifications in circulating nucleosomes as new biomarkers in colorectal cancer. Vivo. 2014;28(3):287–92. Gezer U, Holdenrieder S. Post-translational histone modifications in circulating nucleosomes as new biomarkers in colorectal cancer. Vivo. 2014;28(3):287–92.
65.
Zurück zum Zitat Gezer U, Yörüker EE, Keskin M, Kulle CB, Dharuman Y, Holdenrieder S. Histone methylation marks on circulating nucleosomes as novel blood-based biomarker in colorectal cancer. Int J Mol Sci. 2015;16(12):29654–62.CrossRefPubMedPubMedCentral Gezer U, Yörüker EE, Keskin M, Kulle CB, Dharuman Y, Holdenrieder S. Histone methylation marks on circulating nucleosomes as novel blood-based biomarker in colorectal cancer. Int J Mol Sci. 2015;16(12):29654–62.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Thålin C, Lundström S, Seignez C, Daleskog M, Lundström A, Henriksson P, et al. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PloS One. 2018;13(1):e0191231.CrossRefPubMedPubMedCentral Thålin C, Lundström S, Seignez C, Daleskog M, Lundström A, Henriksson P, et al. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PloS One. 2018;13(1):e0191231.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Versteege I, Sévenet N, Lange J, Rousseau-Merck MF, Ambros P, Handgretinger R, et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998;394(6689):203–6.CrossRefPubMed Versteege I, Sévenet N, Lange J, Rousseau-Merck MF, Ambros P, Handgretinger R, et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998;394(6689):203–6.CrossRefPubMed
69.
Zurück zum Zitat Swensen JJ, Keyser J, Coffin CM, Biegel JA, Viskochil DH, Williams MS. Familial occurrence of schwannomas and malignant rhabdoid tumour associated with a duplication in SMARCB1. J Med Genet. 2009;46(1):68–72.CrossRefPubMedPubMedCentral Swensen JJ, Keyser J, Coffin CM, Biegel JA, Viskochil DH, Williams MS. Familial occurrence of schwannomas and malignant rhabdoid tumour associated with a duplication in SMARCB1. J Med Genet. 2009;46(1):68–72.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Torchia J, Golbourn B, Feng S, Ho KC, Sin-Chan P, Vasiljevic A, et al. Integrated (epi)-genomic analyses identify subgroup-specific therapeutic targets in CNS rhabdoid tumors. Cancer Cell. 2016;30(6):891–908.CrossRefPubMedPubMedCentral Torchia J, Golbourn B, Feng S, Ho KC, Sin-Chan P, Vasiljevic A, et al. Integrated (epi)-genomic analyses identify subgroup-specific therapeutic targets in CNS rhabdoid tumors. Cancer Cell. 2016;30(6):891–908.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Wiegand KC, Lee AF, Al-Agha OM, Chow C, Kalloger SE, Scott DW, et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J Pathol. 2011;224(3):328–33.CrossRefPubMed Wiegand KC, Lee AF, Al-Agha OM, Chow C, Kalloger SE, Scott DW, et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J Pathol. 2011;224(3):328–33.CrossRefPubMed
73.
Zurück zum Zitat Zhang L, Lu Z, Zhao Q, Huang J, Shen H, Zhang Z. Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI-grafted graphene oxide. Small. 2011;7(4):460–4.CrossRefPubMed Zhang L, Lu Z, Zhao Q, Huang J, Shen H, Zhang Z. Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI-grafted graphene oxide. Small. 2011;7(4):460–4.CrossRefPubMed
74.
Zurück zum Zitat Cornen S, Adélaide J, Bertucci F, Finetti P, Guille A, Birnbaum D, et al. Mutations and deletions of ARID1A in breast tumors. Oncogene. 2012;16(31):4255–6.CrossRef Cornen S, Adélaide J, Bertucci F, Finetti P, Guille A, Birnbaum D, et al. Mutations and deletions of ARID1A in breast tumors. Oncogene. 2012;16(31):4255–6.CrossRef
75.
Zurück zum Zitat Huang J, Zhao Y-L, Li Y, Fletcher JA, Xiao S. Genomic and functional evidence for an ARID1A tumor suppressor role. Genes Chromosomes Cancer. 2007;46(8):745–50.CrossRefPubMed Huang J, Zhao Y-L, Li Y, Fletcher JA, Xiao S. Genomic and functional evidence for an ARID1A tumor suppressor role. Genes Chromosomes Cancer. 2007;46(8):745–50.CrossRefPubMed
76.
Zurück zum Zitat Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44(6):694–8.CrossRefPubMedPubMedCentral Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44(6):694–8.CrossRefPubMedPubMedCentral
77.
Zurück zum Zitat Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet. 2011;43(12):1219–23.CrossRefPubMed Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet. 2011;43(12):1219–23.CrossRefPubMed
78.
Zurück zum Zitat Sausen M, Leary RJ, Jones S, Wu J, Reynolds CP, Liu X, et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet. 2013;45(1):12–7.CrossRefPubMed Sausen M, Leary RJ, Jones S, Wu J, Reynolds CP, Liu X, et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet. 2013;45(1):12–7.CrossRefPubMed
79.
Zurück zum Zitat Takao C, Morikawa A, Ohkubo H, Kito Y, Saigo C, Sakuratani T, et al. Downregulation of ARID1A, a component of the SWI/SNF chromatin remodeling complex, in breast cancer. J Cancer. 2017;8(1):1–8.CrossRefPubMedPubMedCentral Takao C, Morikawa A, Ohkubo H, Kito Y, Saigo C, Sakuratani T, et al. Downregulation of ARID1A, a component of the SWI/SNF chromatin remodeling complex, in breast cancer. J Cancer. 2017;8(1):1–8.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Cho HD, Lee JE, Jung HY, Oh MH, Lee JH, Jang SH, et al. Loss of tumor suppressor ARID1A protein expression correlates with poor prognosis in patients with primary breast cancer. J Breast Cancer. 2015;18(4):339–46.CrossRefPubMedPubMedCentral Cho HD, Lee JE, Jung HY, Oh MH, Lee JH, Jang SH, et al. Loss of tumor suppressor ARID1A protein expression correlates with poor prognosis in patients with primary breast cancer. J Breast Cancer. 2015;18(4):339–46.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Yang L, Wei S, Zhao R, Wu Y, Qiu H, Xiong H. Loss of ARID1A expression predicts poor survival prognosis in gastric cancer: a systematic meta-analysis from 14 studies. Sci Rep. 2016;29(6):28919.CrossRef Yang L, Wei S, Zhao R, Wu Y, Qiu H, Xiong H. Loss of ARID1A expression predicts poor survival prognosis in gastric cancer: a systematic meta-analysis from 14 studies. Sci Rep. 2016;29(6):28919.CrossRef
82.
Zurück zum Zitat Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 2010;363(16):1532–43.CrossRefPubMedPubMedCentral Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 2010;363(16):1532–43.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Kubiak M, Lewandowska MA. Can chromatin conformation technologies bring light into human molecular pathology? Acta Biochim Pol. 2015;62(3):483–9.CrossRefPubMed Kubiak M, Lewandowska MA. Can chromatin conformation technologies bring light into human molecular pathology? Acta Biochim Pol. 2015;62(3):483–9.CrossRefPubMed
85.
Zurück zum Zitat Herold M, Bartkuhn M, Renkawitz R. CTCF: insights into insulator function during development. Development. 2012;139(6):1045–57.CrossRefPubMed Herold M, Bartkuhn M, Renkawitz R. CTCF: insights into insulator function during development. Development. 2012;139(6):1045–57.CrossRefPubMed
87.
Zurück zum Zitat Wang XQD, Dostie J. Chromosome folding and its regulation in health and disease. Curr Opin Genet Dev. 2017;43:23–30.CrossRefPubMed Wang XQD, Dostie J. Chromosome folding and its regulation in health and disease. Curr Opin Genet Dev. 2017;43:23–30.CrossRefPubMed
88.
Zurück zum Zitat Liu J, Ding Z, Li G, Tang L, Xu Y, Luo H, et al. Identification and validation of colorectal neoplasia-specific methylation biomarkers based on CTCF-binding sites. Oncotarget. 2017;8(69):114183–94.CrossRefPubMedPubMedCentral Liu J, Ding Z, Li G, Tang L, Xu Y, Luo H, et al. Identification and validation of colorectal neoplasia-specific methylation biomarkers based on CTCF-binding sites. Oncotarget. 2017;8(69):114183–94.CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Green AR, Krivinskas S, Young P, Rakha EA, Paish EC, Powe DG, et al. Loss of expression of chromosome 16q genes DPEP1 and CTCF in lobular carcinoma in situ of the breast. Breast Cancer Res Treat. 2009;113(1):59–66.CrossRefPubMed Green AR, Krivinskas S, Young P, Rakha EA, Paish EC, Powe DG, et al. Loss of expression of chromosome 16q genes DPEP1 and CTCF in lobular carcinoma in situ of the breast. Breast Cancer Res Treat. 2009;113(1):59–66.CrossRefPubMed
90.
Zurück zum Zitat Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54.CrossRefPubMed Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54.CrossRefPubMed
91.
Zurück zum Zitat Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99(24):15524–9.CrossRefPubMed Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99(24):15524–9.CrossRefPubMed
92.
Zurück zum Zitat Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 2005;102(39):13944–9.CrossRefPubMed Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 2005;102(39):13944–9.CrossRefPubMed
93.
Zurück zum Zitat Van Dyke DL, Shanafelt TD, Call TG, Zent CS, Smoley SA, Rabe KG, et al. A comprehensive evaluation of the prognostic significance of 13q deletions in patients with B-chronic lymphocytic leukaemia. Br J Haematol. 2010;148(4):544–50.CrossRefPubMed Van Dyke DL, Shanafelt TD, Call TG, Zent CS, Smoley SA, Rabe KG, et al. A comprehensive evaluation of the prognostic significance of 13q deletions in patients with B-chronic lymphocytic leukaemia. Br J Haematol. 2010;148(4):544–50.CrossRefPubMed
94.
Zurück zum Zitat Li J, Smyth P, Flavin R, Cahill S, Denning K, Aherne S, et al. Comparison of miRNA expression patterns using total RNA extracted from matched samples of formalin-fixed paraffin-embedded (FFPE) cells and snap frozen cells. BMC Biotechnol. 2007;29(7):36.CrossRef Li J, Smyth P, Flavin R, Cahill S, Denning K, Aherne S, et al. Comparison of miRNA expression patterns using total RNA extracted from matched samples of formalin-fixed paraffin-embedded (FFPE) cells and snap frozen cells. BMC Biotechnol. 2007;29(7):36.CrossRef
95.
Zurück zum Zitat Czubak K, Lewandowska MA, Klonowska K, Roszkowski K, Kowalewski J, Figlerowicz M, et al. High copy number variation of cancer-related microRNA genes and frequent amplification of DICER1 and DROSHA in lung cancer. Oncotarget. 2015;6(27):23399–416.CrossRefPubMedPubMedCentral Czubak K, Lewandowska MA, Klonowska K, Roszkowski K, Kowalewski J, Figlerowicz M, et al. High copy number variation of cancer-related microRNA genes and frequent amplification of DICER1 and DROSHA in lung cancer. Oncotarget. 2015;6(27):23399–416.CrossRefPubMedPubMedCentral
96.
Zurück zum Zitat Li N, Kaur S, Greshock J, Lassus H, Zhong X, Wang Y, et al. A combined array-based comparative genomic hybridization and functional library screening approach identifies mir-30d as an oncomir in cancer. Cancer Res. 2012;72(1):154–64.CrossRefPubMed Li N, Kaur S, Greshock J, Lassus H, Zhong X, Wang Y, et al. A combined array-based comparative genomic hybridization and functional library screening approach identifies mir-30d as an oncomir in cancer. Cancer Res. 2012;72(1):154–64.CrossRefPubMed
97.
Zurück zum Zitat Manceau G, Imbeaud S, Thiébaut R, Liébaert F, Fontaine K, Rousseau F, et al. Hsa-miR-31-3p expression is linked to progression-free survival in patients with KRAS wild-type metastatic colorectal cancer treated with anti-EGFR therapy. Clin Cancer Res. 2014;20(12):3338–47.CrossRefPubMed Manceau G, Imbeaud S, Thiébaut R, Liébaert F, Fontaine K, Rousseau F, et al. Hsa-miR-31-3p expression is linked to progression-free survival in patients with KRAS wild-type metastatic colorectal cancer treated with anti-EGFR therapy. Clin Cancer Res. 2014;20(12):3338–47.CrossRefPubMed
98.
Zurück zum Zitat Gilad S, Lithwick-Yanai G, Barshack I, Benjamin S, Krivitsky I, Edmonston TB, et al. Classification of the four main types of lung cancer using a microRNA-based diagnostic assay. J Mol Diagn. 2012;14(5):510–7.CrossRefPubMed Gilad S, Lithwick-Yanai G, Barshack I, Benjamin S, Krivitsky I, Edmonston TB, et al. Classification of the four main types of lung cancer using a microRNA-based diagnostic assay. J Mol Diagn. 2012;14(5):510–7.CrossRefPubMed
99.
Zurück zum Zitat Barh D, Carpi A, Verma M, Gunduz M, editors. Cancer biomarkers: minimal and noninvasive early diagnosis and prognosis. Boca Raton: CRC; 2017. Barh D, Carpi A, Verma M, Gunduz M, editors. Cancer biomarkers: minimal and noninvasive early diagnosis and prognosis. Boca Raton: CRC; 2017.
100.
101.
Zurück zum Zitat Boyerinas B, Park S-M, Hau A, Murmann AE, Peter ME. The role of let-7 in cell differentiation and cancer. Endocr Relat Cancer. 2010;17(1):F19–36.CrossRefPubMed Boyerinas B, Park S-M, Hau A, Murmann AE, Peter ME. The role of let-7 in cell differentiation and cancer. Endocr Relat Cancer. 2010;17(1):F19–36.CrossRefPubMed
103.
Zurück zum Zitat Cmarik JL, Min H, Hegamyer G, Zhan S, Kulesz-Martin M, Yoshinaga H, et al. Differentially expressed protein Pdcd4 inhibits tumor promoter-induced neoplastic transformation. Proc Natl Acad Sci USA. 1999;96(24):14037–42.CrossRefPubMed Cmarik JL, Min H, Hegamyer G, Zhan S, Kulesz-Martin M, Yoshinaga H, et al. Differentially expressed protein Pdcd4 inhibits tumor promoter-induced neoplastic transformation. Proc Natl Acad Sci USA. 1999;96(24):14037–42.CrossRefPubMed
104.
Zurück zum Zitat Jansen AP, Camalier CE, Colburn NH. Epidermal expression of the translation inhibitor programmed cell death 4 suppresses tumorigenesis. Cancer Res. 2005;65(14):6034–41.CrossRefPubMed Jansen AP, Camalier CE, Colburn NH. Epidermal expression of the translation inhibitor programmed cell death 4 suppresses tumorigenesis. Cancer Res. 2005;65(14):6034–41.CrossRefPubMed
105.
Zurück zum Zitat Leupold JH, Yang H-S, Colburn NH, Asangani I, Post S, Allgayer H. Tumor suppressor Pdcd4 inhibits invasion/intravasation and regulates urokinase receptor (u-PAR) gene expression via Sp-transcription factors. Oncogene. 2007;26(31):4550–62.CrossRefPubMed Leupold JH, Yang H-S, Colburn NH, Asangani I, Post S, Allgayer H. Tumor suppressor Pdcd4 inhibits invasion/intravasation and regulates urokinase receptor (u-PAR) gene expression via Sp-transcription factors. Oncogene. 2007;26(31):4550–62.CrossRefPubMed
107.
Zurück zum Zitat Resnick KE, Alder H, Hagan JP, Richardson DL, Croce CM, Cohn DE. The detection of differentially expressed microRNAs from the serum of ovarian cancer patients using a novel real-time PCR platform. Gynecol Oncol. 2009;112(1):55–9.CrossRefPubMed Resnick KE, Alder H, Hagan JP, Richardson DL, Croce CM, Cohn DE. The detection of differentially expressed microRNAs from the serum of ovarian cancer patients using a novel real-time PCR platform. Gynecol Oncol. 2009;112(1):55–9.CrossRefPubMed
108.
Zurück zum Zitat Ryu JK, Matthaei H, Dal Molin M, Hong SM, Canto MI, Schulick RD, et al. Elevated microRNA miR-21 levels in pancreatic cyst fluid are predictive of mucinous precursor lesions of ductal adenocarcinoma. Pancreatology. 2011;11(3):343–50.CrossRefPubMedPubMedCentral Ryu JK, Matthaei H, Dal Molin M, Hong SM, Canto MI, Schulick RD, et al. Elevated microRNA miR-21 levels in pancreatic cyst fluid are predictive of mucinous precursor lesions of ductal adenocarcinoma. Pancreatology. 2011;11(3):343–50.CrossRefPubMedPubMedCentral
109.
Zurück zum Zitat Zeng Z, Wang J, Zhao L, Hu P, Zhang H, Tang X, et al. Potential role of microRNA-21 in the diagnosis of gastric cancer: a meta-analysis. PloS One. 2013;8(9):e73278.CrossRefPubMedPubMedCentral Zeng Z, Wang J, Zhao L, Hu P, Zhang H, Tang X, et al. Potential role of microRNA-21 in the diagnosis of gastric cancer: a meta-analysis. PloS One. 2013;8(9):e73278.CrossRefPubMedPubMedCentral
110.
Zurück zum Zitat Shiotani A, Murao T, Kimura Y, Matsumoto H, Kamada T, Kusunoki H, et al. Identification of serum miRNAs as novel non-invasive biomarkers for detection of high risk for early gastric cancer. Br J Cancer. 2013;109(9):2323–30.CrossRefPubMedPubMedCentral Shiotani A, Murao T, Kimura Y, Matsumoto H, Kamada T, Kusunoki H, et al. Identification of serum miRNAs as novel non-invasive biomarkers for detection of high risk for early gastric cancer. Br J Cancer. 2013;109(9):2323–30.CrossRefPubMedPubMedCentral
111.
Zurück zum Zitat Song L, Jia J, Peng X, Xiao W, Li Y. The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: a meta-analysis. Sci Rep. 2017;7(1):3032.CrossRefPubMedPubMedCentral Song L, Jia J, Peng X, Xiao W, Li Y. The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: a meta-analysis. Sci Rep. 2017;7(1):3032.CrossRefPubMedPubMedCentral
113.
Zurück zum Zitat Nian J, Sun X, Ming S, Yan C, Ma Y, Feng Y, et al. Diagnostic accuracy of methylated SEPT9 for blood-based colorectal cancer detection: a systematic review and meta-analysis. Clin Transl Gastroenterol. 2017;8(1):e216.CrossRefPubMedPubMedCentral Nian J, Sun X, Ming S, Yan C, Ma Y, Feng Y, et al. Diagnostic accuracy of methylated SEPT9 for blood-based colorectal cancer detection: a systematic review and meta-analysis. Clin Transl Gastroenterol. 2017;8(1):e216.CrossRefPubMedPubMedCentral
114.
Zurück zum Zitat Thon N, Kreth S, Kreth FW. Personalized treatment strategies in glioblastoma: MGMT promoter methylation status. Onco Targets Ther. 2013;27(6):1363–72.CrossRef Thon N, Kreth S, Kreth FW. Personalized treatment strategies in glioblastoma: MGMT promoter methylation status. Onco Targets Ther. 2013;27(6):1363–72.CrossRef
115.
Zurück zum Zitat Fiorentino A, Balducci M, De Bonis P, Chiesa S, De Filippo L, Mangiola A, et al. Can elderly patients with newly diagnosed glioblastoma be enrolled in radiochemotherapy trials? Am J Clin Oncol. 2015;38(1):23–7.CrossRefPubMed Fiorentino A, Balducci M, De Bonis P, Chiesa S, De Filippo L, Mangiola A, et al. Can elderly patients with newly diagnosed glioblastoma be enrolled in radiochemotherapy trials? Am J Clin Oncol. 2015;38(1):23–7.CrossRefPubMed
116.
Zurück zum Zitat Wang Z, Jiang W, Wang Y, Guo Y, Cong Z, Du F, et al. MGMT promoter methylation in serum and cerebrospinal fluid as a tumor-specific biomarker of glioma. Biomed Rep. 2015;3(4):543–8.CrossRefPubMedPubMedCentral Wang Z, Jiang W, Wang Y, Guo Y, Cong Z, Du F, et al. MGMT promoter methylation in serum and cerebrospinal fluid as a tumor-specific biomarker of glioma. Biomed Rep. 2015;3(4):543–8.CrossRefPubMedPubMedCentral
118.
Zurück zum Zitat Selaru FM, Olaru AV, Kan T, David S, Cheng Y, Mori Y, et al. MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3. Hepatology. 2009;49(5):1595–601.CrossRefPubMedPubMedCentral Selaru FM, Olaru AV, Kan T, David S, Cheng Y, Mori Y, et al. MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3. Hepatology. 2009;49(5):1595–601.CrossRefPubMedPubMedCentral
119.
Zurück zum Zitat Verma M, Kumar V. Chapter 21—Epigenetic drugs for cancer and precision medicine. In: Moskalev A, Vaiserman AM, editors. Epigenetics of aging and longevity. Boston: Academic; 2018. p. 439–51.CrossRef Verma M, Kumar V. Chapter 21—Epigenetic drugs for cancer and precision medicine. In: Moskalev A, Vaiserman AM, editors. Epigenetics of aging and longevity. Boston: Academic; 2018. p. 439–51.CrossRef
120.
Zurück zum Zitat Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nat Rev Genet. 2016;17(10):630–41.CrossRefPubMed Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nat Rev Genet. 2016;17(10):630–41.CrossRefPubMed
121.
Zurück zum Zitat Biswas S, Rao CM. Epigenetic tools (the writers, the readers and the erasers) and their implications in cancer therapy. Eur J Pharmacol. 2018;15(837):8–24.CrossRef Biswas S, Rao CM. Epigenetic tools (the writers, the readers and the erasers) and their implications in cancer therapy. Eur J Pharmacol. 2018;15(837):8–24.CrossRef
122.
Zurück zum Zitat Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18(7):1414.CrossRefPubMedCentral Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18(7):1414.CrossRefPubMedCentral
123.
124.
Zurück zum Zitat van den Bent MJ, Erdem-Eraslan L, Idbaih A, de Rooi J, Eilers PH, Spliet WG, et al. MGMT-STP27 methylation status as predictive marker for response to PCV in anaplastic oligodendrogliomas and oligoastrocytomas. A report from EORTC study 26951. Clin Cancer Res. 2013;19(19):5513–22.CrossRefPubMed van den Bent MJ, Erdem-Eraslan L, Idbaih A, de Rooi J, Eilers PH, Spliet WG, et al. MGMT-STP27 methylation status as predictive marker for response to PCV in anaplastic oligodendrogliomas and oligoastrocytomas. A report from EORTC study 26951. Clin Cancer Res. 2013;19(19):5513–22.CrossRefPubMed
125.
Zurück zum Zitat Mastoraki S, Strati A, Tzanikou E, Chimonidou M, Politaki E, Voutsina A, et al. ESR1 methylation: a liquid biopsy-based epigenetic assay for the follow-up of patients with metastatic breast cancer receiving endocrine treatment. Clin Cancer Res. 2018;24(6):1500–10.CrossRefPubMed Mastoraki S, Strati A, Tzanikou E, Chimonidou M, Politaki E, Voutsina A, et al. ESR1 methylation: a liquid biopsy-based epigenetic assay for the follow-up of patients with metastatic breast cancer receiving endocrine treatment. Clin Cancer Res. 2018;24(6):1500–10.CrossRefPubMed
126.
Zurück zum Zitat Vedeld HM, Nesbakken A, Lothe RA, Lind GE. Re-assessing ZNF331 as a DNA methylation biomarker for colorectal cancer. Clin Epigenetics. 2018;10:70.CrossRefPubMedPubMedCentral Vedeld HM, Nesbakken A, Lothe RA, Lind GE. Re-assessing ZNF331 as a DNA methylation biomarker for colorectal cancer. Clin Epigenetics. 2018;10:70.CrossRefPubMedPubMedCentral
127.
Zurück zum Zitat Misawa K, Misawa Y, Imai A, Mochizuki D, Endo S, Mima M, et al. Epigenetic modification of SALL1 as a novel biomarker for the prognosis of early stage head and neck cancer. J Cancer. 2018;9(6):941–9.CrossRefPubMedPubMedCentral Misawa K, Misawa Y, Imai A, Mochizuki D, Endo S, Mima M, et al. Epigenetic modification of SALL1 as a novel biomarker for the prognosis of early stage head and neck cancer. J Cancer. 2018;9(6):941–9.CrossRefPubMedPubMedCentral
128.
Zurück zum Zitat Rahier JF, Druez A, Faugeras L, Martinet JP, Gehenot M, Josseaux E, et al. Circulating nucleosomes as new blood-based biomarkers for detection of colorectal cancer. Clin Epigenetics. 2017;9:53.CrossRefPubMedPubMedCentral Rahier JF, Druez A, Faugeras L, Martinet JP, Gehenot M, Josseaux E, et al. Circulating nucleosomes as new blood-based biomarkers for detection of colorectal cancer. Clin Epigenetics. 2017;9:53.CrossRefPubMedPubMedCentral
129.
Zurück zum Zitat Liu H, Li Y, Li J, Liu Y, Cui B. H3K4me3 and Wdr82 are associated with tumor progression and a favorable prognosis in human colorectal cancer. Oncol Lett. 2018;16(2):2125–34.PubMedPubMedCentral Liu H, Li Y, Li J, Liu Y, Cui B. H3K4me3 and Wdr82 are associated with tumor progression and a favorable prognosis in human colorectal cancer. Oncol Lett. 2018;16(2):2125–34.PubMedPubMedCentral
130.
Zurück zum Zitat Bell EH, Chakraborty AR, Mo X, Liu Z, Shilo K, Kirste S, et al. SMARCA4/BRG1 is a novel prognostic biomarker predictive of cisplatin-based chemotherapy outcomes in resected non-small cell lung cancer. Clin Cancer Res. 2016;22(10):2396–404.CrossRefPubMed Bell EH, Chakraborty AR, Mo X, Liu Z, Shilo K, Kirste S, et al. SMARCA4/BRG1 is a novel prognostic biomarker predictive of cisplatin-based chemotherapy outcomes in resected non-small cell lung cancer. Clin Cancer Res. 2016;22(10):2396–404.CrossRefPubMed
131.
Zurück zum Zitat Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer. 2010;127(1):118–26.CrossRefPubMed Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer. 2010;127(1):118–26.CrossRefPubMed
132.
Zurück zum Zitat Krawczyk P, Powrózek T, Olesiński T, Dmitruk A, Dziwota J, Kowalski D, et al. Evaluation of miR-506 and miR-4316 expression in early and non-invasive diagnosis of colorectal cancer. Int J Colorectal Dis. 2017;32(7):1057–60.CrossRefPubMedPubMedCentral Krawczyk P, Powrózek T, Olesiński T, Dmitruk A, Dziwota J, Kowalski D, et al. Evaluation of miR-506 and miR-4316 expression in early and non-invasive diagnosis of colorectal cancer. Int J Colorectal Dis. 2017;32(7):1057–60.CrossRefPubMedPubMedCentral
133.
Zurück zum Zitat Leng Q, Lin Y, Jiang F, Lee CJ, Zhan M, Fang H, et al. A plasma miRNA signature for lung cancer early detection. Oncotarget. 2017;8(67):111902–11.CrossRefPubMedPubMedCentral Leng Q, Lin Y, Jiang F, Lee CJ, Zhan M, Fang H, et al. A plasma miRNA signature for lung cancer early detection. Oncotarget. 2017;8(67):111902–11.CrossRefPubMedPubMedCentral
134.
Zurück zum Zitat Fogli S, Polini B, Carpi S, Pardini B, Naccarati A, Dubbini N, et al. Identification of plasma microRNAs as new potential biomarkers with high diagnostic power in human cutaneous melanoma. Tumour Biol. 2017;39(5):1010428317701646.CrossRefPubMed Fogli S, Polini B, Carpi S, Pardini B, Naccarati A, Dubbini N, et al. Identification of plasma microRNAs as new potential biomarkers with high diagnostic power in human cutaneous melanoma. Tumour Biol. 2017;39(5):1010428317701646.CrossRefPubMed
135.
Zurück zum Zitat Lin HM, Castillo L, Mahon KL, Chiam K, Lee BY, Nguyen Q, et al. Circulating microRNAs are associated with docetaxel chemotherapy outcome in castration-resistant prostate cancer. Br J Cancer. 2014;110(10):2462–71.CrossRefPubMedPubMedCentral Lin HM, Castillo L, Mahon KL, Chiam K, Lee BY, Nguyen Q, et al. Circulating microRNAs are associated with docetaxel chemotherapy outcome in castration-resistant prostate cancer. Br J Cancer. 2014;110(10):2462–71.CrossRefPubMedPubMedCentral
Metadaten
Titel
Prognostic and Predictive Epigenetic Biomarkers in Oncology
verfasst von
Katarzyna Kamińska
Ewelina Nalejska
Marta Kubiak
Joanna Wojtysiak
Łukasz Żołna
Janusz Kowalewski
Marzena Anna Lewandowska
Publikationsdatum
01.02.2019
Verlag
Springer International Publishing
Erschienen in
Molecular Diagnosis & Therapy / Ausgabe 1/2019
Print ISSN: 1177-1062
Elektronische ISSN: 1179-2000
DOI
https://doi.org/10.1007/s40291-018-0371-7

Weitere Artikel der Ausgabe 1/2019

Molecular Diagnosis & Therapy 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.