• <tr id="yyy80"></tr>
  • <sup id="yyy80"></sup>
  • <tfoot id="yyy80"><noscript id="yyy80"></noscript></tfoot>
  • 99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看 ?

    Epigenetic modulators for brain cancer stem cells: Implications for anticancer treatment

    2021-07-30 06:08:36LuanaAbballeEvelinaMiele
    World Journal of Stem Cells 2021年7期

    Luana Abballe, Evelina Miele

    Luana Abballe, Evelina Miele, Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy

    Abstract Primary malignant brain tumors are a major cause of morbidity and mortality in both adults and children, with a dismal prognosis despite multimodal therapeutic approaches. In the last years, a specific subpopulation of cells within the tumor bulk, named cancer stem cells (CSCs) or tumor-initiating cells, have been identified in brain tumors as responsible for cancer growth and disease progression. Stemness features of tumor cells strongly affect treatment response, leading to the escape from conventional therapeutic approaches and subsequently causing tumor relapse. Recent research efforts have focused at identifying new therapeutic strategies capable of specifically targeting CSCs in cancers by taking into consideration their complex nature. Aberrant epigenetic machinery plays a key role in the genesis and progression of brain tumors as well as inducing CSC reprogramming and preserving CSC characteristics. Thus, reverting the cancer epigenome can be considered a promising therapeutic strategy. Three main epigenetic mechanisms have been described: DNA methylation, histone modifications, and non-coding RNA, particularly microRNAs. Each of these mechanisms has been proven to be targetable by chemical compounds, known as epigeneticbased drugs or epidrugs, that specifically target epigenetic marks. We review here recent advances in the study of epigenetic modulators promoting and sustaining brain tumor stem-like cells. We focus on their potential role in cancer therapy.

    Key Words: Cancer stem cells; Epigenetics; Brain tumors; Epigenetic drugs; Histone deacetylase inhibitors; DNA methyltransferase inhibitors

    INTRODUCTION

    Primary malignant brain tumors are a heterogeneous group of tumors arising from the brain parenchyma and its surrounding structures. They represent a major cause of morbidity and mortality in both adults and children. In particular, brain tumors are the most frequently reported solid malignancies in children, accounting for up to 20%of childhood cancers[1]. Brain cancers require a multimodal treatment that includes surgical resection, chemotherapy, and radiation therapy[2]. This therapeutic strategy includes a particularly invasive surgery as well as brain irradiation and can lead to long-lasting side effects, significantly decreasing the patient’s quality of life.

    Brain tumors can harbor genetic and epigenetic alterations that make them resistant to conventional pharmacological treatment[3]. Current therapeutic approaches do not consider the presence of a specific subpopulation of cells within the tumor bulk,known as cancer stem cells (CSCs) or tumor-initiating cells. These cells are not responsive to conventional treatment, promoting tumor relapse[4]. In this scenario, it is of fundamental importance to seek alternative therapeutic strategies that take into account the genetic and epigenetic alterations of the tumor as well as the presence of tumor-initiating cells. Nowadays, to address this challenge, the epigenetic profile of CSCs is being thoroughly studied and several epigenetics drugs are being tested inin vitroandin vivostudies on CSC cultures[5].

    This review aims to summarize and organize the current epigenetics strategies for eradicating brain CSCs based on CSC epigenetic modulators.

    Brain cancer stem cells

    CSCs are considered the “reservoirs” of the tumor. They are defined as a small subset of stem cells capable of proliferating and generating diverse and heterogeneous cell types that constitute the whole tumor[6]. CSCs reside in specific anatomical areas called “niches”, where they interact with the microenvironment surrounding them[7].Like their normal tissue counterpart, CSCs exhibit “stem-like” characteristics: (1) Cell quiescence: A way to preserve self-renewal and to avoid genetic perturbation that could occur during cell division; (2) Self-renewal capacity: The ability to proliferate symmetrically and asymmetrically; (3) Multipotency: The ability to give rise to heterogeneous cells with different proliferative potential; (4) Migration: The ability to migrate and disseminate; (5) Tissue regeneration: The ability to give rise to new tumoral tissues; and (6) Communication: The ability to interact with the microenvironment.

    The substantial difference between CSCs and normal tissue stem cells is that proliferation/death signals are aberrant and dysregulated in cancer, whereas normal tissue stem cells can maintain physiological homeostasis.

    CSCs were discovered for the first time in acute myeloid leukemia[8]. Subsequently,this tumor-initiation subpopulation was also identified in many solid tumors,including brain cancers[9,10]. CSCs have been discovered and isolated in major brain tumors [e.g., medulloblastoma (MB), gliomas, ependymoma][11-13] from both pediatric and adult patients[14-16].

    In the first instance, cell surface markers were used to identify this subset of tumorinitiating cells. The cell surface antigen CD133 has been most frequently used to mark CSCs in various solid tumors[17]. In the context of the brain, Singhet al[11]demonstrated that CD133+human brain tumor cells were able to initiate and recapitulate the original tumor inin vivomouse models. Further study demonstrated that a high expression level of CD133 was correlated with poor prognosis in brain cancer patients, reinforcing CD133’s role as a “brain stemness” marker[18].

    The accuracy of this detection method remains very controversial because cell surface markers evolve rapidly in response to different environmental stimuli, disease states, and tumor progression[19,20]. Moreover, brain CSCs and neural stem cells share common phenotypic markers (e.g., CD133, CD15, CD44)[21]. Hence, the markerdependent identification method alone is insufficient to discriminate correctly CSCs from normal tissue ones. Recent studies[22-24] investigate complementary methods that consider cell dysregulated survival pathways and genetic and epigenetic signatures. As already described by Abbaszadeganet al[23], the gold standard strategy to identify efficiently brain CSCs is to test theirin vivotumorigenicity. Limiting dilution assay is the best tumorigenicity method that is commonly used for evaluation of CSCs frequency. However, this method presents some critical points, being influenced by the number of the cells, the implantation site, and growing time of incubation. Moreover, it is not feasible on large scale studies. Complementaryin vitrofunctional assays could be used to identify CSCs based on: (1) Their intrinsic properties (e.g., self-renewal, asymmetrical division, slow proliferation phenotype, and aldehyde dehydrogenase 1 expression); and (2) Their survival pathways (e.g., Wnt/βcatenin, Hedgehog, and Notch signaling pathway), in terms of expression of transcription factors/key proteins/microRNAs (miRNAs). Among the recently developed approaches to isolate CSCs, there are next generation sequencing (NGS)technologies. For example, Jonassonet al[24] isolated the stem-like subpopulation using a functional cellular assay that enriches for cells that can self-renew and differentiate, combined with NGS technologies (single-cell RNA sequencing) to identify CSCs[22-24]. Moreover, Rodriguez-Meiraet al[25] in their scientific work developed an NGS platform that combines single-cell RNA-seq with mutational analysis allowing the identification of distinct subclones of cancer cells[26]. This evidence suggests that a combination of cell surface markers and functional assays provide an efficient tool for their identification.

    CSCs’ properties have profound implications for treatment response due to their ability to evade conventional therapy and cause subsequent tumor relapse[27,28]. In this case, it is crucial to identify new treatment strategies that take into consideration the complexities of CSCs. Currently, global researchers are making great efforts to understand the biological properties of CSCs and develop new therapeutic approaches targeting CSCs.

    EPIGENETIC MODULATORS

    The term "epigenetics" is used to refer to information that controls gene expression that is stable and inheritable during cell division and happens without changes in the DNA sequence. Aberrant epigenetic landscapes control cell fate specification,promoting tumor initiation and progression[29,30].

    Epigenetics controls gene expression through three main mechanisms: DNA methylation, histone modifications, and non-coding RNA, particularly miRNAs[31](Figure 1).

    DNA methylation is the most studied epigenetic modification. It is associated with transcriptional inactivation and closed chromatin structure, mainly regulating gene silencing or repression. It depends on the action of specialized enzymes, known as DNA methyltransferases (DNMTs) that transfer a methyl group from S-adenosyl methionine to the fifth carbon of the pyrimidine ring of a DNA base cytosine. DNA methylation occurs mainly at CpG dinucleotides, which are concentrated in specific regions of DNA called “CpG islands”. CpG islands are located at gene promoters, in regulatory regions, and in gene bodies[32], but DNA methylation could also be present in non-coding DNA sequences such as repetitive elements, transposons, non-coding RNAs, and introns[33,34].

    A methylation pattern represents the fingerprint of a cell. It is established during early embryogenesis, and it is maintained during cell division by the action of DNMTs.

    Methylation status is altered in cancers, mainly through two mechanisms: Regional hypermethylation and global hypomethylation[35]. DNA hypermethylation involves the CpG islands, which are usually unmethylated in normal cells. The result of DNA hypermethylation is the transcriptional repression of tumor-suppressor and tissuespecific genes, and the inactivation of miRNA, which are involved in the initiation and progression of cancer[34]. This mechanism occurs in different stages of carcinogenesis including CSC formation[36]. Conversely, global hypomethylation consists of the loss of the methyl group on cytosine, mainly in repetitive elements across DNA.Hypomethylation was one of the first epigenetic features discovered in human cancers[37], causing the reactivation of methylated regions of DNA such as transposons,introns, and germ-line genes that are silent in differentiated cells[34,38]. In high-grade cancers, such as glioblastoma (GBM), hypomethylation is a mechanism used by CSCs to reactivate key stem cell genes[39].

    Abnormalities of DNA methylation are early events in pre-malignant transformation and are maintained in the global tumor population. However, the epigenome is in continuous evolution, and some of the changes are detectable in later steps of tumorigenesis as a result of positive selection. In this way, epigenome contributes to tumor heterogeneity and plasticity, which gives rise to a heterogeneous tumor composed of different cell subpopulations, one of them could have “stem-like” features.Additionally, compared to the bulk tumor, CSCs could acquire further epigenetic alterations in response to stress of different nature (e.g., chemotherapy/radiotherapy,chronic inflammation, and environmental exposures), contributing to tumor relapse[40].

    The development of powerful “next-generation” techniques makes it possible to describe the entire epigenome of cells or tissues[38]. Capperet al[41] developed a DNA methylation-based classification for central nervous system cancers that allows discrimination between different subtypes of tumors, some of them previously considered as homogeneous diseases.

    Histones are subject to reversible post-translational modifications (PTMs) that cooperate to govern the chromatin state. Histone PTMs influence chromatin structure that is conducive to the expression or repression of target genes. Histone aminoterminal regions can undergo diverse PTMs: Methylation, acetylation, phosphorylation, ubiquitylation, biotinylation, sumoylation, and ADP-ribosylation[42] that work in concert to define the chromatin status of the specific region of DNA.

    Histone PTMs are controlled by different enzymes: "Writers" catalyze histone modifications; “erasers” cut histone modifications; and “readers” translate the PTMs’language into cellular signals. All enzymes work together in a very specific manner to create the “histone code”.

    Histone methylation and acetylation are the best-known PTMs on histone residues.Histone methylation is a reversible mechanism that occurs on lysine and arginine residues, leading to a different degree of methylation. Up to three methyl groups can be added to a single lysine residue (un-, mono-, di-, and tri-methylated states) and up to two groups to a single arginine residue (mono- and di-methylated states).Methylation can be either an activating or a repressive mark, depending on the target histone and lysine residue: For example, histone H3 Lysine 27 (H3K27) and histone H4 Lysine 20 (H4K20) are usually associated with gene silencing, while H3 Lysine 4(H3K4) and H3 Lysine 36 (H3K36) are transcriptional activation marks. Histone acetylation is associated with relaxed chromatin structure and promotes the binding of transcription factors and RNA polymerase to DNA, resulting in the activation of gene expression[42].

    Recent efforts have tried to highlight the role of epigenetic modulators in the development and plasticity of brain tumor CSCs[43,44]. In GBM CSCs (GSCs), Liauet al[45] showed in patient-derived GSCs, which effectively initiate tumors in mice models, that KDM6-mediated demethylation of histone H3 Lysine 27 trimethylation(H3K27me3), a repressive mark, has a central role in the maintenance and persistence of GSCs. Maramponet al[46] demonstrated the fundamental role of two histone deacetylases (HDAC4 and HDAC6) in regulating the DNA repair machinery, survival,and stemness characteristics of radioresistant GSCs derived from U87MG and U251MG human GMB cell lines subjected to irradiation. Moreover, a recent study by Banelliet al[47] on human GSCs derived from human tumors describes a subset of GSCs resistant to chemotherapy that are particularly dependent on KDM5A action,making them strongly sensitive to HDAC inhibitor (HDACi) treatments in term of cell viability, percentage of apoptotic cells, and reducing capacity of clonal growth. In medulloblastoma, pharmacological inhibition of histone methyltransferase enhancer of zeste 2 (EZH2) impairs proliferation and self-renewal of human and mouse stem-like cells, derived from primary human Sonic Hedgehog MB (SHH-MBs) and from tumors arisen in Ptc+/- mice, inin vitroandin vivostudies[48].

    miRNAs and epigenetic modulators create a miRNA-epigenetic feedback loop: On the one hand, miRNA can regulate the transcription of epigenetics-associated enzymes, while on the other miRNA transcription is under the control of epigenetics machinery (DNA methylation, histone PTMs, and RNA modifications)[49].

    In literature, there are several examples of miRNA-epigenetic machinery in CSCs.For example, Ferrettiet al[50] described the role of the miR-326-ARRB1-E2F1 axis in medulloblastoma CSCs survival. For this study, MB CSCs were derived from tissues freshly resected from pediatric patients and cultured in CSC-enriched cultures.Notably, miR-326 is already described as onco-suppressor miRNA[50]. It is involved in neuronal differentiation, and its expression is under control of the histone-lysine Nmethyltransferase EZH2. High levels of EZH2 are responsible for the presence of H3K27me3 on the promoter region of miR-326[22]. In another study, Lizarte Netoet al[51] investigated the role of miR-181d and the methylation status of the MGMT gene in response to pharmacological treatment in CD133+ GBM CSCs. Their results showed an increase of miR-181d and MGMT transcription levels after treatment, due to cellular mechanism of drug resistance[51].

    The tumor microenvironment (TME) also acts as an epigenetic regulator for cancer cells. TME communicates with cancer cells through extracellular vesicles secreted by many TME cell types that contain various mediators including proteins and nucleic acids. Also, miRNAs can be charged in the extracellular vesicles and thereby alter the epigenome of the recipient cancer cell[52].

    All this evidence makes brain CSCs an excellent candidate for pharmacological epigenetic treatments.

    TRANSLATIONAL SIGNIFICANCE OF EPIGENETICS: EPIDRUGS

    Epigenetics-based drugs (epidrugs) are chemical compounds that specifically target epigenetic marks (Figure 1). CSCs could acquire mutations in epigenetic marks or changing in methylation/acetylation status, or again in miRNAs signature, that make them sensitive to epigenetics-based drugs’ approaches. Epidrugs are based on the idea that by changing the epigenome of CSCs, it may be possible to remodel the fate of cells from CSCs to differentiated tumor cells[53-55]. Epidrugs are used as monotherapy or in combination with chemo-/radio-therapy to target both CSCs and bulk tumors.Today, several HDAC and DNMT inhibitors are in different clinical trial phases.

    HDACi

    Among the various epigenetic modulators, the acetylation state of histone proteins is one of the major targets for anticancer therapy. Histone acetylation is controlled by two types of enzymes: HDACs and histone acetyltransferases (Figure 1).

    Human HDACs are categorized into four different classes: Class I (HDAC1, 2, 3,and 8), class IIa (HDAC4, 5, 7, and 9), class IIb (HDAC6 and 10), class III (sirt 1-7), and class IV (HDAC11). HDACs are enzymes that remove the acetyl group from lysine residues on histones, which compact the chromatin structure into a non-permissive state, resulting in transcriptional repression.

    Based on isoform selectivity, HDACi can be classified as: (1) Pan-inhibitors, if they act against all HDACs; or (2) HDAC isoform-selective inhibitors if they target a specific HDAC class[56]. Pan-inhibitors are in turn classified according to their chemical structure as: (1) Hydroxamic acids; (2) Aliphatic carboxylic acids; (3)Benzamides; (4) Cyclic peptides; or (5) Sirtuin inhibitors.

    HDACs control several cellular mechanisms. They have been implicated in different types of cancers[46], and several HDACs are overexpressed in brain cancers[57,58]. For example, Staberget al[59] found an up-expression of HDAC1, 3, and 6 in 21 primary GBM cell cultures, grown as neurospheres, compared to non-neoplastic brain control cells (normal human astrocytes), and confirmed these findings in a panel of primary GBM tissue samples compared to normal brain tissues. They demonstrated the efficacy of HDACi therapy (trichostatin A) in GBM treatment inin vitroexperiments.

    HDACi exhibit anti-cancer activity against CSCs in tumors with a predominant stem-like population such as brain cancers. HDACi target the escape mechanism of CSCs, reversing chemo-radio-therapy resistance by inducing cell differentiation,apoptosis, inhibition of angiogenesis, and upregulation of tumor suppressor genes[60].

    In line with this observation, da Cunha Jaegeret al[61] demonstrated that HDACi and mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitors could modulate the stemness markers (BMI1 and CD133), viability, and neurosphere formation capacity of MB cell lines (DAOY and D283 cultured in serum-free sphereinduction medium). Their results proposed HDACi as a valid candidate for MB CSC treatment[61]. Coniet al[62] reported antitumor effects of selective HDAC1 and HDAC2 inhibition on SHH-MB cells and mouse models. Also, in diffuse intrinsic pontine glioma tumor models (patient-derived neurospheres, xenografts, and allografts), combination of HDAC and AXL inhibition modulate the H3K27M epigenetic mark resulting in a down-regulation of stemness markers (SOX2 and its target genes directly correlated with stem cell characteristics)[63].

    Several HDACi are emerging as promising anticancer drugs in clinical trials for brain cancer treatment, both as combinatorial- and as mono-therapy[64]. Table 1 shows the HDACi used in clinical trials for brain cancers (Vorinostat, Valproic acid,Panobinostat, and Entinostat). Most clinical trials are focused on the usage of Vorinostat (suberoylanilide hydroxamic acid or SAHA), a potent inhibitor of HDAC classes I and II. Vorinostat was initially approved by the Food and Drug Administration for treating refractory cutaneous T-cell lymphoma, and its use was subsequently extended to various solid cancers[56]. In a phase 2 clinical trial,Vorinostat was tested as a mono-therapeutic agent in GBM treatment to target both CSCs and differentiated cancer cells. In fact, preclinical studies demonstrated the efficacy of Vorinostat in reducing EZH2 and CD133 stemness marker in patientderived GBM CSCs[65] and in modulating senescenceviathe p38-p53 axis and inducing apoptosis in GBM cell lines cultured in serum-free sphere-induction medium[66]. Sunget al[67] demonstrated that a combination of Vorinostat with melatonin can overcome the pharmacological resistance of human GBM CSCs, reducing self-renewal and proliferation of stem cell compartments and increasing apoptosis markers(cleaved poly-ADP ribose polymerase and p-γH2AX).

    Table 1 Histone deacetylases inhibitors in clinical trial for brain cancer treatment

    Adult gliosarcoma acids)Recurrent adult brain tumor Adult giant cell GBM Adult GBM Adult gliosarcoma Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Recurrent adult brain tumor Phase II Vorinostat in Treating Patients With Progressive or Recurrent Glioblastoma Multiforme Recurrent GBM multiforme Malignant glioma Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Adult brain tumor Phase II Ph II SAHA and Bevacizumab for Recurrent Malignant Glioma Patients Brain cancer Neoplasm metastasis Lung cancer Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Carcinoma, non-small-cell lung Phase I Phase I Vorinostat Concurrent With Stereotactic Radiosurgery (SRS) in Brain Metastases From Non-Small Cell Lung Cancer Brain tumor Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC GBM Phase I/II Suberoylanilide Hydroxamic Acid (SAHA),Bevacizumab, Daily Temozolomide for Recurrent Malignant Gliomas Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Brain metastases Phase I Study of the Combination of Vorinostat and Radiation Therapy for the Treatment of Patients With Brain Metastases Adult GBM Depression Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Recurrent adult brain tumor Not applicable Magnetic Resonance Spectroscopy Imaging in Predicting Response to Vorinostat and Temozolomide in Patients With Recurrent or Progressive Glioblastoma Adult anaplastic astrocytoma Adult anaplastic oligodendrogliomaAdult giant cell GBM Adult GBM Adult gliosarcomaAdult mixed glioma Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Recurrent adult brain neoplasm Phase I Vorinostat and Temozolomide in Treating Patients With Malignant Gliomas Medulloblastoma Pineoblastoma Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Supratentorial embryonal tumor Phase I Vorinostat Combined With Isotretinoin and Chemotherapy in Treating Younger Patients With Embryonal Tumors of the Central Nervous System Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Brain metastasis Phase II Vorinostat and Concurrent Whole Brain Radiotherapy for Brain Metastasis Adult anaplastic astrocytoma Adult anaplastic oligodendroglioma Adult giant cell GBM Adult GBM Adult gliosarcoma Adult mixed glioma Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC Recurrent adult brain neoplasm Phase I High-Dose Vorinostat and Fractionated Stereotactic Body Radiation Therapy in Treating Patients With Recurrent Glioma GBM Brain tumor Vorinostat(SAHA)iHDAC(hydroxymic acids)Pan-HDAC GBM Phase I Pembrolizumab and Vorinostat Combined With Temozolomide for Newly Diagnosed Glioblastoma Brain stem glioma Vorinostat iHDAC Pan-Vorinostat, Temozolomide, or Bevacizumab in Phase II/III

    HDAC: Histone deacetylases; iHDAC: HDAC inhibitors; DIPG: Diffuse intrinsic pontine gliomas; WHO: World Health Organization; Ph: Phase; SRS:Stereotactic radiosurgery; GBM: Glioblastoma.

    Vorinostat acts by inducing the acetylation of proteins, including histones and transcription factors at both transcriptional and non-transcriptional levels, leading to different cellular effects[68]. Vorinostat, like other pan-HDACi, has variable activity against HDAC isoenzymes, some of which are important in antitumor response.

    Vorinostat shows a different toxicity profile compared to classical chemotherapeutic drugs. In fact, common pan-HDACi side effects include fatigue, nausea/vomiting,anemia, anorexia, increased blood urea, hyperglycemia, and thrombocytopenia. Some of these adverse effects can be routinely managed by physicians, but some others need more careful monitoring[69]. However, side effects are dose-dependent and Vorinostat is effective at very low concentrations[70]. There are several reasons behind the epidrugs’ toxicity. It is partly due to “on-target” effects, which could be explained with the concept of “pleiotropy”. Specifically, a single target gene could be involved in different signaling and controls multiple phenotypic effects. Another reason is “off-target” effects. Epi-drugs are designed to inhibit aberrant epigenetic enzymes, but it is known that they could also affect other classes of substrates belonging to unintended cellular pathways, at both intracellular and extracellular levels[71].

    In medulloblastoma, the administration of epigenetic modifiers such as Vorinostat and Valproic acid shows radiosynergistic action on the proliferation and cloning capacity of three human MB cell lines (DAOY, MEB-Med8a, and D283-Med), offering a new opportunity to treat MB patients[72].

    DNA methyltransferase inhibitors

    The readout of DNMTs is hypermethylation of the DNA sequence and resulting silencing of gene expression. Aberrant DNA methylation has been associated with different stages of cancers. This evidence supports a rationale for using DNMT inhibitors (DNMTi) in cancer treatment (Figure 1).

    There are two different classes of DNMTi: (1) Nucleoside analogues, which could be considered analogues to cytosine and act as a natural substrate for DNMT (e.g., 5-azacytidine); and (2) Nonnucleoside compounds, which inhibit DNA methyltransferase activity through mechanisms other than DNA incorporation.

    This second class of DNMTi appears to be less toxic and more stable than nucleoside analogues[5,73,74], and Valente and colleagues have focused on the study of nonnucleoside compounds[75].

    The archetypal DNA methyltransferase inhibitor is 5-azacytidine (also known as 5-aza), it is a nucleoside inhibitor that is incorporated into the DNA sequence and covalently bonds to and inactivates DNA methyltransferase. As shown in Table 2, 5-azacytidine has been tested in different phase I trials against various brain cancers,particularly in recurrent brain tumors, GBM, and ependymoma.

    Table 2 DNA methyltransferase inhibitors in clinical trial for brain cancer treatment

    Several studies have also demonstrated the differentiation potential of DNMTi in cancer therapy. Liaoet al[76] showed that decitabine, an analogue of cytosine, in combinatorial treatment with a differentiation drug, could provide an effective route to enhancing cell differentiation (oligodendrocyte-like morphology and mRNA expression of terminal differentiation marker MBP) and inhibiting cell growth in two malignant glioma human cell lines. Andradeet al[77] tested the efficacy of zebularine,another DNMTi, in four pediatric SHH-MB cell lines (DAOY, ONS-76, UW402, and UW473). Zebularine decreased MB cell growth by targeting the Sonic Hedgehog pathway’s components (GLI1, SMO and PTCH1), evaluated at transcriptional levels.This provides a rationale for furtherin vivoinvestigation into the combination of zebularine with chemotherapy[77]. Valenteet al[75] tested two nonnucleosides DNMTi(compounds 2 and 5) in mouse MB stem cells isolated from fresh tumor specimens from Ptch1+/- mouse models; compound 2 significantly blocked cell proliferation,while compound 5 was stronger in differentiation potential evaluated by both βIIItubulin reverse transcriptase polymerase chain reaction and morphology images.

    CONCLUSION

    Aberrant epigenetic regulation has emerged as a key player in the genesis and progression of brain tumors, influencing malignant phenotypes at various stages of the disease and possibly underlying individual variability in drug response. In particular,the epigenome of brain tumors can be modulated by both cell-intrinsic (e.g., mutations)and cell-extrinsic (e.g., microenvironment) mechanisms, favoring those characteristics of CSCs responsible for cancer growth and disease progression. Reprogramming the epigenetic landscape in the cancer epigenome is among the most promising target therapies, both as a treatment itself and for reversing drug resistance. In this review,we discussed how epigenetic alterations regulate the "stem-like" properties of CSCs and the epigenetic drugs available to blockade epi-mutations. We have reported several examples of epidrugs in Phase I/II clinical trials, providing evidence on the benefit of using epidrugs as single agents or in combination-therapy in brain tumors.We believe this is thus a viable avenue for clinical trials aiming at the development of more affordable and efficient anticancer drugs and treatments.

    ACKNOWLEDGEMENTS

    We thank Ralf Mouthaan for manuscript editing.

    国产成人av激情在线播放| 欧美日韩中文字幕国产精品一区二区三区| 亚洲一区中文字幕在线| 久久久久免费精品人妻一区二区| 国产av麻豆久久久久久久| 国产探花在线观看一区二区| 99久久综合精品五月天人人| 操出白浆在线播放| 久久久久国内视频| 俄罗斯特黄特色一大片| 国产精品一区二区三区四区久久| 免费av毛片视频| 人人妻人人澡欧美一区二区| 亚洲电影在线观看av| 亚洲成人免费电影在线观看| 国产精品亚洲一级av第二区| 久久久久国产精品人妻aⅴ院| 国产av麻豆久久久久久久| 正在播放国产对白刺激| 日韩欧美在线二视频| 国产成人欧美在线观看| 亚洲人成电影免费在线| 免费搜索国产男女视频| 亚洲五月天丁香| 十八禁人妻一区二区| 这个男人来自地球电影免费观看| 夜夜爽天天搞| 日本a在线网址| 精品免费久久久久久久清纯| 18禁美女被吸乳视频| 性色av乱码一区二区三区2| 久久久精品欧美日韩精品| 女生性感内裤真人,穿戴方法视频| 亚洲熟妇熟女久久| 久久99热这里只有精品18| 色噜噜av男人的天堂激情| 亚洲乱码一区二区免费版| 国产精品久久久久久亚洲av鲁大| 久久欧美精品欧美久久欧美| 午夜视频精品福利| 人妻夜夜爽99麻豆av| 久久香蕉精品热| 老司机靠b影院| 免费在线观看影片大全网站| 亚洲电影在线观看av| 熟女电影av网| 夜夜看夜夜爽夜夜摸| 日本黄大片高清| 日本免费一区二区三区高清不卡| 99久久久亚洲精品蜜臀av| 此物有八面人人有两片| 成人国产一区最新在线观看| 99久久久亚洲精品蜜臀av| 大型黄色视频在线免费观看| 悠悠久久av| 国产高清视频在线播放一区| 男人舔女人的私密视频| 男人舔奶头视频| 国产男靠女视频免费网站| 久久欧美精品欧美久久欧美| 嫩草影视91久久| 国产av在哪里看| 日韩欧美精品v在线| 欧美av亚洲av综合av国产av| 午夜福利欧美成人| 国产私拍福利视频在线观看| 亚洲18禁久久av| 国产蜜桃级精品一区二区三区| 成人三级黄色视频| 神马国产精品三级电影在线观看 | 丰满人妻一区二区三区视频av | 成人av在线播放网站| 三级男女做爰猛烈吃奶摸视频| 免费在线观看视频国产中文字幕亚洲| 老司机深夜福利视频在线观看| 国语自产精品视频在线第100页| 久久国产精品影院| 麻豆久久精品国产亚洲av| 亚洲 国产 在线| 免费看a级黄色片| 午夜福利在线在线| 91字幕亚洲| 亚洲全国av大片| 成人国产综合亚洲| 亚洲自偷自拍图片 自拍| 精品日产1卡2卡| 日本在线视频免费播放| 一级毛片女人18水好多| 色综合婷婷激情| 无限看片的www在线观看| 两性午夜刺激爽爽歪歪视频在线观看 | 欧美日韩精品网址| 国产亚洲精品一区二区www| 亚洲va日本ⅴa欧美va伊人久久| 免费在线观看视频国产中文字幕亚洲| 亚洲国产日韩欧美精品在线观看 | 两个人视频免费观看高清| 午夜福利视频1000在线观看| 免费在线观看黄色视频的| 性欧美人与动物交配| 伦理电影免费视频| 91大片在线观看| 亚洲成av人片在线播放无| av福利片在线观看| 大型黄色视频在线免费观看| 97人妻精品一区二区三区麻豆| 宅男免费午夜| 久久久国产精品麻豆| av超薄肉色丝袜交足视频| 久久九九热精品免费| 国产区一区二久久| 亚洲欧洲精品一区二区精品久久久| 国产成人av激情在线播放| 在线观看舔阴道视频| 国产乱人伦免费视频| 国产一区二区三区视频了| 免费看a级黄色片| 美女高潮喷水抽搐中文字幕| 欧美一区二区国产精品久久精品 | 欧美一级毛片孕妇| 黑人欧美特级aaaaaa片| 成人永久免费在线观看视频| 久久香蕉精品热| 国产男靠女视频免费网站| 1024香蕉在线观看| 色综合站精品国产| 大型av网站在线播放| x7x7x7水蜜桃| 伦理电影免费视频| 欧美乱码精品一区二区三区| 久久国产精品影院| 精品电影一区二区在线| 国产高清激情床上av| 精品久久久久久久毛片微露脸| 久久亚洲真实| 久99久视频精品免费| 精品久久久久久成人av| 黄色毛片三级朝国网站| 国语自产精品视频在线第100页| 亚洲中文字幕一区二区三区有码在线看 | 午夜激情福利司机影院| 成熟少妇高潮喷水视频| 国产探花在线观看一区二区| 国产精品一区二区三区四区免费观看 | 人妻丰满熟妇av一区二区三区| 国产熟女xx| 午夜两性在线视频| 日韩精品青青久久久久久| 丰满的人妻完整版| 亚洲狠狠婷婷综合久久图片| 精品久久久久久久末码| 久久这里只有精品19| 日韩欧美国产在线观看| 国产亚洲精品av在线| 国产精品99久久99久久久不卡| 一区二区三区高清视频在线| 国产又色又爽无遮挡免费看| 久久久久久大精品| 禁无遮挡网站| 欧美中文综合在线视频| 亚洲国产欧美网| 亚洲一区二区三区色噜噜| 亚洲国产看品久久| 欧美性猛交黑人性爽| 日本 欧美在线| 操出白浆在线播放| xxx96com| 免费在线观看视频国产中文字幕亚洲| 国产高清视频在线播放一区| 久久精品夜夜夜夜夜久久蜜豆 | 欧美成人性av电影在线观看| 国产三级中文精品| 日本黄大片高清| 男女下面进入的视频免费午夜| 色综合站精品国产| 欧美黑人巨大hd| 成年女人毛片免费观看观看9| 一级毛片女人18水好多| 手机成人av网站| av天堂在线播放| 国产精品一区二区三区四区久久| 日日干狠狠操夜夜爽| 在线观看免费日韩欧美大片| 精品欧美一区二区三区在线| 国产激情欧美一区二区| 又黄又爽又免费观看的视频| 日韩精品青青久久久久久| 亚洲精品色激情综合| 国产亚洲精品久久久久5区| 日韩有码中文字幕| 亚洲欧美日韩高清在线视频| 欧美激情久久久久久爽电影| 人人妻,人人澡人人爽秒播| 99热只有精品国产| 日日干狠狠操夜夜爽| 日韩有码中文字幕| 国产高清videossex| 欧美乱色亚洲激情| 日韩免费av在线播放| 日韩欧美一区二区三区在线观看| 国产午夜精品久久久久久| 久久精品国产亚洲av高清一级| 国产成人av教育| 中文字幕最新亚洲高清| 在线十欧美十亚洲十日本专区| 久久婷婷成人综合色麻豆| 日韩成人在线观看一区二区三区| 88av欧美| 男女视频在线观看网站免费 | 熟女电影av网| 欧美黑人精品巨大| 免费观看人在逋| 欧美色视频一区免费| 一本精品99久久精品77| 欧美激情久久久久久爽电影| 亚洲乱码一区二区免费版| 最近视频中文字幕2019在线8| 别揉我奶头~嗯~啊~动态视频| 天堂影院成人在线观看| 欧美日韩精品网址| 亚洲自拍偷在线| 国产午夜精品久久久久久| 制服诱惑二区| 久久国产精品人妻蜜桃| 欧美日韩国产亚洲二区| 国产爱豆传媒在线观看 | 国产久久久一区二区三区| 免费一级毛片在线播放高清视频| 国产视频一区二区在线看| 国产一区在线观看成人免费| 天天添夜夜摸| x7x7x7水蜜桃| 亚洲精品av麻豆狂野| 亚洲精品中文字幕在线视频| 18禁国产床啪视频网站| 一个人观看的视频www高清免费观看 | 一本综合久久免费| 国产爱豆传媒在线观看 | 国产精品亚洲av一区麻豆| 国产成人精品久久二区二区免费| 熟女电影av网| av欧美777| 淫秽高清视频在线观看| 成人av在线播放网站| 国内精品久久久久精免费| 三级男女做爰猛烈吃奶摸视频| 美女午夜性视频免费| 制服丝袜大香蕉在线| 国产麻豆成人av免费视频| 脱女人内裤的视频| 免费在线观看影片大全网站| 亚洲av美国av| 一个人观看的视频www高清免费观看 | 日韩中文字幕欧美一区二区| 欧美在线黄色| 一个人免费在线观看电影 | 99久久精品国产亚洲精品| 中文字幕高清在线视频| 国产黄片美女视频| 18禁黄网站禁片免费观看直播| 最近视频中文字幕2019在线8| 精品无人区乱码1区二区| 国产99久久九九免费精品| 欧美三级亚洲精品| 亚洲熟妇中文字幕五十中出| 久久久国产成人免费| 最近最新中文字幕大全电影3| 在线观看66精品国产| 久久这里只有精品中国| 午夜福利在线观看吧| 欧美日韩亚洲国产一区二区在线观看| www日本在线高清视频| 在线视频色国产色| 男女之事视频高清在线观看| 两人在一起打扑克的视频| 99久久99久久久精品蜜桃| 神马国产精品三级电影在线观看 | 91大片在线观看| 又紧又爽又黄一区二区| 久久99热这里只有精品18| 中国美女看黄片| 中文在线观看免费www的网站 | 久久久久久久精品吃奶| 欧美另类亚洲清纯唯美| 国产精品一区二区精品视频观看| avwww免费| 久久精品人妻少妇| 日韩欧美精品v在线| 啪啪无遮挡十八禁网站| 成人18禁高潮啪啪吃奶动态图| 精品少妇一区二区三区视频日本电影| avwww免费| 丝袜人妻中文字幕| 国产在线精品亚洲第一网站| 国产99白浆流出| 国产av不卡久久| 亚洲成人久久爱视频| 真人做人爱边吃奶动态| 神马国产精品三级电影在线观看 | 精品高清国产在线一区| www.自偷自拍.com| av在线天堂中文字幕| 国产单亲对白刺激| 欧美 亚洲 国产 日韩一| www日本黄色视频网| 国产精品 国内视频| 成人18禁在线播放| 亚洲电影在线观看av| 亚洲18禁久久av| 日本一区二区免费在线视频| 色综合婷婷激情| 少妇人妻一区二区三区视频| 两个人视频免费观看高清| 精品久久久久久成人av| 欧美日韩精品网址| 久久天堂一区二区三区四区| 午夜免费激情av| 日日爽夜夜爽网站| 久久久久久九九精品二区国产 | 一区二区三区国产精品乱码| 大型黄色视频在线免费观看| 免费电影在线观看免费观看| 后天国语完整版免费观看| 麻豆一二三区av精品| 男女午夜视频在线观看| 亚洲av第一区精品v没综合| 欧美日韩亚洲国产一区二区在线观看| 999久久久精品免费观看国产| 久久久久久久午夜电影| 欧美丝袜亚洲另类 | 亚洲人成77777在线视频| 女人被狂操c到高潮| 老司机在亚洲福利影院| 丰满人妻一区二区三区视频av | 黄色成人免费大全| 亚洲精品色激情综合| 亚洲人与动物交配视频| 香蕉久久夜色| 亚洲精品久久成人aⅴ小说| 嫁个100分男人电影在线观看| 日韩大尺度精品在线看网址| ponron亚洲| 欧美一区二区国产精品久久精品 | 久久久久免费精品人妻一区二区| 美女免费视频网站| 啪啪无遮挡十八禁网站| 最近最新中文字幕大全电影3| 一区二区三区高清视频在线| 国产精品自产拍在线观看55亚洲| 精品日产1卡2卡| 熟女电影av网| 国产精品爽爽va在线观看网站| www国产在线视频色| 一级黄色大片毛片| 床上黄色一级片| 国产成人av教育| 亚洲av五月六月丁香网| 丰满的人妻完整版| 女警被强在线播放| 757午夜福利合集在线观看| 香蕉av资源在线| 中文字幕精品亚洲无线码一区| 日韩精品中文字幕看吧| 岛国视频午夜一区免费看| 18禁黄网站禁片午夜丰满| www日本在线高清视频| 国产精品免费一区二区三区在线| 国产欧美日韩一区二区精品| 熟妇人妻久久中文字幕3abv| 色综合亚洲欧美另类图片| 欧美日韩福利视频一区二区| 国内精品一区二区在线观看| 天堂√8在线中文| 免费看日本二区| 亚洲精品在线美女| 香蕉丝袜av| 亚洲在线自拍视频| 精品久久久久久,| 午夜精品在线福利| 好看av亚洲va欧美ⅴa在| 男女做爰动态图高潮gif福利片| 日韩有码中文字幕| 日本熟妇午夜| 1024香蕉在线观看| 麻豆国产av国片精品| 日韩国内少妇激情av| 色综合亚洲欧美另类图片| a级毛片在线看网站| 性欧美人与动物交配| 亚洲一区高清亚洲精品| 又大又爽又粗| 两个人免费观看高清视频| 亚洲精品中文字幕在线视频| 欧美一级a爱片免费观看看 | 久久精品aⅴ一区二区三区四区| 国产精华一区二区三区| 黑人操中国人逼视频| 久久精品国产清高在天天线| 成年免费大片在线观看| 国产精品久久久久久久电影 | 国产精品久久久久久久电影 | 曰老女人黄片| 老汉色av国产亚洲站长工具| 最新在线观看一区二区三区| 日本黄色视频三级网站网址| 国产黄a三级三级三级人| 欧美另类亚洲清纯唯美| 欧美一区二区精品小视频在线| 一级片免费观看大全| 久久久久久久久中文| 日韩欧美一区二区三区在线观看| 久久久精品国产亚洲av高清涩受| 久久国产精品影院| 亚洲无线在线观看| 成人国语在线视频| 欧美日韩国产亚洲二区| 国产av一区二区精品久久| 女警被强在线播放| 一级黄色大片毛片| 丝袜美腿诱惑在线| 50天的宝宝边吃奶边哭怎么回事| 亚洲国产精品sss在线观看| a在线观看视频网站| 亚洲欧美日韩东京热| 变态另类丝袜制服| 欧美中文综合在线视频| 91字幕亚洲| 午夜福利视频1000在线观看| 免费在线观看视频国产中文字幕亚洲| 国产欧美日韩精品亚洲av| √禁漫天堂资源中文www| 99国产极品粉嫩在线观看| 18禁裸乳无遮挡免费网站照片| 国产成+人综合+亚洲专区| 国产不卡一卡二| 最近最新中文字幕大全免费视频| 国内久久婷婷六月综合欲色啪| 欧美一级毛片孕妇| 熟妇人妻久久中文字幕3abv| 久久久久久久久久黄片| 亚洲欧美精品综合久久99| 国产爱豆传媒在线观看 | 国产精品亚洲美女久久久| 波多野结衣高清作品| 精品久久久久久成人av| 97碰自拍视频| tocl精华| 欧美日韩精品网址| 欧美日韩亚洲综合一区二区三区_| www.熟女人妻精品国产| 女人高潮潮喷娇喘18禁视频| 午夜精品在线福利| 欧美激情久久久久久爽电影| 哪里可以看免费的av片| 久久草成人影院| 我的老师免费观看完整版| 成人午夜高清在线视频| 久久 成人 亚洲| 日韩欧美三级三区| 又爽又黄无遮挡网站| 两个人的视频大全免费| 俺也久久电影网| 天天一区二区日本电影三级| 舔av片在线| 亚洲精品av麻豆狂野| 久久婷婷人人爽人人干人人爱| 黄色片一级片一级黄色片| 日韩中文字幕欧美一区二区| 男女之事视频高清在线观看| 男女那种视频在线观看| 欧美av亚洲av综合av国产av| 青草久久国产| 国产精品亚洲美女久久久| 亚洲熟妇熟女久久| 国产精品 欧美亚洲| 波多野结衣高清无吗| 高清毛片免费观看视频网站| 一本综合久久免费| 免费高清视频大片| 精品久久久久久成人av| 亚洲 国产 在线| 亚洲五月婷婷丁香| 国产精品一区二区三区四区免费观看 | 国内精品一区二区在线观看| 岛国视频午夜一区免费看| 亚洲av电影不卡..在线观看| 日本免费一区二区三区高清不卡| 国产精品久久久久久人妻精品电影| 脱女人内裤的视频| 午夜免费观看网址| 日本撒尿小便嘘嘘汇集6| 婷婷精品国产亚洲av| 国产成人精品久久二区二区91| 真人做人爱边吃奶动态| 中文字幕人妻丝袜一区二区| 伦理电影免费视频| 一区福利在线观看| 两人在一起打扑克的视频| 伊人久久大香线蕉亚洲五| 青草久久国产| 国产亚洲精品久久久久久毛片| 51午夜福利影视在线观看| 黑人操中国人逼视频| 视频区欧美日本亚洲| 午夜精品一区二区三区免费看| 丰满的人妻完整版| 老司机福利观看| 午夜影院日韩av| 非洲黑人性xxxx精品又粗又长| 12—13女人毛片做爰片一| 午夜福利视频1000在线观看| 久热爱精品视频在线9| 欧美日韩亚洲综合一区二区三区_| 国产精品电影一区二区三区| 99久久精品热视频| 特大巨黑吊av在线直播| 欧美+亚洲+日韩+国产| 桃红色精品国产亚洲av| 麻豆成人av在线观看| 日日干狠狠操夜夜爽| 黑人操中国人逼视频| 亚洲av电影在线进入| 日韩大尺度精品在线看网址| 免费av毛片视频| svipshipincom国产片| 国产精品久久久av美女十八| 久久久国产成人免费| 亚洲中文字幕日韩| 精品久久久久久久末码| 黄色成人免费大全| 99久久无色码亚洲精品果冻| 欧美 亚洲 国产 日韩一| 一夜夜www| 757午夜福利合集在线观看| 日本a在线网址| 99国产精品一区二区三区| 搡老妇女老女人老熟妇| 欧美日本视频| 国产一区二区三区在线臀色熟女| 中文字幕av在线有码专区| 色综合站精品国产| 日韩欧美在线乱码| 国产精品自产拍在线观看55亚洲| 中文字幕av在线有码专区| 久久久久久久午夜电影| 高潮久久久久久久久久久不卡| www.999成人在线观看| 淫妇啪啪啪对白视频| 久久久水蜜桃国产精品网| 99精品在免费线老司机午夜| 久久久水蜜桃国产精品网| 天堂av国产一区二区熟女人妻 | 无人区码免费观看不卡| 久久精品人妻少妇| 在线观看午夜福利视频| 夜夜看夜夜爽夜夜摸| 久久精品91无色码中文字幕| 夜夜看夜夜爽夜夜摸| 给我免费播放毛片高清在线观看| 亚洲熟女毛片儿| 亚洲色图 男人天堂 中文字幕| 成人av一区二区三区在线看| 久久久国产成人免费| www.自偷自拍.com| 97超级碰碰碰精品色视频在线观看| 久久人人精品亚洲av| 精品久久蜜臀av无| 亚洲国产看品久久| 亚洲avbb在线观看| 免费av毛片视频| 后天国语完整版免费观看| 麻豆久久精品国产亚洲av| 在线观看免费午夜福利视频| 亚洲 国产 在线| e午夜精品久久久久久久| 午夜福利18| 成人亚洲精品av一区二区| 婷婷六月久久综合丁香| 91大片在线观看| 欧美黑人欧美精品刺激| 久久人妻av系列| 亚洲精品粉嫩美女一区| 久久精品国产综合久久久| 一个人免费在线观看电影 | 亚洲欧美日韩东京热| 啪啪无遮挡十八禁网站| 黑人巨大精品欧美一区二区mp4| 午夜福利视频1000在线观看| 国产高清视频在线播放一区| 亚洲人成伊人成综合网2020| 天天躁狠狠躁夜夜躁狠狠躁| 法律面前人人平等表现在哪些方面| 久久天堂一区二区三区四区| 日韩大尺度精品在线看网址| 亚洲无线在线观看| 好看av亚洲va欧美ⅴa在| 岛国在线免费视频观看| 手机成人av网站| 亚洲国产欧美一区二区综合| 999久久久精品免费观看国产| 久久香蕉激情| 最好的美女福利视频网| 亚洲avbb在线观看| 久久中文看片网| 天堂av国产一区二区熟女人妻 | 国产成人欧美在线观看| 久久精品aⅴ一区二区三区四区| 日本一本二区三区精品| 九九热线精品视视频播放| 两性午夜刺激爽爽歪歪视频在线观看 | 男女做爰动态图高潮gif福利片| 国产av一区在线观看免费| 长腿黑丝高跟| 亚洲一区二区三区不卡视频| 日韩av在线大香蕉|