Abstract
Prostate cancer (PCa) is a heterogenous disease exhibiting both genetic and epigenetic deregulations. Epigenetic alterations are defined as changes not based on DNA sequence, which include those of DNA methylation, histone modification and chromatin remodeling. Androgen receptor (AR) is the main driver for PCa and androgen deprivation therapy (ADT) remains a backbone treatment for patients with PCa; however, ADT resistance almost inevitably occurs and advanced diseases develop termed castration resistant PCa (CRPC), due to both genetic and epigenetic changes. Due to the reversible nature of epigenetic modifications, inhibitors targeting epigenetic factors have become promising anti-cancer agents. In this chapter, we focus on recent studies about the dysregulation of epigenetic regulators crucially involved in the initiation, development and progression of PCa and discuss the potential use of inhibitors targeting epigenetic modifiers for treatment of advanced PCa.
1. Introduction
Prostate cancer (PCa) is the second most commonly diagnosed cancer in the men and one of the leading causes of cancer-related death (Sung et al., 2021). According to Globocan estimates, approximately 1.4 million of new PCa cases were diagnosed worldwide in 2020 (Sung et al., 2021). Androgen receptor (AR), a nuclear hormone receptor and transcription factor that mediates signaling of testosterone and 5-α-dihydrotestosterone (DHT), not only is essential for normal prostate development but also remains a main driver in PCa (Watson et al., 2015). Accordingly, androgen deprivation therapy (ADT) has been a backbone of treatment for PCa patients (Huggins and Hodges, 1972). However, most PCa patients eventually become refractory to ADT and, almost inevitably, the disease develops to castration-resistant prostate cancer (CRPC) (Watson et al., 2015). Patients with CRPC often develop metastases (termed metastatic CRPC or mCRPC), mainly in the skeleton, and become incurable due to a lack of effective therapy (Tsuzuki et al., 2016). CRPC exhibits a diverse disease presentation with variable outcomes, including neuroendocrine PCa (NEPC) (Conteduca et al., 2019). NEPC is relatively rare at initial diagnosis of PCa but its incidence increases dramatically following ADT (thus termed therapy induced-NEPC or t-NEPC) and ranges from ~17–30% based on different clinical studies (Patel et al., 2019a). The available therapy for CRPC is very limited, therefore, it remains an urgent task to dissect precise mechanisms responsible for CRPC progression and develop new methods to overcome therapy resistance.
Genetic and epigenetic alterations, as well as changes in tumor microenvironment (TME), collectively contribute to malignant transformation of cancer (You and Jones, 2012). Whole genome sequencing (WGS) studies in patient samples have identified numerous genomic alterations in primary and advanced PCa, which include the loss of tumor suppressor genes such as PTEN, RB and TP53, amplification of oncogenes such as AR and MYC, formation of structural variants such as TMPRSS2-ERG, and mutations of genes important for various signaling pathways (Quigley et al., 2018, Armenia et al., 2018, Robinson et al., 2015, Cancer Genome Atlas Research, 2015, Grasso et al., 2012, Tomlins et al., 2005, Taylor et al., 2010). On the other hand, epigenetics, here by a relatively loose definition, refers to biological processes that regulate gene expression and function without altering DNA sequences (Sharma et al., 2010). Genetic information or DNA of mammalian cells is organized in the form of chromatin and the building unit of chromatin is termed as nucleosomal core particle, which consists of a histone octamer (two copies of each of the four core histones H2A, H2B, H3 and H4) surrounded by double-stranded DNA (Felsenfeld and Groudine, 2003). Epigenetic regulations include DNA methylation, histone modification, chromatin remodeling, amongst other mechanisms. DNA methylation mainly occurs in CpG dinucleotides in which a methyl group is added to the 5’-carbon position of cytosine by a family of DNA methyltransferases (DNMTs) (Greenberg and Bourc’his, 2019). Histone modification refers to covalent post-translational modification (PTM) of histone proteins. The cellular machineries that mediate the epigenetic regulation can be grouped as writers, erasers and readers (Zhao et al., 2021). The writer refers to an enzyme that deposits a specific histone modification or DNA methylation. The common writer enzymes include histone acetyltransferases (HATs), histone methyltransferases (HMTs) and DNA methyltransferases (DNMTs). Histone modifications and DNA methylation are reversible and can be removed by erasers. Common erasers include histone deacetylases (HDACs), histone demethylases (HDMs) and TET family proteins. Readers refer to a set of specialized proteins that recognize and bind specific histone modification or DNA methylation and convey epigenetic information to downstream effectors (Zhao et al., 2021). The change of chromatin accessibility is mediated by chromatin remodeling complexes such as the SWI/SNF complex (Clapier et al., 2017). DNA methylation, histone modifications and chromatin remodeling are important epigenetic mechanisms to mediate gene expression, and their deregulations act independently and/or cooperatively in the process of PCa initiation and progression and during the development of drug resistance (Kumaraswamy et al., 2021, Kukkonen et al., 2021, Natesan et al., 2019, Ruggero et al., 2018). In this book chapter, we will use prostate cancer as an example to highlight the recent discoveries on how to deregulation in DNA methylation and histone modification modifiers and chromatin remodelers contributes to the cancer initiation and progression, as well as potential targeting strategies, as summarized in Figure 1.
Figure 1. Overview of key epigenetic regulators and targeting drugs in PCa.
This schematic illustration only covers a set of selectively focused epigenetic modifications including DNA methylation (top), histone methylation and acetylation (middle), and chromatin remodeling (bottom) involved in PCa. Certain key writers, erasers and readers, as well as the respective targeting compounds, are listed. *, the compound used in the ongoing clinical trials in PCa as shown in Table 1. Many other histone modifications, epigenetic modifiers and targeting drugs, which might have equally important functions in PCa, are not listed. The figure is created with BioRender.com.
2. DNA Methylation in prostate cancer
Patterns of cytosine methylation within CpG dinucleotide sequences are important for regulating gene expression, DNA damage repair, DNA recombination and DNA replication (Chin et al., 2011). Cytosine methylation is de novo ‘written’ by DNMT3A and DNMT3B and maintained by DNMT1, which transfer a methyl group from S-adenosylmethionine, a methyl donor, to the 5’-carbon of the cytosine ring to form 5-methylcytosine (5mC) (Greenberg and Bourc’his, 2019). 5mC can be ‘erased’ through sequential oxidation by Ten-eleven translocation (TET) family proteins, with the assistance of base excision repair (BER) pathway, first to 5-hydroxymethyl cytosine (5hmC), then 5-formyl cytosine (5fC) and finally 5-carboxyl cytosine (5caC) (Ikeda and Kinoshita, 2009). CpG islands refer to the CpG-rich regions of ~200 bp to several kilobases in length, usually located near the promoters of highly expressed genes (Illingworth et al., 2008). CpG islands are often subject to hyper-methylation in human tumors including PCa, compared to normal tissues (Nowacka-Zawisza and Wisnik, 2017). Hypermethylated CpG islands are often associated with target gene silencing (Greenberg and Bourc’his, 2019).
Mechanistically, 5mC can regulate gene expression by recruiting readers such as methyl-CpG binding proteins (MBP1, MBP2, MBP3), Kaiso, and methyl CpG-binding protein-2 (MeCP2) (Buck-Koehntop and Defossez, 2013, Wu et al., 2015, Jones et al., 1998, Nan et al., 1998). Once bound to promoter-associated 5mC, methyl-CpG-binding domain (MBD) family proteins can further recruit corepressors such as HDACs, thereby mediating repression of target gene transcription (Du et al., 2015). Meanwhile, DNA methylation can profoundly modulate binding of various DNA-binding proteins such as transcription factors (TFs) and thus directly influences gene activity (Zhu et al., 2016). In this case, 5mC may either inhibit or promote TF binding (Ren et al., 2018, Zhu et al., 2016).
In 1987, Bedford and van Helden reported that global DNA methylation was significantly lower in metastatic PCa compared to benign prostatic hyperplasia (BPH) (Bedford and van Helden, 1987). This is the first study to demonstrate a correlation between global hypomethylation and metastatic potential of PCa. In general, promoter hypomethylation and gene body hypermethylation are positively correlated with gene expression (Jones, 2012). In PCa, DNA hypomethylation has been linked to higher expression of genes and usually occurs later in PCa progression and contributes to tumor metastasis (Yegnasubramanian et al., 2004). Over the last decade, several studies have compared genome-wide DNA methylation patterns in benign prostate tissues and various stages of PCa (Kirby et al., 2017, Bhasin et al., 2015, Geybels et al., 2015, Cancer Genome Atlas Research, 2015, Zelic et al., 2015). Very recently, Zhao et al. used whole genome bisulfide sequencing (WGBS) and further confirmed that advanced PCa is overall less methylated than primary PCa, which is predominantly less methylated than benign prostate (Zhao et al., 2020). Numerous oncogenic factors involved in various signaling pathways are reported to be hypomethylated in PCa including Urokinase plasminogen activator (uPA) (Pakneshan et al., 2003), Heparanase (HSPE) (Hulett et al., 1999), Cytochrome P450 1B1 (CYP1B1) (Tokizane et al., 2005), and wingless-related MMTV integration site 5A (WNT5A) (Wang et al., 2007). In the aforementioned study, Zhao et al. demonstrated promoter hypomethylation at AR and key androgen-responsive genes including KLK3 and NKX3–1 in advanced PCa compared to benign prostate samples (Zhao et al., 2020). Global DNA hypomethylation has also been proposed to be correlated with genetic instability by promoting formation of more open chromatin (Ehrlich, 2002). Therefore, DNA hypomethylation potentially contributes to PCa development and progression through altering transcriptomics and/or genome stability.
Conversely, DNA hypermethylation is one of the most commonly observed phenomena and best characterized epigenetic alterations in PCa. A plethora of genome-wide DNA methylation analyses have been performed which suggest that differential methylation among distal and genic regions is one of key drivers of PCa tumorigenesis and progression (Wang et al., 2016b). For example, Zhao et al. reported that about 22% of CRPC tumors exhibit a hypermethylation subtype termed a CpG methylator phenotype (CMP), which is enriched with somatic mutations of DNMT3B, TET2, BRAF and IDH1 (Zhao et al., 2020). In contrast to promoter hypomethylation, hypermethylation of gene promoters often coincides with transcriptional repression of target genes, most of which are tumor suppressor genes (TSGs) such as adenomatosis polyposis coli (APC) (Richiardi et al., 2009), retinoic acid receptor β (RARβ) (Jeronimo et al., 2004) and RAS-associated domain family 1 (RASSF1) (Liu et al., 2002, Kuzmin et al., 2002), DNA damage repair related genes such as Glutathione S-Transferase Pi 1 (GSTP1) (Mahon et al., 2014, Lee et al., 1994) and O-6-methylguanine-DNA methyltransferase (MGMT) (Gerson, 2004), hormonal response related genes such as AR (Jarrard et al., 1998), ER-α (Sasaki et al., 2002) and ER-β (Lau et al., 2000), as well as genes involved in cell cycle control, apoptosis and cell adhesion such as cyclin dependent kinase inhibitor 2A (CDKN2A) (Herman et al., 1995), Target of Methylation-induced Silencing 1 (TMS1) (Das et al., 2006) and E-cadherin (CDH1) (Graff et al., 1995, Li et al., 2001).
Measurement of global or targeted DNA methylation can be used as biomarkers to help the risk stratification of PCa patients. For instance, ConfirmMDx, an epigenetic test that evaluates the methylation status of three genes, GSTP1, APC and RASSF1, can distinguish true negative prostate biopsies from occult PCa, therefore avoiding unnecessary repeat prostate biopsies of patients (Partin et al., 2014). Likewise, the methylation panel of GSTP1, together with those of GAS6 and HAPLN3, was identified by Patel et al. to separate PCa and benign prostate tissues in a sensitive and specific way (Patel et al., 2019b).
While 5mC is commonly associated with transcriptional repression, further conversion to 5hmC is related to transcriptional activation (Xu et al., 2011). The laboratory of Dr. Felix Feng has recently profiled 5hmC among 145 PCa samples representing different disease stages (including 52 localized PCa and 93 mCRPC) and integrated datasets of WGS, WGBS and RNA-seq from the same samples (Sjostrom et al., 2022). In this study, 5hmC in mCRPC was shown to mark the activation of major cancer driver genes (such as AR and EZH2) at both gene-regulatory sites and their downstream binding sites (Sjostrom et al., 2022), consistent with the transcriptional changes during disease progression captured by RNA-seq (Bolis et al., 2021). Since metastatic PCa is notoriously heterogenous and hard to biopsy, circulating tumor DNA (ctDNA) obtained from liquid biopsy provides an alternative approach for understanding the underlying tumor biology (Vandekerkhove et al., 2019, Wu et al., 2020). Indeed, 5hmC level in ctDNA represents the individual advanced tumors, making it a potential liquid biomarker for mCRPC (Sjostrom et al., 2022, Bolis et al., 2021). A very exciting work done by Chen et al. performed plasma DNA methylome analysis from patients with localized (60 samples) and metastatic PCa (175 samples) using the cell-free methylated DNA immunoprecipitation sequencing (cfMeDIP-seq) technology (Chen et al., 2022, Shen et al., 2018). They observed global hypermethylation in metastatic samples, together with hypomethylation in the pericentromeric regions (Chen et al., 2022). This study indicated that the cell-free DNA methylome may accurately distinguish different disease stages, highlighting a potential use in PCa diagnosis and prognosis (Chen et al., 2022).
Deregulation of DNMT occurs commonly in cancer and is associated with tumorigenesis (Zhang and Xu, 2017). DNMT1 is often overexpressed in localized and metastatic PCa when compared to BPH (Valdez et al., 2013, Zhang et al., 2015). Increased expression of DNMT3A and DNMT3B led to hypermethylation at a substantial subset of CpG sites in PCa (Kobayashi et al., 2011). TET2 binds AR and TET2 loss is correlated with PCa progression (Nickerson et al., 2017). To target aberrant DNA hypermethylation, DNMT inhibitors such as Azacytidine and Decitabine have been developed and tested in various types of cancers including PCa (Cheng et al., 2019). Currently, there are two ongoing clinical trials using Decitabine/Cedazuridine together with Enzalutamide (NCT05037500), or Guadecitabine (SGI-110) together with Perbrolizumab (NCT02998567), for the treatment of CRPC patients (see Table 1).
Table I.
Selective ongoing clinical trials of epigenetic drugs for the treatment of PCa (Source: https://clinicaltrials.gov/)
Trial ID | Drug name | Phase | Conditions | Status |
---|---|---|---|---|
DNMT inhibitors | ||||
NCT05037500 | Decitabine/Cedazuridine | I/II | mCRPC (+Enza) | Recruiting |
NCT02998567 | Guadecitabine(SGI-110) | I | CRPC (+Pembrolizumab) | Active |
LSD1/KMD1A inhibitors | ||||
NCT04628988 | CC-90011 | I | mCRPC(+Abi/Pred) | Recruiting |
EZH2 inhibitors | ||||
NCT03460977 | PF-06821497 | I | mCRPC (alone or with SOC) | Recruiting |
NCT04179864 | Tazemetostat | I/II | mCRPC (+Enza or Abi/Pred) | Recruiting |
NCT04846478 | Tazemetostat | I | mCRPC(+Talazoparib) | Recruiting |
NCT04104776 | CPI-0209 | II | mCPRC | Recruiting |
NCT03480646 | CPI-1205 | I/II | mCRPC (+Enza or Abi/Pred) | Active |
NCT04388852 | DS3201(Valemetostat) | I | CRPC(+Ipilimumab) | Recruiting |
p300/CBP inhibitor | ||||
NCT03568656 | CCS1477 | I/II | mCRPC(+Enza or Abi or Daro) | Recruiting |
HDAC inhibitors | ||||
NCT04703920 | Belinostat | I | mCRPC(+Talazoparib) | Recruiting |
BET protein inhibitors | ||||
NCT05252390 | Nuv-868 | I/II | mCRPC(alone or +Enza or Talazoparib) | Recruiting |
NCT04471974 | ZEN-3694 | II | mCRPC(+Enza and Pembrolizumab | Recruiting |
NCT04986423 | ZEN-3694 | II | CRPC(+Enza) | Recruiting |
Abbreviation in the table: mCRPC, Metastatic castration-resistant prostate cancer; Enza, Enzalutamide; Abi, Abiraterone acetate; SOC, Standard of Care; Pred, Prednisone/Prednisolone; Daro, Darolutamide
3. Histone methylation in prostate cancer.
Histones are a group of highly conserved, highly basic proteins that are responsible for chromatin organization and compaction by folding DNA into the nucleus (Marino-Ramirez et al., 2005). There are four core histones, namely H2A, H2B, H3 and H4. Amino acid residues of histones, especially those in the unstructured N-terminal tails, are known to be potential sites for various types of post-translational modifications (PTMs), such as methylation, acetylation, phosphorylation and ubiquitination (Allis and Jenuwein, 2016). Histone methylation and acetylation have been relatively intensively studied in cancer and hence, we will focus the next sections on the recent findings showing the involvement of these modifications in PCa.
Histone methylation and acetylation are established, respectively, by histone lysine methyltransferases (KMTs) and histone acetyltransferases (HATs), and can be removed, respectively, by histone lysine demethylases (KDMs) and histone deacetylases (HDACs). Histone methylation is critically involved in a wide variety of biological processes ranging from transcriptional regulation to heterochromatin formation (Greer and Shi, 2012). Lysine (Lys or K) and arginine (Arg or R) residues serve as the most common sites of methylation. Here we mainly discuss about lysine methylation. The histone H3 lysine 4, 9, 27, 36 and 79 (H3K4, H3K9, H3K27, H3K36 and H3K79) and histone H4 lysine 20 (H4K20) are among the most extensively studied sites of histone lysine methylation. Lysine can be mono-, di- or tri-methylated (Kme1, Kme2, or Kme3) on its ε-amine group (Greer and Shi, 2012). The degree of histone lysine methylation is controlled by the relative activities of site-specific methyltransferases and demethylases.
3.1. H3K4 methylation
Highly methylated H3K4, such as H3K4me3 and H3K4me2, is localized to the promoter-proximal regions of actively transcribed genes, with H3K4me3 exhibiting a more punctuate pattern than H3K4me2 (Ng et al., 2003, Santos-Rosa et al., 2002). H3K4me3 and H3K4me2 are associated with active transcription. H3K4me1, on the other hand, is a marker of gene enhancer (Creyghton et al., 2010, Rada-Iglesias et al., 2011). While the poised enhancers are marked by a presence of both H3K4me1 and H3K27me3 and a lack of H3K27 acetylation (H3K27ac), those active ones carry both H3K4me1 and H3K27ac and a lack of H3K27me3 (Creyghton et al., 2010, Rada-Iglesias et al., 2011). In mammalian cells, methylation of H3K4 is catalyzed by the KMT2/MLL family methyltransferases: KMT2A/MLL1, KMT2D/MLL2, KMT2C/MLL3, KMT2B/MLL4, KMT2F/SETD1A and KMT2G/SETD1B. Another SMYD family of H3K4 methyltransferases include SMYD1, SMYD2, and SMYD3. On the contrary, H3K4 demethylation is primarily catalyzed by KDM1A/LSD1, KDM1B/LSD2, KDM5A/JARID1A, KDM5B/JARID1B, KDM5C/JARID1C and KDM5D/JARID1D (Zhao et al., 2021).
The H3K4 methyltransferase KMT2A/MLL1 can function as a co-activator for AR in CRPC (Malik et al., 2015, Grasso et al., 2012). Here, AR directly interacts with the MLL complex via the menin subunit, and the inhibition of menin-MLL complex suppresses AR signaling (Malik et al., 2015). Notably, a phase I clinical trial with revumenib (SNDX-5613), a potent and selective oral inhibitor of the menin–MLL interaction, demonstrated strong efficacy in MLL-rearranged or NPM1-mutated acute leukemia (Issa et al., 2023). It would be worthwhile to test this inhibitor in advanced PCa. KMT2D/MLL2 is mutated in 8.6% of PCa cases (Grasso et al., 2012). KMT2D/MLL2 promotes prostate tumor growth through epigenetically activating LIFR and KLF4, which are involved in PI3K/AKT and epithelial-mesenchymal transition (EMT) pathways (Lv et al., 2018). KMT2F/SETD1A promotes tumor cell proliferation in metastatic CRPC by regulating FOXM1 transcription (Yang et al., 2020). Upregulation of SMYD3 expression occurs frequently in multiple cancer types including PCa (Huang and Xu, 2017). SMYD3 was shown to promote prostate tumorigenesis through upregulation of AR expression (Liu et al., 2013).
LSD1 (also known as KDM1A) is the first identified histone demethylase (Shi et al., 2004) and also represents one of the most widely studied KDMs in cancers including PCa (Rotili and Mai, 2011). LSD1 is overexpressed in patients with advanced PCa compared to normal tissues (Crea et al., 2012). LSD1 was reported to interact with AR and activate AR signaling through demethylating histone H3K9 methylation (Wissmann et al., 2007, Metzger et al., 2005, He et al., 2018). Cai et al. demonstrated that LSD1 can play dual roles in PCa (Cai et al., 2014). In the presence of high concentration of androgen, LSD1 acts as a co-repressor of AR expression by inducing demethylation of H3K4me1 and H3K4me2 at AR intronic region, and additionally, AR recruits LSD1 for mediating silencing of AR-repressed gene targets (Cai et al., 2011). However, in CRPC cells where androgen level is generally low, the expression of AR and AR-repressed genes is increased due to an impaired recruitment of LSD1, which in turn promotes CRPC cell proliferation (Cai et al., 2011). LSD1-mediated epigenetic reprogramming also drives PCa progression through regulating CENPE expression (He et al., 2018, Liang et al., 2017). However, it remains to be elucidated as of the molecular determinants responsible for LSD1 specificity towards H3K4 versus H3K9 demethylation under different biological contexts. In addition to its role in demethylating histone substrates (He et al., 2018), LSD1 also demethylates a number of non-histone substrates such as p53 and FOXA1. LSD1-mediated repression of p53 functionality was reported to be relevant during oncogenesis (Huang et al., 2007). More recently, LSD1 was shown to demethylate FOXA1, an oncogenic TF and functional partner of AR in PCa, at its residue K270, which positively regulates FOXA1’s binding to chromatin targets and then enhances AR’s transcriptional activity (Gao et al., 2020). LSD1 blockade, alone or in synergy with AR antagonist treatment, dramatically decreased PCa growth in vivo (Gao et al., 2020). LSD1 also has demethylase-independent functions to promote survival of the AR-independent prostate cancer (Sehrawat et al., 2018).
A number of LSD1 inhibitors, which include CC-90011 (Kanouni et al., 2020), SP-2577 (Soldi et al., 2020), ORY-1001 (Maes et al., 2018), TCP (Yang et al., 2007) and GSK-2879552 (Mohammad et al., 2015), have been reported and are under clinical evaluation for the treatment of cancers, either alone or in combination with other drugs (Fang et al., 2019, Cheng et al., 2019). An ongoing clinical trial (NCT04628988) is testing the LSD1 inhibitor CC-90011 together with ADT in mCRPC (Table 1).
The KDM5 family of Jumonji domain-containing KDMs are H3K4-specific and also frequently altered in PCa (Vieira et al., 2014). For instance, KDM5A is upregulated in PCa (Vieira et al., 2014). KDM5B was reported to be frequently upregulated in PCa tissues and associate with AR to regulate its transcriptional activity (Xiang et al., 2007b). KDM5B is also a key regulator of PI3K/AKT signaling in PCa through directly binding the PIK3CA promoter (Li et al., 2020). Loss of KDM5B results in a significant reduction of P110α and PIP3 levels and inhibition of the proliferation of PCa cells (Li et al., 2020). KDM5C overexpression in PCa was found to be associated with a reduced biochemical recurrence-free survival in patients after prostatectomy (Stein et al., 2014). KDM5C is highly expressed in metastatic PCa and promotes tumor growth and metastasis (Lemster et al., 2022). In contrast, KMD5D, a male-specific protein, was shown to suppress the invasion of PCa cells via demethylating H3K4me3 on the promoters of invasion-associated genes or recruiting co-repressor ZMYND8 (Li et al., 2016, Padeken et al., 2022). Furthermore, KDM5D is associated with altered docetaxel sensitivity in PCa through modulating the AR signaling (Komura et al., 2016). Loss of KDM5D expression results in aggressive PCa and confers a poorer prognosis in PCa patients (Komura et al., 2018). Taken together, these findings underscore the roles of KDMs in PCa initiation, progression and metastasis, indicating that KDMs might represent attractive therapeutic targets.
The reader proteins for H3K4 methylation also play a role in PCa progression. For example, an in vivo open reading frame (ORF) screening identified PYGO2, a H3K4me2 and H3K4me3 reader, as a driver for metastatic PCa (Lu et al., 2018). While overexpression of PYGO2 promotes PCa tumor growth and invasion, depletion of PYGO2 has the opposite effects in vitro and in vivo (Lu et al., 2018). Upregulation of PYGO2 is associated with higher Gleason score and metastasis in PCa patients, further indicating PYGO2 as a potential therapeutic target in advanced PCa (Lu et al., 2018). A recent follow-up study showed that targeting PYGO2 through genetic or pharmacological inhibition enhanced cytotoxic T cell responses and sensitized PCa to immunotherapy (Zhu et al., 2023).
3.2. H3K9 methylation
H3K9 methylation is deposited by the KMT1 writer enzymes (including KMT1A/SUV39H1, KMT1B/SUV39H2, KMT1C/G9a/EHMT2, KMT1D/GLP/EHMT1, KMT1E/SETDB1 and KMT1F/SETDB2), recognized by readers such as HP1α and MPP8, and eliminated by erasers such as the KDM3 (KDM3A-C/JHDM2A-C) and KDM4 (KDM4A-E/JMJD2A-E) family enzymes (Padeken et al., 2022). H3K9me2 or H3K9me3 is a gene silencing-related histone PTM and often found in high levels at constitutive heterochromatin regions (Barski et al., 2007). H3K9me3 mediates transcriptional silencing of various transposable elements (TEs) and regular genes as well. Malfunction of H3K9 methylation-associated regulators and modifier enzymes can profoundly affect either the level or the genomic distribution of H3K9 methylation, thus resulting in de-regulation of gene expression and/or genome instability during the course of pathogenesis (Padeken et al., 2022). H3K9 methylation writers, readers and erasers were reported to influence PCa development and progression by a variety of different mechanisms.
KMT1B/SUV39H2 is the writer responsible for H3K9me1/2 deposition and its overexpression was reported in PCa (Vieira et al., 2014). Through yeast two hybrid screening, SUV39H2 was identified as a coactivator of AR (Askew et al., 2017). Similarly, the H3K9me3 writer KMT1E/SETDB1 was also upregulated in PCa (Strepkos et al., 2021, Sun et al., 2014). Knockdown of SETDB1 induced the G0/G1 cell cycle arrest and inhibited PCa cell proliferation and migration (Sun et al., 2014). Dutta et al. discovered a NKX3.1-G9a-UTY transcriptional axis, which is essential for prostate differentiation (Dutta et al., 2016). More studies, however, are required to decipher the role of this axis in PCa. Notably, H3K9 methylation was found to be essential to drive antiandrogen resistance in advanced PCa (Baratchian et al., 2022). ADT induced de-repression of retroelements (REs) leading to a phenomenon termed viral mimicry, which activates the immune-related signaling pathways and thus inhibits tumor growth (Baratchian et al., 2022). Overexpression of H3K9me3 writers (for example, GLP or G9a) conferred drug resistance through mediating repression of antiandrogen-induced activation of REs, whereas inhibition of these writers and readers (such as CBX5, also known as HP1α) restored RE expression and abolished antiandrogen resistance (Baratchian et al., 2022). Therefore, a combined treatment with antiandrogen and inhibitors targeting H3K9me3 regulators serves as a promising therapeutic strategy for treating CRPC, based on the linkage between H3K9me3-mediated RE silencing and tumor microenvironment.
The H3K9me3 reader HP1α showed elevated expression in NEPCs, which was found to be associated with poor prognosis (Ci et al., 2018). HP1α promoted neuroendocrine trans-differentiation and silencing of HP1α inhibited NEPC cell proliferation (Ci et al., 2018). Another H3K9me3 reader MPP8 was reported to repress E-cadherin through binding to H3K9 methylation and promote EMT phenotypes in PC3 cells (Sun et al., 2015).
H3K9 methylation is removed by the KDM3 and KDM4 family of erasers, which are often highly expressed in PCa (Bjorkman et al., 2012). KDM3A (also known as JMJD1A) regulates activities of AR and c-Myc and promotes prostate cancer progression (Fan et al., 2016). Acetylation of KDM3A by P300 blocks its degradation and enhances AR activity in CRPC (Xu et al., 2020). In addition, KDM3A promotes alternative splicing of AR to generate the constitutively active AR spliced variant 7 (AR-V7), the formation of which represents one of the main ADT resistance mechanisms (Fan et al., 2018). Collectively, multifaceted roles of KDM3A in regulating the AR activation, c-Myc activity and AR-V7 alternative splicing suggested it to be a promising therapeutic target for PCa. KDM3B (also known as JMJD1B) was identified as a key regulator of cell proliferation in CRPC cells by a focused shRNA screening (Sarac et al., 2020). KDM3C has been shown to have a synthetic lethal relationship with AR and the AR-negative PCa cells are sensitive to KDM3C inhibition (Yoshihama et al., 2021). Knockdown of PHF8, another H3K9 demethylase, showed that it plays a role in PCa cell proliferation, migration and invasion (Bjorkman et al., 2012).
KDM4A and KDM4D form complexes with ligand-bound AR and function as AR coactivators (Shin and Janknecht, 2007). KDM4A (also known as JMJD2A) is a coactivator of E2F1 and regulates PCa metabolism through transcriptional regulation of pyruvate dehydrogenase kinase (PDK) (Wang et al., 2016a). Overexpression of KDM4A is positively correlated with Gleason score and metastasis in human PCa (Kim et al., 2016). Kim et al. further identified a KDM4A-ETV1-YAP1 axis that operates to promote PCa initiation (Kim et al., 2016). KDM4B interacts with AR and regulates its stability and activity (Coffey et al., 2013). In addition, KDM4B promotes alternative splicing to generate AR-V7, thus contributing to the development of CRPC (Duan et al., 2019). Furthermore, KDM4B promotes CRPC cell proliferation through physically interacting with c-Myc to activate a set of metabolic genes such as LDHA (Wu et al., 2021). Overexpression of KDM4B may act promote the recruitment of AR to the enhancer of c-Myc gene and induces its expression, which causes anti-androgen resistance (Tang et al., 2020). Together, targeting KDM4B may theoretically repress the nodes of AR-FL, AR-V7 and c-Myc, making it an attractive therapeutic strategy in PCa. In fact, small-molecule inhibitors targeting KDM4 were shown to suppress PCa cell proliferation (Duan et al., 2015, Chu et al., 2014).
In summary, H3K9 methylation and its various regulators profoundly affect PCa cell proliferation and survival, as well as PCa-TME interaction, through different molecular mechanisms. Targeting the H3K9 methylation-related modifiers and regulators provide promising clinical strategies to fight against PCa, which awaits additional studies.
3.3. H3K27 methylation
Trimethylation of histone H3 lysine 27 (H3K27me3) is another histone PTM closely associated with transcriptional repression (Di Croce and Helin, 2013). H3K27me3 deposition is carried out by Polycomb Repressive Complex 2 (PRC2), a multi-subunit methyltransferase complex that contains Enhancer of zeste homolog 2 (EZH2) as the catalytic subunit. PRC2 core complex is composed of EZH2 or related EZH1, EED, SUZ12 and RbAP46/48 (Guo et al., 2021). To gain a comprehensive view of PRC2 function in development and cancer, please refer to recent comprehensive reviews (Kim and Kingston, 2022, Piunti and Shilatifard, 2021, Blackledge and Klose, 2021).
EZH2 mRNA and protein levels were found to be progressively increased in metastatic PCa samples compared with benign prostate tissues (Varambally et al., 2002). In a clinical PCa cohort, higher expression of EZH2 was found to be correlated with a worse prognosis (Varambally et al., 2002). EZH2 overexpression promotes the proliferative and invasive capacity of PCa cells (Varambally et al., 2002, Varambally et al., 2008). Loss of EZH2-targeting microRNAs such as micro-RNA-101, which negatively regulates EZH2 at a posttranscriptional level, contributes to overexpression of EZH2 during PCa progression (Varambally et al., 2008).
As an oncogenic driver of PCa, overexpression of EZH2 inhibits the expression of TSGs through deposition of H3K27me3, which then promotes tumorigenesis (Jain and Di Croce, 2016). In addition to this canonical role of EZH2 in H3K27me3 deposition and gene silencing, EZH2 also carries non-canonical oncogenic functions independent of PRC2 and H3K27me3 (Wang and Wang, 2020). For instance, EZH2 can methylate STAT3 to promote its activity in glioblastoma stem-like cells (Kim et al., 2013). EZH2 methylates elongin A to regulate target gene transcription in embryonic stem cells (Ardehali et al., 2017). Besides its canonical function to repress TSGs in PCa, EZH2 was reported to be involved in transcriptional activation of AR target genes, and PI3K/AKT phosphorylation of EZH2 at serine 21 was proposed to promote such a gene-activation effect by EZH2 (Xu et al., 2012). EZH2 was further demonstrated to directly bind the AR gene promoter to activate its transcription, thereby potentiating AR signaling in PCa (Kim et al., 2018). A more recent study from the same group showed that EZH2 can methylate FOXA1 at the K295 residue to enhance FOXA1’s stability, thereby regulating the cell cycle-related genes in PCa cells (Park et al., 2021a). Furthermore, Yi et al. demonstrated that EZH2 increases rRNA 2’-O methylation and regulates translation through its direct interaction with fibrillarin in PCa (Yi et al., 2021). EZH2 also plays a significant role in regulating lineage plasticity, drug resistance and antitumor immunity (Morel et al., 2021, Abida et al., 2019, Bai et al., 2019, Xiao et al., 2018, Davies et al., 2021, Berger et al., 2019, Puca et al., 2018, Zhang et al., 2018, Dardenne et al., 2016, Clermont et al., 2015, Ku et al., 2017).
Because of the aforementioned important roles of EZH2, many EZH2 inhibitors, such as DZNeP, GSK126, UNC1999, EPZ-6438, PF-06821497, CPI-1205, CPI-0209 and DS3201, have been developed and tested in preclinical and clinical settings (Park et al., 2021b, Li et al., 2021, Duan et al., 2020). Tazemetostat (EPZ-6438) is the first FDA-approved EZH2 inhibitor for the treatment of epithelioid sarcoma and follicular lymphoma (Hoy, 2020). In the domain of mCRPC, there are a couple of ongoing clinical trials, either with EZH2 inhibitors alone or together with antiandrogen treatment, PARP inhibitor or immune checkpoint blockade (ICB) therapy (NCT03460977, NCT04846478, NCT04179864, NCT03480646, NCT04104776, NCT04388852; Table 1). Despite much effort, there seems a lack of efficacy of EZH2 inhibitors in solid tumors, which could be explained, at least partially, by the enzymatically independent functions of EZH2.
Interestingly, a gene-activation-related function EZH2 has been linked to an intrinsic transactivation activity harbored within a cryptic transactivation domain (TAD) of EZH2 (Jiao et al., 2020, Wang et al., 2022b). Here, EZH2 TAD was found to directly interact with p300 and c-Myc, which then act to mediate gene activation (Jiao et al., 2020, Wang et al., 2022b). To more thoroughly inhibit multifaceted activities of EZH2 in cancers, a Proteolysis-Targeting Chimera (PROTAC)-based degrader specific to EZH2 was recently developed for blocking both H3K27me3-dependent and H3K27me3-independent tumor-promoting functions of EZH2 (Wang et al., 2022b, Wang et al., 2022a). Wang et al. further reported that, in advanced PCa, EZH2’s TAD associates with both AR and AR-V7 to recruit the transactivation-related machineries at target sites that lack binding of PRC2 and H3K27me3 (Wang et al., 2022a). These non-canonical target sites of EZH2 and AR/AR-V7 were found to be enriched for the clinically relevant oncogenes, notably CDK2 and MYBL2 (also known as B-Myb) (Wang et al., 2022a). EZH2 TAD facilitates EZH2’s recruitment to these non-canonical target oncogenes, stimulating their activation to enhance CRPC growth in vitro and in vivo (Wang et al., 2022a). And when compared to the matched EZH2 catalytic inhibitor, the EZH2 degrader MS177 showed superior antitumor efficacy, presumably through on-target depletion of both EZH2’s canonical (EZH2:PRC2) and non-canonical (EZH2TAD:AR/AR-V7:co-activators) complexes in PCa (Wang et al., 2022a). EZH2-targeting PROTACs emerge as a promising therapeutic method for treating the aggressive cancers (Wang et al., 2022b, Wang et al., 2022a).
H3K27me demethylases include KDM6A (or UTX), KDM6B (or JMJD3), and KDM7A. KDM6A interacts with AR in advanced PCa (Grasso et al., 2012). Although KDM6A was found mutated in metastatic CRPC, its exact function in PCa progression or AR signaling remains elusive. A role for KDM6B was also implicated in PCa (Xiang et al., 2007a, Farzaneh et al., 2022). KDM7A is upregulated in PCa, which is correlated with Gleason score (Lee et al., 2018). KDM7A directly interacts with AR and regulates expression of its downstream target genes (Lee et al., 2018). Knockdown of KDM7A inhibits PCa cell proliferation in vitro and in the tumor xenografted animal model, indicating that targeting KDM7A might be a possible strategy to treat advanced PCa (Lee et al., 2018).
3.4. H3K36 methylation
H3K36 methylation is involved in a range of biological processes including transcriptional regulation, mRNA splicing, DNA replication and DNA repair, and it also cross-talks with other epigenetic modifications, such as DNA methylation and H3K27me3 (Husmann and Gozani, 2019, Li et al., 2019). H3K36me2 is enriched at the 5’ regions of actively transcribed genes and intergenic area, while H3K36me3 is mainly localized to the gene body of actively transcribed genes (Pokholok et al., 2005, Li et al., 2019, Husmann and Gozani, 2019). There exist several H3K36me2-specific writers in the human cells, which include ASH1L and the nuclear receptor binding SET domain (NSD) family proteins, NSD1, NSD2 (also known as WHSC1 and MMSET) and NSD3. In contrast, SETD2 is the sole H3K36me3 writer (Husmann and Gozani, 2019). H3K36 methylation is removed by the KDM2 (KDM2A and KDM2B) and KDM4 (KDM4A-KDM4D) subfamilies of erasers (Greer and Shi, 2012). H3K36 methylation can be recognized by reads that contain the PWWP, Chromodomain, or Tudor domain (Li et al 2019).
Deregulation of NSD family writer is known to be involved in development of human cancers including PCa (Topchu et al., 2022). For example, NSD1 and NSD2 were both shown to interact with AR, enhancing transactivation of AR signaling in PCa (Wang et al., 2001, Kang et al., 2009). The role of NSD2 in PCa has been extensively studied. NSD2 mediates constitutive activation of NF-κB signaling by proinflammatory cytokines in CRPC through direct interaction with NF-κB, which involves NSD2’s enzymatic activity (Yang et al., 2012). NSD2 is overexpressed in PCa and required for PCa tumor growth (Yang et al., 2012). In addition, NSD2 has been found to be overexpressed in metastatic PCa compared to primary tumors and associated with disease stage and biochemical recurrence (Li et al., 2017b). NSD2, together with PTEN loss, promotes PCa metastasis (Li et al., 2017b). Aytes et al. further showed NSD2 to be a conserved driver of metastatic PCa through analysis of genetically engineered mouse models (GEMM) of PCa and correlative study of human PCa patient datasets (Aytes et al., 2018). Interestingly, H3K36me2 has recently been associated with epithelial plasticity and metastasis in pancreatic ductal adenocarcinoma (PDAC) through the H3K36 to H3M36 mutation (H3K36M), which directly and globally inhibits this mark (Yuan et al., 2020b). via CRISPR-Cas9 screening, the authors discovered both NSD2 and KDM2A (the writer and eraser of H3K36me2) to be involved in regulating EMT and tumor metastasis of PDAC (Yuan et al., 2020b). NSD2 also cross-talks with other epigenetic players. For instance, EZH2 regulates the expression of NSD2 through repressing the NSD2-targeting microRNAs (Asangani et al., 2013). On the other hand, the oncogenic function of EZH2 was mediated by NSD2, which affects PCa tumor formation and metastasis (Asangani et al., 2013). NSD2 has also been shown to be involved in regulating immune infiltration in PCa (Want et al., 2021). NSD3 is amplified in lung squamous cell carcinoma (LUSC) and functions as a key regulator of tumorigenesis (Yuan et al., 2021). In breast cancer, NSD3-mediated H3K36 methylation is crucial for tumor initiation and metastasis (Jeong et al., 2021). However, the function of NSD3 in PCa remains to be identified.
SETD2 serves as the sole methyltransferase for writing H3K36me3 (Edmunds et al., 2008) and is frequently mutated in a variety of human cancers (Li et al., 2019). Via H3K36me3 deposition, SETD2 has been shown to be involved in regulation of DNA damage repair and mRNA splicing (Luco et al., 2010). Additionally, SETD2 also methylates non-histone substrates to regulate oncogenesis. For example, Yuan et al. reported that loss of SETD2 cooperates with PTEN deletion to promote PCa metastasis in an EZH2-dependent manner (Yuan et al., 2020a). Mechanistically, SETD2 methylates EZH2 at its K735 residue, an event that promotes degradation of EZH2, an oncoprotein in PCa, and thus inhibits progression of PCa (Yuan et al., 2020a). The K735R mutant of EZH2 (a SETD2 methylation deficient mutant) induces metastatic PCa in mice (Yuan et al., 2020a). These results thus provide an explanation for the known antagonism between SETD2-mediated H3K36me3 and EZH2-catalyzed H3K27me3 (Yuan et al., 2011, Schmitges et al., 2011, Yuan et al., 2020a).
PHF19, a component of PRC2 complex, promotes the binding and spreading of PRC2 complex into active chromatin region through its Tudor domain-mediated recognition of H3K36me3, thereby promoting gene silencing during development (Li et al., 2017a, Brien et al., 2012, Ballare et al., 2012, Cai et al., 2013). PHF19 also plays a role in many cancers including PCa (Jain et al., 2020, Abdelfettah et al., 2020). In vitro studies with the AR-negative PCa indicated that PHF19 regulates cell proliferation, invasiveness and angiogenesis (Jain et al., 2020). Another H3K36 methylation reader, LEDGF (or PSIP1), is overexpressed in PCa and knocking down LEDGF sensitizes chemo-resistant PCa cells to taxanes (Rios-Colon et al., 2017). The exact functions of these H3K36 methylation readers in PCa tumorigenesis and progression warrant more detailed study in future.
The H3K36 methylation erasers include the family members of KDM2 and KDM4. KDM2B has recently been shown to play a role in PCa cell motility (Zacharopoulou et al., 2020). The four KDM4 family members (namely, KDM4A, KDM4B, KDM4C and KDM4D) can act as the erasers of both H3K36me2/3 and H3K9me2/3 methylation (Klose et al., 2006, Whetstine et al., 2006), as mentioned in the previous H3K9 methylation section. KDM4 proteins can form complex with AR and promote its activity in an enzymatic dependent fashion (Coffey et al., 2013, Chu et al., 2014, Wissmann et al., 2007, Shin and Janknecht, 2007).
3.5. H3K79 methylation
H3K79 methylation, located inside the globular domain of histone H3, is associated with the actively transcribed genes (Zhao et al., 2021). Disrupter of telomeric silencing 1L (DOT1L) is the only identified writer for H3K79 methylation (Feng et al., 2002). So far, eraser of H3K79 methylation has not been reported. Very recently, Menin is identified as the first H3K79me2 reader protein (Lin et al., 2023). Menin is a well-known component of MLL complex, the H3K4 methylation writer, and also a critical regulator of AR signaling (Malik et al., 2015). The expression of Menin is higher in CRPC when compared to primary PCa and benign prostate tissues, which also correlates with poor overall survival of PCa patients (Malik et al., 2015). How exactly such a newly identified function of Menin as a H3K79me2 reader is involved in PCa development warrants further investigation. H3K79me2 and H3K79me3 marks participate in the regulation of gene transcription, cell cycle progression and DNA damage response (Nguyen and Zhang, 2011). Besides its function in the development and maintenance of MLL-rearranged (MLL-r) leukemia (Okada et al., 2005), DOT1L has also become a promising therapeutic target for treating the solid tumors (Alexandrova et al., 2022). DOT1L was identified as a novel cofactor for ERα and regulates transcription of ERα target genes in breast cancer (Nassa et al., 2019). DOT1L inhibition impairs the growth of antiestrogen-resistant breast cancer cells (Nassa et al., 2019). Vatapalli et al. found that DOT1L expression was significantly increased in PCa relative to normal prostate and correlated with Gleason score and poor clinical outcome in multiple datasets of PCa patients (Vatapalli et al., 2020). Interestingly, DOT1L blockade specifically reduces the viability of AR-positive PCa cells, including enzalutamide-resistant ones (Vatapalli et al., 2020). Mechanistically, DOT1L and AR co-bind a distal enhancer of MYC, thus promoting the MYC expression in AR-positive PCa cells; conversely, treatment with the DOT1L inhibitor promotes AR and MYC degradation via upregulation of the E3 ubiquitin ligases HECTD4 and MYCBP2, thus impairs the PCa tumor growth (Vatapalli et al., 2020). Taken together, these studies provide a foundation for rationalizing the targeting of DOT1L in endocrine therapy resistant AR-positive or ERα-positive cancers.
4. Histone acetylation in prostate cancer
Histone acetylation is closely associated with transcriptional activation and this histone PTM is achieved through the transfer of an acetyl group to the histone lysine by HATs (Struhl, 1998). Conversely, HDACs conduct deacetylation reaction by the removal of an acetyl group off lysine (Struhl, 1998). There are over 40 different histone lysine residues that have been reported to be modified by acetylation (Zhao and Garcia, 2015). Histone acetylation influences numerous physiological and pathogenic processes (Shvedunova and Akhtar, 2022). For instance, HATs and HDACs act as AR’s coactivator and corepressor, respectively, thus profoundly affecting the AR-mediated gene transcription in PCa (Lavery and Bevan, 2011). H3K27ac is a marker for active enhancers and promoters. Patterns of global AR binding and H3K27ac are reprogrammed in mCRPC compared to localized PCa (Pomerantz et al., 2020), suggesting the importance of chromatin states in governing tumor progression. In addition, H3K27ac profiling before and after AR-targeted therapy revealed that a subset of H3K27ac peaks are associated with treatment resistance (Severson et al., 2023).
Super-enhancers are defined as a cluster of enhancers formed by exceedingly high levels of binding of master transcription factors and mediator complex, also marked with strong histone acetylation such as H3K27ac (Whyte et al., 2013), which play a crucial role as oncogenic drivers in various tumor types including PCa. Activation of AR-associated enhancers is well correlated with histone acetylation in PCa (Valdes-Mora et al., 2017).
4.1. Histone acetyltransferases (HATs)
Classic HATs include p300, CREB-binding protein (CBP), TIP60, GCN5 and PCAF. These HATs are involved in AR signaling and PCa tumorigenesis, which are nicely summarized in a recent review (Jaiswal et al., 2022). Here, we focus on p300 and CBP, the two highly homologous HAT proteins, each containing two ZZ-type zinc finger (ZZ) domains and three cysteine/histidine-rich (CH) domains (CH1, CH2 and CH3), a bromodomain (BD) that recognizes acetylated residues, and a catalytic histone acetyltransferase (HAT) domain that catalyzes lysine acetylation (Dancy and Cole, 2015). p300 and CBP are well-known AR coactivators through directly acetylating AR to enhance AR’s transcriptional activity (Fu et al., 2000). They are overexpressed and involved in the progression of PCa (Comuzzi et al., 2004, Debes et al., 2003). A number of CBP/p300 inhibitors have recently been developed. For example, GNE-049, a CBP/p300 bromodomain inhibitor, inhibits the growth of CRPC in vitro and in vivo (Jin et al., 2017). A-485, a highly selective catalytic inhibitor of p300/CBP, potently downregulated the AR transcriptional output in both androgen-sensitive PCa and CRPC cells (Lasko et al., 2017). Furthermore, A-485 inhibited the growth of LUCaP-77 xenograft, a patient-derived AR-positive CRPC model (Lasko et al., 2017). In addition, A-485 treatment greatly increases the efficacy of PD-L1 blockade in a syngeneic PCa mouse model through inhibiting the secretion of exosomal PD-L1 (Liu et al., 2020). More recently, CCS1477, another CBP/p300 inhibitor targeting the bromodomain, exerts anti-tumor activity through regulating AR, AR-V7 and c-MYC nodes and it is currently in a phase I/II clinical trial NCT03568656 (Table 1) (Welti et al., 2021). In summary, these findings underscore CBP/p300 as a compelling therapeutic target, either alone or in combination with other drugs, for the treatment of advanced PCa.
4.2. Histone deacetylases (HDACs)
HDACs removes acetylation of histones and non-histone substrates (Shvedunova and Akhtar, 2022). So far, eighteen different HDACs have been identified, which comprise four major classes, namely, HDAC Class I, II, III and IV (Seto and Yoshida, 2014). Overexpression of HDACs is observed in different types of cancers, including PCa (Ropero and Esteller, 2007).
Class I HDACs include HDAC1, 2, 3 and 8, which are highly expressed in PCa compared to normal prostate tissue (Weichert et al., 2008). HDAC inhibitors block AR-mediated activation of target genes in both hormone-sensitive and refractory PCa (Welsbie et al., 2009). Similarly, HDAC3 inhibitors were shown to block the activity of AR-V7, inhibiting the growth of 22Rv1 xenografted tumors (McLeod et al., 2018). A very recent study further demonstrated that selective elimination of senescent neutrophils through HDAC inhibitors delays PCa progression in vivo (Bancaro et al., 2023). Class III HDACs, namely sirtuins (SIRT1–7), are nicotinamide adenine dinucleotide (NAD+)-dependent and different from other zinc-dependent HDAC classes (Chalkiadaki and Guarente, 2015). The function of sirtuins in PCa is complex and context dependent. For example, loss of SIRT1 results PIN formation (Powell et al., 2011); and SIRT1 inhibits PCa cell proliferation through AR deacetylation (Lavery and Bevan, 2011, Fu et al., 2006). In contrast, Huang et al. showed that lowering SIRT1 leads to inhibition of PCa growth (Huang et al., 2021).
HDAC inhibitors, which cover five different classes of compounds (namely, hydroxamic acids, aliphatic acids, cyclic peptides, benzamides and sirtuin inhibitor), have been developed and tested in clinical trials (Eckschlager et al., 2017). Suberoylanilide hydroxamic acids (SAHA) was the first FDA-approved HDAC inhibitor for the treatment of cancer in the US (Grant et al., 2007). SAHA treatment has been found to inhibit the proliferation of PCa cell lines in vitro and the progression of PCa tumors in vivo (Butler et al., 2000, Chinnaiyan et al., 2005, Lakshmikanthan et al., 2006). Although HDAC inhibitors showed efficacy for some hematological malignancy subtypes, a majority of the clinical trials involving HDAC inhibitors in PCa failed due to high toxicity and a lack of specificity (Rana et al., 2020). There are still ongoing clinical trials with HDAC inhibitor in PCa (Thompson et al., 2022). For example, the FDA-approved HDAC inhibitor Belinostat is currently tested together with PARP inhibitor Talazoparib in mCRPC (NCT04703920, Table 1). Nevertheless, different HDACs can exert either tumor-promoting or anti-tumorigenic functions (Chalkiadaki and Guarente, 2015), therefore demanding more mechanistic studies to provide guidance on how to employ class-specific HDAC inhibitors, either as a single-agent therapy or in combination with other therapies, to treat advanced PCa.
4.3. Histone acetylation readers
The bromodomain-containing family proteins recognize histone acetylation to regulate gene expression (Fujisawa and Filippakopoulos, 2017). The bromodomain and extra terminal domain (BET) subfamily of bromodomain-containing proteins include BRD2, BRD3, BRD4, and BRDT, all of which contain two tandem bromodomains (BD1 and BD2) (Perez-Salvia and Esteller, 2017, Stathis and Bertoni, 2018). BD1 and BD2 play an important role in regulating transcription via recognizing acetylated lysine residues of histones and non-histone proteins (Dhalluin et al., 1999). BRD4, the best characterized BET family protein, recruits the elongation factor p-TEFb to stimulate RNA polymerase II-dependent transcription (Jang et al., 2005). BET proteins are frequently overexpressed in various types of human cancer including PCa (Urbanucci et al., 2017, Urbanucci and Mills, 2018, Fujisawa and Filippakopoulos, 2017, Stathis and Bertoni, 2018). They play a critical role in tumorigenesis and represent a class of attractive therapeutic targets for cancer treatment (Perez-Salvia and Esteller, 2017).
Pan-BET inhibitors, such as JQ1, I-BET151, I-BET762, ABBV-075, OTX-015 and ZEN-3694, were developed to block interaction between BD domain and the acetylated residue, which were shown to affect a large variety of cellular processes and have antitumor effects in numerous preclinical and clinical models (Perez-Salvia and Esteller, 2017). In PCa, BRD4 physically interacts with the N-terminal domain of AR and promotes AR-related gene-expression programs in CRPC (Asangani et al., 2014). JQ1, an inhibitor of BET family member proteins, inhibited the binding of BRD4 to AR enhancers globally, thereby repressing the AR-mediated gene transcription (Asangani et al., 2016). Welti et al. reported that the nuclear BRD4 level increases significantly over disease progression and its higher expression at diagnosis is associated with a worse clinical outcome (Welti et al., 2018). Furthermore, they found that BET inhibitors reduce AR and AR-V7 signaling in patient-derived xenograft model of CRPC (Welti et al., 2018). Furthermore, BET inhibitors also overcome antiandrogen resistance in advanced PCa (Shah et al., 2017, Asangani et al., 2016). In addition, BRD4 seems to have AR-independent functions (Civenni et al., 2019, Coleman et al., 2019). BRD4 also recognizes the acetylated non-histone proteins such as NF-κB and TMPRSS2-ERG (Hajmirza et al., 2018, Li et al., 2018). Treatment of the BET inhibitor OTX-015 increases the efficacy of radiation therapy (RT) and overcomes radio-resistance through blocking DNA repair (Li et al., 2022b). In summary, these aforementioned studies lay a strong foundation for the future clinical exploration of BET inhibitors.
The mechanisms for resistance to BET inhibitors have been reported, some of which involve PCa-associated somatic mutation of SPOP, an E3 ligase substrate binding protein (Zhang et al., 2017, Janouskova et al., 2017, Dai et al., 2017). Due to its frequent mutation in PCa, mutant SPOP failed to induce the ubiquitination and proteasomal degradation of BET domain proteins (Zhang et al., 2017, Janouskova et al., 2017, Dai et al., 2017). This resistance mechanism then led to activation of AKT-mTORC1 signaling, which can be overcome by combination treatment with AKT inhibitors (Zhang et al., 2017).
PROTAC-based small-molecule degrader for specifically targeting BET domain proteins have also emerged (Raina et al., 2016, Zhou et al., 2018). For Instance, ARV-771, a potent pan-BET degrader, more potently suppressed the AR signaling and tumor progression in the CRPC-xenografted mouse models, compared to the original inhibitor (Raina et al., 2016). ZBC-260, another BET degrader, preferentially affects AR-positive PCa cells over AR-negative ones, and suppresses PCa growth in vitro and in vivo (Kregel et al., 2019).
Interestingly, a study based on CRISPR-Cas9-directed protein domain scanning has previously uncovered that the BD1 and BD2 domains of BRD4 have distinct functions (Shi et al., 2015). Accordingly, selective inhibitors targeting either BD1 or BD2 have recently been developed (Gilan et al., 2020, Faivre et al., 2020). While the BD1-specific inhibitor iBET-BD1 phenocopies the pan-BET inhibitors in a majority of the tested cancer cell lines, the BD2-specific inhibitor iBET-BD2 is predominantly effective in models of inflammatory and autoimmune disease (Gilan et al., 2020). The activity of another BD2-specific inhibitor, ABBV-744, is predominantly restricted to AML and AR-positive PCa (Faivre et al., 2020). ABBV-744 displaces BRD4 from AR-containing super-enhancers, inhibits the AR-dependent transcription, and reduces tumor growth in PCa xenografts (Faivre et al., 2020). There are a plethora of ongoing clinical trials with inhibitors or degraders targeting BET domain proteins (Guo et al., 2023). In mCRPC, a number of trials with the pan-BET inhibitor ZEN-3694 are currently underway (Table 1). A completed trial (NCT02711956) showed that ZEN-3694 plus Enzalutamide demonstrated acceptable tolerability and potential efficacy in mCRPC patients, and further prospective study is required (Aggarwal et al., 2020). Interestingly, a phase I/II clinical trial with NUV-868, a BD2-selective oral small-molecule BET inhibitor, is ongoing in mCRPC patients, either alone or in combination with Enzalutamide or Talazoparib (NCT05252390, Table 1).
5. Chromatin remodeling in prostate cancer
Chromatin accessibility is regulated by chromatin-remodeling complexes, which use the energy from ATP hydrolysis to slide or eject nucleosomes (Clapier et al., 2017). There are four subfamilies of chromatin-remodeling complexes, namely Chromodomain Helicase DNA-binding (CHD), Inositol Requiring 80 (INO80), Imitation Switch (ISWI) and Switch/Sucrose Non-Fermentable (SWI/SNF, also known as BRG1/BRM associated factor (BAF) complex) (Clapier et al., 2017). In this section, we mainly highlight the recent discoveries on SWI/SNF and CHD remodeling complexes in PCa.
Alteration of the SWI/SNF complex subunit occurs in about 20–25% of all cancers including PCa (Shain and Pollack, 2013, Kadoch et al., 2013). The catalytic ATPase components, SMARCA4 (BRG1) and SMARCA2 (BRM), mediate ATP hydrolysis to reposition nucleosomes at non-coding regulatory elements, thereby enabling the transcription factors to bind DNA and promote gene expression (Mittal and Roberts, 2020). The mutant SWI/SNF complex usually enhances oncogenic transcriptional programs, making it a promising therapeutic target (Centore et al., 2020). BRG1 has been reported to be overexpressed in PCa and associated with tumor progression (Cyrta et al., 2020, Muthuswami et al., 2019, Sun et al., 2007). Sandoval et al. identified a strong interaction between TMPRSS2-ERG and BAF complex in VCaP cells, a PCa cell line containing endogenous TMPRSS2-ERG fusion (Sandoval et al., 2018). They further found that this interaction drives retargeting of BAF complex globally to ETS target sites; meanwhile, the ATP-dependent catalytic activity of BAF complex is required for ERG’s chromatin association, ERG’s downstream target gene expression and ERG’s oncogenic function in PCa (Sandoval et al., 2018). The interdependency between ERG and BAF complex indicates that TMPRSS2-ERG-positive PCa could be sensitive to BAF complex inhibitors. Indeed, Xiao et al. reported that the treatment of AU-15330, a PROTAC degrader of BRG1 and BRM, dismissed the main transcription factors such as AR, FOXA1, ERG and MYC from chromatin, disrupted their super-enhancer and promoter looping, inhibited the down-stream oncogenic pathways, and induced strong inhibition of CRPC growth, either alone or in combination with enzalutamide (Xiao et al., 2022). BRD9, a component of non-canonical SWI/SNF complex (ncBAF), has recently been shown to interact with AR and regulate its activity and promote tumor progression (Alpsoy et al., 2021). In addition, the expression of BAF57 is positively correlated with Gleason score and functions as a critical regulator of AR (Link et al., 2008, Balasubramaniam et al., 2013).
Chromatin-remodeling complex often intertwines with other signaling pathways. For example, a synthetic lethal relationship was characterized between PTEN and BRG1 by the Qin laboratory (Ding et al., 2019). In this study, they found that PTEN loss stabilized BRG1 via inhibiting the AKT/GSK3b/FBXW7 pathway, which drove a tumor-promoting transcriptome through chromatin remodeling (Ding et al., 2019). Additionally, PTEN-deficient PCa is sensitive to the treatment with BRG1 antagonist, indicating a therapeutic strategy (Ding et al., 2019).
Another chromatin remodeler CHD1 is altered in 7–10% of PCa cases (Baca et al., 2013, Grasso et al., 2012). CHD1 has been reported to play significant roles in PCa in a context-dependent manner (Li et al., 2023, Zhao et al., 2017). CHD1 depletion leads to the defects of DNA double-strand break (DSB) repair via homologous recombination (HR) and thus sensitizes PCa cells to PARP inhibitors (Kari et al., 2016). Coordinate loss of MAP3K7 and CHD1 occurs in 10% to 20% of localized PCa, correlating with poor disease-free survival (Rodrigues et al., 2015). Likewise, combined loss of MAP3K7 and CHD1 increased tumor growth and decreased survival in LNCaP xenografted animal models (Rodrigues et al., 2015). In addition, CHD1 loss alters AR binding and regulates different sets of target genes to promote PCa (Augello et al., 2019). Zhang et al. identified that depletion of CHD1 leads to lineage plasticity and antiandrogen resistance through an in vivo shRNA screening (Zhang et al., 2020). In a mCRPC patient cohort, low CHD1 expression is correlated with a worse response to the second-generation antiandrogen treatment (Zhang et al., 2020).
Interestingly, both SWI/SNF complex and CHD1 have been shown to play an important role in cancer immunotherapy including PCa (Li et al., 2022a, Belk et al., 2022). Since PCa is known to be immune-cold (Topalian et al., 2012), it would be worthwhile to test whether targeting chromatin remodeling pathway makes PCa sensitive to immunotherapy.
6. Conclusions and future perspectives
This chapter provides an overview of epigenetic alterations, focusing on DNA methylation, histone methylation and acetylation, and chromatin remodeling during the development and progression of PCa. AR-mediated signaling is one of the most critical drivers for PCa initiation and progression. Various histone-modifying enzymes regulate ligand-dependent or ligand-independent transcription of AR target genes through interacting with AR. Additionally, epigenetic regulators are also critically involved in the activation of other oncogenic signaling pathways, independent of AR signaling and usually in a more advanced setting of PCa such as mCRPC. Due to the reversible nature of epigenetic modifications, targeting epigenetic regulators has become a promising therapeutic intervention. Table I lists some of the ongoing clinical trials for testing drug candidates targeting epigenetic factors in patients with mCRPC. Development of epigenetically centered therapy alone generally showed less efficacy in PCa, probably because of the heterogeneity nature of this disease. However, we remain optimistic about potential combination therapies with other drugs, such as antiandrogen, PARP inhibitor and immune therapy, for the treatment of patients with advanced PCa. We look forward to new exciting development along these lines in the years to come.
Acknowledgements
We thank all members of the Cai laboratory for helpful discussion. This work was supported in part by the US National Institutes of Health (NIH) grant (R01CA262903 to L.C.). We apologize to colleagues whose studies cannot be cited in this book chapter due to space limitations. The figure in this chapter was created using tools from BioRender.com.
Footnotes
Competing interest statement
The authors declare no competing interests.
References
- ABDELFETTAH S, BOULAY G, DUBUISSEZ M, SPRUYT N, GARCIA SP, RENGARAJAN S, LOISON I, LEROY X, RIVERA MN & LEPRINCE D 2020. hPCL3S promotes proliferation and migration of androgen-independent prostate cancer cells. Oncotarget, 11, 1051–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ABIDA W, CYRTA J, HELLER G, PRANDI D, ARMENIA J, COLEMAN I, CIESLIK M, BENELLI M, ROBINSON D, VAN ALLEN EM, SBONER A, FEDRIZZI T, MOSQUERA JM, ROBINSON BD, DE SARKAR N, KUNJU LP, TOMLINS S, WU YM, NAVA RODRIGUES D, LODA M, GOPALAN A, REUTER VE, PRITCHARD CC, MATEO J, BIANCHINI D, MIRANDA S, CARREIRA S, RESCIGNO P, FILIPENKO J, VINSON J, MONTGOMERY RB, BELTRAN H, HEATH EI, SCHER HI, KANTOFF PW, TAPLIN ME, SCHULTZ N, DEBONO JS, DEMICHELIS F, NELSON PS, RUBIN MA, CHINNAIYAN AM & SAWYERS CL 2019. Genomic correlates of clinical outcome in advanced prostate cancer. Proc Natl Acad Sci U S A, 116, 11428–11436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- AGGARWAL RR, SCHWEIZER MT, NANUS DM, PANTUCK AJ, HEATH EI, CAMPEAU E, ATTWELL S, NOREK K, SNYDER M, BAUMAN L, LAKHOTIA S, FENG FY, SMALL EJ, ABIDA W & ALUMKAL JJ 2020. A Phase Ib/IIa Study of the Pan-BET Inhibitor ZEN-3694 in Combination with Enzalutamide in Patients with Metastatic Castration-resistant Prostate Cancer. Clin Cancer Res, 26, 5338–5347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ALEXANDROVA E, SALVATI A, PECORARO G, LAMBERTI J, MELONE V, SELLITTO A, RIZZO F, GIURATO G, TARALLO R, NASSA G & WEISZ A 2022. Histone Methyltransferase DOT1L as a Promising Epigenetic Target for Treatment of Solid Tumors. Front Genet, 13, 864612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ALLIS CD & JENUWEIN T 2016. The molecular hallmarks of epigenetic control. Nat Rev Genet, 17, 487–500. [DOI] [PubMed] [Google Scholar]
- ALPSOY A, UTTURKAR SM, CARTER BC, DHIMAN A, TORREGROSA-ALLEN SE, CURRIE MP, ELZEY BD & DYKHUIZEN EC 2021. BRD9 Is a Critical Regulator of Androgen Receptor Signaling and Prostate Cancer Progression. Cancer Res, 81, 820–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ARDEHALI MB, ANSELMO A, COCHRANE JC, KUNDU S, SADREYEV RI & KINGSTON RE 2017. Polycomb Repressive Complex 2 Methylates Elongin A to Regulate Transcription. Mol Cell, 68, 872–884 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ARMENIA J, WANKOWICZ SAM, LIU D, GAO J, KUNDRA R, REZNIK E, CHATILA WK, CHAKRAVARTY D, HAN GC, COLEMAN I, MONTGOMERY B, PRITCHARD C, MORRISSEY C, BARBIERI CE, BELTRAN H, SBONER A, ZAFEIRIOU Z, MIRANDA S, BIELSKI CM, PENSON AV, TOLONEN C, HUANG FW, ROBINSON D, WU YM, LONIGRO R, GARRAWAY LA, DEMICHELIS F, KANTOFF PW, TAPLIN ME, ABIDA W, TAYLOR BS, SCHER HI, NELSON PS, DE BONO JS, RUBIN MA, SAWYERS CL, CHINNAIYAN AM, TEAM PSCIPCD, SCHULTZ N & VAN ALLEN EM 2018. The long tail of oncogenic drivers in prostate cancer. Nat Genet, 50, 645–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ASANGANI IA, ATEEQ B, CAO Q, DODSON L, PANDHI M, KUNJU LP, MEHRA R, LONIGRO RJ, SIDDIQUI J, PALANISAMY N, WU YM, CAO X, KIM JH, ZHAO M, QIN ZS, IYER MK, MAHER CA, KUMAR-SINHA C, VARAMBALLY S & CHINNAIYAN AM 2013. Characterization of the EZH2-MMSET histone methyltransferase regulatory axis in cancer. Mol Cell, 49, 80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ASANGANI IA, DOMMETI VL, WANG X, MALIK R, CIESLIK M, YANG R, ESCARA-WILKE J, WILDER-ROMANS K, DHANIREDDY S, ENGELKE C, IYER MK, JING X, WU YM, CAO X, QIN ZS, WANG S, FENG FY & CHINNAIYAN AM 2014. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature, 510, 278–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ASANGANI IA, WILDER-ROMANS K, DOMMETI VL, KRISHNAMURTHY PM, APEL IJ, ESCARA-WILKE J, PLYMATE SR, NAVONE NM, WANG S, FENG FY & CHINNAIYAN AM 2016. BET Bromodomain Inhibitors Enhance Efficacy and Disrupt Resistance to AR Antagonists in the Treatment of Prostate Cancer. Mol Cancer Res, 14, 324–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ASKEW EB, BAI S, PARRIS AB, MINGES JT & WILSON EM 2017. Androgen receptor regulation by histone methyltransferase Suppressor of variegation 3–9 homolog 2 and Melanoma antigen-A11. Mol Cell Endocrinol, 443, 42–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- AUGELLO MA, LIU D, DEONARINE LD, ROBINSON BD, HUANG D, STELLOO S, BLATTNER M, DOANE AS, WONG EWP, CHEN Y, RUBIN MA, BELTRAN H, ELEMENTO O, BERGMAN AM, ZWART W, SBONER A, DEPHOURE N & BARBIERI CE 2019. CHD1 Loss Alters AR Binding at Lineage-Specific Enhancers and Modulates Distinct Transcriptional Programs to Drive Prostate Tumorigenesis. Cancer Cell, 35, 603–617 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- AYTES A, GIACOBBE A, MITROFANOVA A, RUGGERO K, CYRTA J, ARRIAGA J, PALOMERO L, FARRAN-MATAS S, RUBIN MA, SHEN MM, CALIFANO A & ABATE-SHEN C 2018. NSD2 is a conserved driver of metastatic prostate cancer progression. Nat Commun, 9, 5201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BACA SC, PRANDI D, LAWRENCE MS, MOSQUERA JM, ROMANEL A, DRIER Y, PARK K, KITABAYASHI N, MACDONALD TY, GHANDI M, VAN ALLEN E, KRYUKOV GV, SBONER A, THEURILLAT JP, SOONG TD, NICKERSON E, AUCLAIR D, TEWARI A, BELTRAN H, ONOFRIO RC, BOYSEN G, GUIDUCCI C, BARBIERI CE, CIBULSKIS K, SIVACHENKO A, CARTER SL, SAKSENA G, VOET D, RAMOS AH, WINCKLER W, CIPICCHIO M, ARDLIE K, KANTOFF PW, BERGER MF, GABRIEL SB, GOLUB TR, MEYERSON M, LANDER ES, ELEMENTO O, GETZ G, DEMICHELIS F, RUBIN MA & GARRAWAY LA 2013. Punctuated evolution of prostate cancer genomes. Cell, 153, 666–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BAI Y, ZHANG Z, CHENG L, WANG R, CHEN X, KONG Y, FENG F, AHMAD N, LI L & LIU X 2019. Inhibition of enhancer of zeste homolog 2 (EZH2) overcomes enzalutamide resistance in castration-resistant prostate cancer. J Biol Chem, 294, 9911–9923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BALASUBRAMANIAM S, COMSTOCK CE, ERTEL A, JEONG KW, STALLCUP MR, ADDYA S, MCCUE PA, OSTRANDER WF JR., AUGELLO MA & KNUDSEN KE 2013. Aberrant BAF57 signaling facilitates prometastatic phenotypes. Clin Cancer Res, 19, 2657–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BALLARE C, LANGE M, LAPINAITE A, MARTIN GM, MOREY L, PASCUAL G, LIEFKE R, SIMON B, SHI Y, GOZANI O, CARLOMAGNO T, BENITAH SA & DI CROCE L 2012. Phf19 links methylated Lys36 of histone H3 to regulation of Polycomb activity. Nat Struct Mol Biol, 19, 1257–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BANCARO N, CALI B, TROIANI M, ELIA AR, ARZOLA RA, ATTANASIO G, LAI P, CRESPO M, GUREL B, PEREIRA R, GUO C, MOSOLE S, BRINA D, D’AMBROSIO M, PASQUINI E, SPATARO C, ZAGATO E, RINALDI A, PEDOTTI M, DI LASCIO S, MEANI F, MONTOPOLI M, FERRARI M, GALLINA A, VARANI L, PEREIRA MESTRE R, BOLIS M, GILLESSEN SOMMER S, DE BONO J, CALCINOTTO A & ALIMONTI A 2023. Apolipoprotein E induces pathogenic senescent-like myeloid cells in prostate cancer. Cancer Cell, 41, 602–619 e11. [DOI] [PubMed] [Google Scholar]
- BARATCHIAN M, TIWARI R, KHALIGHI S, CHAKRAVARTHY A, YUAN W, BERK M, LI J, GUERINOT A, DE BONO J, MAKAROV V, CHAN TA, SILVERMAN RH, STARK GR, VARADAN V, DE CARVALHO DD, CHAKRABORTY AA & SHARIFI N 2022. H3K9 methylation drives resistance to androgen receptor-antagonist therapy in prostate cancer. Proc Natl Acad Sci U S A, 119, e2114324119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BARSKI A, CUDDAPAH S, CUI K, ROH TY, SCHONES DE, WANG Z, WEI G, CHEPELEV I & ZHAO K 2007. High-resolution profiling of histone methylations in the human genome. Cell, 129, 823–37. [DOI] [PubMed] [Google Scholar]
- BEDFORD MT & VAN HELDEN PD 1987. Hypomethylation of DNA in pathological conditions of the human prostate. Cancer Res, 47, 5274–6. [PubMed] [Google Scholar]
- BELK JA, YAO W, LY N, FREITAS KA, CHEN YT, SHI Q, VALENCIA AM, SHIFRUT E, KALE N, YOST KE, DUFFY CV, DANIEL B, HWEE MA, MIAO Z, ASHWORTH A, MACKALL CL, MARSON A, CARNEVALE J, VARDHANA SA & SATPATHY AT 2022. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell, 40, 768–786 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BERGER A, BRADY NJ, BAREJA R, ROBINSON B, CONTEDUCA V, AUGELLO MA, PUCA L, AHMED A, DARDENNE E, LU X, HWANG I, BAGADION AM, SBONER A, ELEMENTO O, PAIK J, YU J, BARBIERI CE, DEPHOURE N, BELTRAN H & RICKMAN DS 2019. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest, 129, 3924–3940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BHASIN JM, LEE BH, MATKIN L, TAYLOR MG, HU B, XU Y, MAGI-GALLUZZI C, KLEIN EA & TING AH 2015. Methylome-wide Sequencing Detects DNA Hypermethylation Distinguishing Indolent from Aggressive Prostate Cancer. Cell Rep, 13, 2135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BJORKMAN M, OSTLING P, HARMA V, VIRTANEN J, MPINDI JP, RANTALA J, MIRTTI T, VESTERINEN T, LUNDIN M, SANKILA A, RANNIKKO A, KAIVANTO E, KOHONEN P, KALLIONIEMI O & NEES M 2012. Systematic knockdown of epigenetic enzymes identifies a novel histone demethylase PHF8 overexpressed in prostate cancer with an impact on cell proliferation, migration and invasion. Oncogene, 31, 3444–56. [DOI] [PubMed] [Google Scholar]
- BLACKLEDGE NP & KLOSE RJ 2021. The molecular principles of gene regulation by Polycomb repressive complexes. Nat Rev Mol Cell Biol, 22, 815–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BOLIS M, BOSSI D, VALLERGA A, CESERANI V, CAVALLI M, IMPELLIZZIERI D, DI RITO L, ZONI E, MOSOLE S, ELIA AR, RINALDI A, PEREIRA MESTRE R, D’ANTONIO E, FERRARI M, STOFFEL F, JERMINI F, GILLESSEN S, BUBENDORF L, SCHRAML P, CALCINOTTO A, COREY E, MOCH H, SPAHN M, THALMANN G, KRUITHOF-DE JULIO M, RUBIN MA & THEURILLAT JP 2021. Dynamic prostate cancer transcriptome analysis delineates the trajectory to disease progression. Nat Commun, 12, 7033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BRIEN GL, GAMBERO G, O’CONNELL DJ, JERMAN E, TURNER SA, EGAN CM, DUNNE EJ, JURGENS MC, WYNNE K, PIAO L, LOHAN AJ, FERGUSON N, SHI X, SINHA KM, LOFTUS BJ, CAGNEY G & BRACKEN AP 2012. Polycomb PHF19 binds H3K36me3 and recruits PRC2 and demethylase NO66 to embryonic stem cell genes during differentiation. Nat Struct Mol Biol, 19, 1273–81. [DOI] [PubMed] [Google Scholar]
- BUCK-KOEHNTOP BA & DEFOSSEZ PA 2013. On how mammalian transcription factors recognize methylated DNA. Epigenetics, 8, 131–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BUTLER LM, AGUS DB, SCHER HI, HIGGINS B, ROSE A, CORDON-CARDO C, THALER HT, RIFKIND RA, MARKS PA & RICHON VM 2000. Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res, 60, 5165–70. [PubMed] [Google Scholar]
- CAI C, HE HH, CHEN S, COLEMAN I, WANG H, FANG Z, CHEN S, NELSON PS, LIU XS, BROWN M & BALK SP 2011. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell, 20, 457–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CAI C, HE HH, GAO S, CHEN S, YU Z, GAO Y, CHEN S, CHEN MW, ZHANG J, AHMED M, WANG Y, METZGER E, SCHULE R, LIU XS, BROWN M & BALK SP 2014. Lysine-specific demethylase 1 has dual functions as a major regulator of androgen receptor transcriptional activity. Cell Rep, 9, 1618–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CAI L, ROTHBART SB, LU R, XU B, CHEN WY, TRIPATHY A, ROCKOWITZ S, ZHENG D, PATEL DJ, ALLIS CD, STRAHL BD, SONG J & WANG GG 2013. An H3K36 methylation-engaging Tudor motif of polycomb-like proteins mediates PRC2 complex targeting. Mol Cell, 49, 571–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CANCER GENOME ATLAS RESEARCH, N. 2015. The Molecular Taxonomy of Primary Prostate Cancer. Cell, 163, 1011–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CENTORE RC, SANDOVAL GJ, SOARES LMM, KADOCH C & CHAN HM 2020. Mammalian SWI/SNF Chromatin Remodeling Complexes: Emerging Mechanisms and Therapeutic Strategies. Trends Genet, 36, 936–950. [DOI] [PubMed] [Google Scholar]
- CHALKIADAKI A & GUARENTE L 2015. The multifaceted functions of sirtuins in cancer. Nat Rev Cancer, 15, 608–24. [DOI] [PubMed] [Google Scholar]
- CHEN S, PETRICCA J, YE W, GUAN J, ZENG Y, CHENG N, GONG L, SHEN SY, HUA JT, CRUMBAKER M, FRASER M, LIU S, BRATMAN SV, VAN DER KWAST T, PUGH T, JOSHUA AM, DE CARVALHO DD, CHI KN, AWADALLA P, JI G, FENG F, WYATT AW & HE HH 2022. The cell-free DNA methylome captures distinctions between localized and metastatic prostate tumors. Nat Commun, 13, 6467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CHENG Y, HE C, WANG M, MA X, MO F, YANG S, HAN J & WEI X 2019. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther, 4, 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CHIN SP, DICKINSON JL & HOLLOWAY AF 2011. Epigenetic regulation of prostate cancer. Clin Epigenetics, 2, 151–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CHINNAIYAN P, VALLABHANENI G, ARMSTRONG E, HUANG SM & HARARI PM 2005. Modulation of radiation response by histone deacetylase inhibition. Int J Radiat Oncol Biol Phys, 62, 223–9. [DOI] [PubMed] [Google Scholar]
- CHU CH, WANG LY, HSU KC, CHEN CC, CHENG HH, WANG SM, WU CM, CHEN TJ, LI LT, LIU R, HUNG CL, YANG JM, KUNG HJ & WANG WC 2014. KDM4B as a target for prostate cancer: structural analysis and selective inhibition by a novel inhibitor. J Med Chem, 57, 5975–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CI X, HAO J, DONG X, CHOI SY, XUE H, WU R, QU S, GOUT PW, ZHANG F, HAEGERT AM, FAZLI L, CREA F, ONG CJ, ZOUBEIDI A, HE HH, GLEAVE ME, COLLINS CC, LIN D & WANG Y 2018. Heterochromatin Protein 1alpha Mediates Development and Aggressiveness of Neuroendocrine Prostate Cancer. Cancer Res, 78, 2691–2704. [DOI] [PubMed] [Google Scholar]
- CIVENNI G, BOSOTTI R, TIMPANARO A, VAZQUEZ R, MERULLA J, PANDIT S, ROSSI S, ALBINO D, ALLEGRINI S, MITRA A, MAPELLI SN, VIERLING L, GIURDANELLA M, MARCHETTI M, PAGANONI A, RINALDI A, LOSA M, MIRA-CATO E, D’ANTUONO R, MORONE D, REZAI K, D’AMBROSIO G, OUAFIK L, MACKENZIE S, RIVEIRO ME, CVITKOVIC E, CARBONE GM & CATAPANO CV 2019. Epigenetic Control of Mitochondrial Fission Enables Self-Renewal of Stem-like Tumor Cells in Human Prostate Cancer. Cell Metab, 30, 303–318 e6. [DOI] [PubMed] [Google Scholar]
- CLAPIER CR, IWASA J, CAIRNS BR & PETERSON CL 2017. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat Rev Mol Cell Biol, 18, 407–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CLERMONT PL, LIN D, CREA F, WU R, XUE H, WANG Y, THU KL, LAM WL, COLLINS CC, WANG Y & HELGASON CD 2015. Polycomb-mediated silencing in neuroendocrine prostate cancer. Clin Epigenetics, 7, 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- COFFEY K, ROGERSON L, RYAN-MUNDEN C, ALKHARAIF D, STOCKLEY J, HEER R, SAHADEVAN K, O’NEILL D, JONES D, DARBY S, STALLER P, MANTILLA A, GAUGHAN L & ROBSON CN 2013. The lysine demethylase, KDM4B, is a key molecule in androgen receptor signalling and turnover. Nucleic Acids Res, 41, 4433–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- COLEMAN DJ, GAO L, KING CJ, SCHWARTZMAN J, URRUTIA J, SEHRAWAT A, TAYOU J, BALTER A, BURCHARD J, CHIOTTI KE, DERRICK DS, SUN D, XIA Z, HEISER LM & ALUMKAL JJ 2019. BET bromodomain inhibition blocks the function of a critical AR-independent master regulator network in lethal prostate cancer. Oncogene, 38, 5658–5669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- COMUZZI B, NEMES C, SCHMIDT S, JASAREVIC Z, LODDE M, PYCHA A, BARTSCH G, OFFNER F, CULIG Z & HOBISCH A 2004. The androgen receptor co-activator CBP is up-regulated following androgen withdrawal and is highly expressed in advanced prostate cancer. J Pathol, 204, 159–66. [DOI] [PubMed] [Google Scholar]
- CONTEDUCA V, OROMENDIA C, ENG KW, BAREJA R, SIGOUROS M, MOLINA A, FALTAS BM, SBONER A, MOSQUERA JM, ELEMENTO O, NANUS DM, TAGAWA ST, BALLMAN KV & BELTRAN H 2019. Clinical features of neuroendocrine prostate cancer. Eur J Cancer, 121, 7–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CREA F, SUN L, MAI A, CHIANG YT, FARRAR WL, DANESI R & HELGASON CD 2012. The emerging role of histone lysine demethylases in prostate cancer. Mol Cancer, 11, 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CREYGHTON MP, CHENG AW, WELSTEAD GG, KOOISTRA T, CAREY BW, STEINE EJ, HANNA J, LODATO MA, FRAMPTON GM, SHARP PA, BOYER LA, YOUNG RA & JAENISCH R 2010. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A, 107, 21931–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CYRTA J, AUGSPACH A, DE FILIPPO MR, PRANDI D, THIENGER P, BENELLI M, COOLEY V, BAREJA R, WILKES D, CHAE SS, CAVALIERE P, DEPHOURE N, ULDRY AC, LAGACHE SB, ROMA L, COHEN S, JAQUET M, BRANDT LP, ALSHALALFA M, PUCA L, SBONER A, FENG F, WANG S, BELTRAN H, LOTAN T, SPAHN M, KRUITHOF-DE JULIO M, CHEN Y, BALLMAN KV, DEMICHELIS F, PISCUOGLIO S & RUBIN MA 2020. Role of specialized composition of SWI/SNF complexes in prostate cancer lineage plasticity. Nat Commun, 11, 5549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DAI X, GAN W, LI X, WANG S, ZHANG W, HUANG L, LIU S, ZHONG Q, GUO J, ZHANG J, CHEN T, SHIMIZU K, BECA F, BLATTNER M, VASUDEVAN D, BUCKLEY DL, QI J, BUSER L, LIU P, INUZUKA H, BECK AH, WANG L, WILD PJ, GARRAWAY LA, RUBIN MA, BARBIERI CE, WONG KK, MUTHUSWAMY SK, HUANG J, CHEN Y, BRADNER JE & WEI W 2017. Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat Med, 23, 1063–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DANCY BM & COLE PA 2015. Protein lysine acetylation by p300/CBP. Chem Rev, 115, 2419–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DARDENNE E, BELTRAN H, BENELLI M, GAYVERT K, BERGER A, PUCA L, CYRTA J, SBONER A, NOORZAD Z, MACDONALD T, CHEUNG C, YUEN KS, GAO D, CHEN Y, EILERS M, MOSQUERA JM, ROBINSON BD, ELEMENTO O, RUBIN MA, DEMICHELIS F & RICKMAN DS 2016. N-Myc Induces an EZH2-Mediated Transcriptional Program Driving Neuroendocrine Prostate Cancer. Cancer Cell, 30, 563–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DAS PM, RAMACHANDRAN K, VANWERT J, FERDINAND L, GOPISETTY G, REIS IM & SINGAL R 2006. Methylation mediated silencing of TMS1/ASC gene in prostate cancer. Mol Cancer, 5, 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DAVIES A, NOURUZI S, GANGULI D, NAMEKAWA T, THAPER D, LINDER S, KARAOGLANOGLU F, OMUR ME, KIM S, KOBELEV M, KUMAR S, SIVAK O, BOSTOCK C, BISHOP J, HOOGSTRAAT M, TALAL A, STELLOO S, VAN DER POEL H, BERGMAN AM, AHMED M, FAZLI L, HUANG H, TILLEY W, GOODRICH D, FENG FY, GLEAVE M, HE HH, HACH F, ZWART W, BELTRAN H, SELTH L & ZOUBEIDI A 2021. An androgen receptor switch underlies lineage infidelity in treatment-resistant prostate cancer. Nat Cell Biol, 23, 1023–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DEBES JD, SEBO TJ, LOHSE CM, MURPHY LM, HAUGEN DA & TINDALL DJ 2003. p300 in prostate cancer proliferation and progression. Cancer Res, 63, 7638–40. [PubMed] [Google Scholar]
- DHALLUIN C, CARLSON JE, ZENG L, HE C, AGGARWAL AK & ZHOU MM 1999. Structure and ligand of a histone acetyltransferase bromodomain. Nature, 399, 491–6. [DOI] [PubMed] [Google Scholar]
- DI CROCE L & HELIN K 2013. Transcriptional regulation by Polycomb group proteins. Nat Struct Mol Biol, 20, 1147–55. [DOI] [PubMed] [Google Scholar]
- DING Y, LI N, DONG B, GUO W, WEI H, CHEN Q, YUAN H, HAN Y, CHANG H, KAN S, WANG X, PAN Q, WU P, PENG C, QIU T, LI Q, GAO D, XUE W & QIN J 2019. Chromatin remodeling ATPase BRG1 and PTEN are synthetic lethal in prostate cancer. J Clin Invest, 129, 759–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DU Q, LUU PL, STIRZAKER C & CLARK SJ 2015. Methyl-CpG-binding domain proteins: readers of the epigenome. Epigenomics, 7, 1051–73. [DOI] [PubMed] [Google Scholar]
- DUAN L, CHEN Z, LU J, LIANG Y, WANG M, ROGGERO CM, ZHANG QJ, GAO J, FANG Y, CAO J, LU J, ZHAO H, DANG A, PONG RC, HERNANDEZ E, CHANG CM, HOANG DT, AHN JM, XIAO G, WANG RT, YU KJ, KAPUR P, RIZO J, HSIEH JT, LUO J & LIU ZP 2019. Histone lysine demethylase KDM4B regulates the alternative splicing of the androgen receptor in response to androgen deprivation. Nucleic Acids Res, 47, 11623–11636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DUAN L, RAI G, ROGGERO C, ZHANG QJ, WEI Q, MA SH, ZHOU Y, SANTOYO J, MARTINEZ ED, XIAO G, RAJ GV, JADHAV A, SIMEONOV A, MALONEY DJ, RIZO J, HSIEH JT & LIU ZP 2015. KDM4/JMJD2 Histone Demethylase Inhibitors Block Prostate Tumor Growth by Suppressing the Expression of AR and BMYB-Regulated Genes. Chem Biol, 22, 1185–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DUAN R, DU W & GUO W 2020. EZH2: a novel target for cancer treatment. J Hematol Oncol, 13, 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DUTTA A, LE MAGNEN C, MITROFANOVA A, OUYANG X, CALIFANO A & ABATE-SHEN C 2016. Identification of an NKX3.1-G9a-UTY transcriptional regulatory network that controls prostate differentiation. Science, 352, 1576–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ECKSCHLAGER T, PLCH J, STIBOROVA M & HRABETA J 2017. Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci, 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- EDMUNDS JW, MAHADEVAN LC & CLAYTON AL 2008. Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation. EMBO J, 27, 406–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- EHRLICH M 2002. DNA methylation in cancer: too much, but also too little. Oncogene, 21, 5400–13. [DOI] [PubMed] [Google Scholar]
- FAIVRE EJ, MCDANIEL KF, ALBERT DH, MANTENA SR, PLOTNIK JP, WILCOX D, ZHANG L, BUI MH, SHEPPARD GS, WANG L, SEHGAL V, LIN X, HUANG X, LU X, UZIEL T, HESSLER P, LAM LT, BELLIN RJ, MEHTA G, FIDANZE S, PRATT JK, LIU D, HASVOLD LA, SUN C, PANCHAL SC, NICOLETTE JJ, FOSSEY SL, PARK CH, LONGENECKER K, BIGELOW L, TORRENT M, ROSENBERG SH, KATI WM & SHEN Y 2020. Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer. Nature, 578, 306–310. [DOI] [PubMed] [Google Scholar]
- FAN L, PENG G, SAHGAL N, FAZLI L, GLEAVE M, ZHANG Y, HUSSAIN A & QI J 2016. Regulation of c-Myc expression by the histone demethylase JMJD1A is essential for prostate cancer cell growth and survival. Oncogene, 35, 2441–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FAN L, ZHANG F, XU S, CUI X, HUSSAIN A, FAZLI L, GLEAVE M, DONG X & QI J 2018. Histone demethylase JMJD1A promotes alternative splicing of AR variant 7 (AR-V7) in prostate cancer cells. Proc Natl Acad Sci U S A, 115, E4584–E4593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FANG Y, LIAO G & YU B 2019. LSD1/KDM1A inhibitors in clinical trials: advances and prospects. J Hematol Oncol, 12, 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FARZANEH M, KUCHAKI Z, RASHID SHEYKHAHMAD F, MEYBODI SM, ABBASI Y, GHOLAMI E, GHAEDRAHMATI F & ANBIYAEE O 2022. Emerging roles of JMJD3 in cancer. Clin Transl Oncol, 24, 1238–1249. [DOI] [PubMed] [Google Scholar]
- FELSENFELD G & GROUDINE M 2003. Controlling the double helix. Nature, 421, 448–53. [DOI] [PubMed] [Google Scholar]
- FENG Q, WANG H, NG HH, ERDJUMENT-BROMAGE H, TEMPST P, STRUHL K & ZHANG Y 2002. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr Biol, 12, 1052–8. [DOI] [PubMed] [Google Scholar]
- FU M, LIU M, SAUVE AA, JIAO X, ZHANG X, WU X, POWELL MJ, YANG T, GU W, AVANTAGGIATI ML, PATTABIRAMAN N, PESTELL TG, WANG F, QUONG AA, WANG C & PESTELL RG 2006. Hormonal control of androgen receptor function through SIRT1. Mol Cell Biol, 26, 8122–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FU M, WANG C, REUTENS AT, WANG J, ANGELETTI RH, SICONOLFI-BAEZ L, OGRYZKO V, AVANTAGGIATI ML & PESTELL RG 2000. p300 and p300/cAMP-response element-binding protein-associated factor acetylate the androgen receptor at sites governing hormone-dependent transactivation. J Biol Chem, 275, 20853–60. [DOI] [PubMed] [Google Scholar]
- FUJISAWA T & FILIPPAKOPOULOS P 2017. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat Rev Mol Cell Biol, 18, 246–262. [DOI] [PubMed] [Google Scholar]
- GAO S, CHEN S, HAN D, WANG Z, LI M, HAN W, BESSCHETNOVA A, LIU M, ZHOU F, BARRETT D, LUONG MP, OWIREDU J, LIANG Y, AHMED M, PETRICCA J, PATALANO S, MACOSKA JA, COREY E, CHEN S, BALK SP, HE HH & CAI C 2020. Chromatin binding of FOXA1 is promoted by LSD1-mediated demethylation in prostate cancer. Nat Genet, 52, 1011–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GERSON SL 2004. MGMT: its role in cancer aetiology and cancer therapeutics. Nat Rev Cancer, 4, 296–307. [DOI] [PubMed] [Google Scholar]
- GEYBELS MS, ZHAO S, WONG CJ, BIBIKOVA M, KLOTZLE B, WU M, OSTRANDER EA, FAN JB, FENG Z & STANFORD JL 2015. Epigenomic profiling of DNA methylation in paired prostate cancer versus adjacent benign tissue. Prostate, 75, 1941–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GILAN O, RIOJA I, KNEZEVIC K, BELL MJ, YEUNG MM, HARKER NR, LAM EYN, CHUNG CW, BAMBOROUGH P, PETRETICH M, URH M, ATKINSON SJ, BASSIL AK, ROBERTS EJ, VASSILIADIS D, BURR ML, PRESTON AGS, WELLAWAY C, WERNER T, GRAY JR, MICHON AM, GOBBETTI T, KUMAR V, SODEN PE, HAYNES A, VAPPIANI J, TOUGH DF, TAYLOR S, DAWSON SJ, BANTSCHEFF M, LINDON M, DREWES G, DEMONT EH, DANIELS DL, GRANDI P, PRINJHA RK & DAWSON MA 2020. Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science, 368, 387–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GRAFF JR, HERMAN JG, LAPIDUS RG, CHOPRA H, XU R, JARRARD DF, ISAACS WB, PITHA PM, DAVIDSON NE & BAYLIN SB 1995. E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res, 55, 5195–9. [PubMed] [Google Scholar]
- GRANT S, EASLEY C & KIRKPATRICK P 2007. Vorinostat. Nat Rev Drug Discov, 6, 21–2. [DOI] [PubMed] [Google Scholar]
- GRASSO CS, WU YM, ROBINSON DR, CAO X, DHANASEKARAN SM, KHAN AP, QUIST MJ, JING X, LONIGRO RJ, BRENNER JC, ASANGANI IA, ATEEQ B, CHUN SY, SIDDIQUI J, SAM L, ANSTETT M, MEHRA R, PRENSNER JR, PALANISAMY N, RYSLIK GA, VANDIN F, RAPHAEL BJ, KUNJU LP, RHODES DR, PIENTA KJ, CHINNAIYAN AM & TOMLINS SA 2012. The mutational landscape of lethal castration-resistant prostate cancer. Nature, 487, 239–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GREENBERG MVC & BOURC’HIS D 2019. The diverse roles of DNA methylation in mammalian development and disease. Nat Rev Mol Cell Biol, 20, 590–607. [DOI] [PubMed] [Google Scholar]
- GREER EL & SHI Y 2012. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet, 13, 343–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUO J, ZHENG Q & PENG Y 2023. BET proteins: Biological functions and therapeutic interventions. Pharmacol Ther, 243, 108354. [DOI] [PubMed] [Google Scholar]
- GUO Y, ZHAO S & WANG GG 2021. Polycomb Gene Silencing Mechanisms: PRC2 Chromatin Targeting, H3K27me3 ‘Readout’, and Phase Separation-Based Compaction. Trends Genet, 37, 547–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- HAJMIRZA A, EMADALI A, GAUTHIER A, CASASNOVAS O, GRESSIN R & CALLANAN MB 2018. BET Family Protein BRD4: An Emerging Actor in NFkappaB Signaling in Inflammation and Cancer. Biomedicines, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- HE Y, ZHAO Y, WANG L, BOHRER LR, PAN Y, WANG L & HUANG H 2018. LSD1 promotes S-phase entry and tumorigenesis via chromatin co-occupation with E2F1 and selective H3K9 demethylation. Oncogene, 37, 534–543. [DOI] [PubMed] [Google Scholar]
- HERMAN JG, MERLO A, MAO L, LAPIDUS RG, ISSA JP, DAVIDSON NE, SIDRANSKY D & BAYLIN SB 1995. Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res, 55, 4525–30. [PubMed] [Google Scholar]
- HOY SM 2020. Tazemetostat: First Approval. Drugs, 80, 513–521. [DOI] [PubMed] [Google Scholar]
- HUANG J, SENGUPTA R, ESPEJO AB, LEE MG, DORSEY JA, RICHTER M, OPRAVIL S, SHIEKHATTAR R, BEDFORD MT, JENUWEIN T & BERGER SL 2007. p53 is regulated by the lysine demethylase LSD1. Nature, 449, 105–8. [DOI] [PubMed] [Google Scholar]
- HUANG L & XU AM 2017. SET and MYND domain containing protein 3 in cancer. Am J Transl Res, 9, 1–14. [PMC free article] [PubMed] [Google Scholar]
- HUANG SB, THAPA D, MUNOZ AR, HUSSAIN SS, YANG X, BEDOLLA RG, OSMULSKI P, GACZYNSKA ME, LAI Z, CHIU YC, WANG LJ, CHEN Y, RIVAS P, SHUDDE C, REDDICK RL, MIYAMOTO H, GHOSH R & KUMAR AP 2021. Androgen deprivation-induced elevated nuclear SIRT1 promotes prostate tumor cell survival by reactivation of AR signaling. Cancer Lett, 505, 24–36. [DOI] [PubMed] [Google Scholar]
- HUGGINS C & HODGES CV 1972. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J Clin, 22, 232–40. [DOI] [PubMed] [Google Scholar]
- HULETT MD, FREEMAN C, HAMDORF BJ, BAKER RT, HARRIS MJ & PARISH CR 1999. Cloning of mammalian heparanase, an important enzyme in tumor invasion and metastasis. Nat Med, 5, 803–9. [DOI] [PubMed] [Google Scholar]
- HUSMANN D & GOZANI O 2019. Histone lysine methyltransferases in biology and disease. Nat Struct Mol Biol, 26, 880–889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- IKEDA Y & KINOSHITA T 2009. DNA demethylation: a lesson from the garden. Chromosoma, 118, 37–41. [DOI] [PubMed] [Google Scholar]
- ILLINGWORTH R, KERR A, DESOUSA D, JORGENSEN H, ELLIS P, STALKER J, JACKSON D, CLEE C, PLUMB R, ROGERS J, HUMPHRAY S, COX T, LANGFORD C & BIRD A 2008. A novel CpG island set identifies tissue-specific methylation at developmental gene loci. PLoS Biol, 6, e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ISSA GC, ALDOSS I, DIPERSIO J, CUGLIEVAN B, STONE R, ARELLANO M, THIRMAN MJ, PATEL MR, DICKENS DS, SHENOY S, SHUKLA N, KANTARJIAN H, ARMSTRONG SA, PERNER F, PERRY JA, ROSEN G, BAGLEY RG, MEYERS ML, ORDENTLICH P, GU Y, KUMAR V, SMITH S, MCGEEHAN GM & STEIN EM 2023. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature, 615, 920–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JAIN P, BALLARE C, BLANCO E, VIZAN P & DI CROCE L 2020. PHF19 mediated regulation of proliferation and invasiveness in prostate cancer cells. Elife, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JAIN P & DI CROCE L 2016. Mutations and deletions of PRC2 in prostate cancer. Bioessays, 38, 446–54. [DOI] [PubMed] [Google Scholar]
- JAISWAL B, AGARWAL A & GUPTA A 2022. Lysine Acetyltransferases and Their Role in AR Signaling and Prostate Cancer. Front Endocrinol (Lausanne), 13, 886594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JANG MK, MOCHIZUKI K, ZHOU M, JEONG HS, BRADY JN & OZATO K 2005. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol Cell, 19, 523–34. [DOI] [PubMed] [Google Scholar]
- JANOUSKOVA H, EL TEKLE G, BELLINI E, UDESHI ND, RINALDI A, ULBRICHT A, BERNASOCCHI T, CIVENNI G, LOSA M, SVINKINA T, BIELSKI CM, KRYUKOV GV, CASCIONE L, NAPOLI S, ENCHEV RI, MUTCH DG, CARNEY ME, BERCHUCK A, WINTERHOFF BJN, BROADDUS RR, SCHRAML P, MOCH H, BERTONI F, CATAPANO CV, PETER M, CARR SA, GARRAWAY LA, WILD PJ & THEURILLAT JP 2017. Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat Med, 23, 1046–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JARRARD DF, KINOSHITA H, SHI Y, SANDEFUR C, HOFF D, MEISNER LF, CHANG C, HERMAN JG, ISAACS WB & NASSIF N 1998. Methylation of the androgen receptor promoter CpG island is associated with loss of androgen receptor expression in prostate cancer cells. Cancer Res, 58, 5310–4. [PubMed] [Google Scholar]
- JEONG GY, PARK MK, CHOI HJ, AN HW, PARK YU, CHOI HJ, PARK J, KIM HY, SON T, LEE H, MIN KW, OH YH, LEE JY & KONG G 2021. NSD3-Induced Methylation of H3K36 Activates NOTCH Signaling to Drive Breast Tumor Initiation and Metastatic Progression. Cancer Res, 81, 77–90. [DOI] [PubMed] [Google Scholar]
- JERONIMO C, HENRIQUE R, HOQUE MO, RIBEIRO FR, OLIVEIRA J, FONSECA D, TEIXEIRA MR, LOPES C & SIDRANSKY D 2004. Quantitative RARbeta2 hypermethylation: a promising prostate cancer marker. Clin Cancer Res, 10, 4010–4. [DOI] [PubMed] [Google Scholar]
- JIAO L, SHUBBAR M, YANG X, ZHANG Q, CHEN S, WU Q, CHEN Z, RIZO J & LIU X 2020. A partially disordered region connects gene repression and activation functions of EZH2. Proc Natl Acad Sci U S A, 117, 16992–17002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JIN L, GARCIA J, CHAN E, DE LA CRUZ C, SEGAL E, MERCHANT M, KHARBANDA S, RAISNER R, HAVERTY PM, MODRUSAN Z, LY J, CHOO E, KAUFMAN S, BERESINI MH, ROMERO FA, MAGNUSON S & GASCOIGNE KE 2017. Therapeutic Targeting of the CBP/p300 Bromodomain Blocks the Growth of Castration-Resistant Prostate Cancer. Cancer Res, 77, 5564–5575. [DOI] [PubMed] [Google Scholar]
- JONES PA 2012. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet, 13, 484–92. [DOI] [PubMed] [Google Scholar]
- JONES PL, VEENSTRA GJ, WADE PA, VERMAAK D, KASS SU, LANDSBERGER N, STROUBOULIS J & WOLFFE AP 1998. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet, 19, 187–91. [DOI] [PubMed] [Google Scholar]
- KADOCH C, HARGREAVES DC, HODGES C, ELIAS L, HO L, RANISH J & CRABTREE GR 2013. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet, 45, 592–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KANG HB, CHOI Y, LEE JM, CHOI KC, KIM HC, YOO JY, LEE YH & YOON HG 2009. The histone methyltransferase, NSD2, enhances androgen receptor-mediated transcription. FEBS Lett, 583, 1880–6. [DOI] [PubMed] [Google Scholar]
- KANOUNI T, SEVERIN C, CHO RW, YUEN NY, XU J, SHI L, LAI C, DEL ROSARIO JR, STANSFIELD RK, LAWTON LN, HOSFIELD D, O’CONNELL S, KREILEIN MM, TAVARES-GRECO P, NIE Z, KALDOR SW, VEAL JM, STAFFORD JA & CHEN YK 2020. Discovery of CC-90011: A Potent and Selective Reversible Inhibitor of Lysine Specific Demethylase 1 (LSD1). J Med Chem, 63, 14522–14529. [DOI] [PubMed] [Google Scholar]
- KARI V, MANSOUR WY, RAUL SK, BAUMGART SJ, MUND A, GRADE M, SIRMA H, SIMON R, WILL H, DOBBELSTEIN M, DIKOMEY E & JOHNSEN SA 2016. Loss of CHD1 causes DNA repair defects and enhances prostate cancer therapeutic responsiveness. EMBO Rep, 17, 1609–1623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KIM E, KIM M, WOO DH, SHIN Y, SHIN J, CHANG N, OH YT, KIM H, RHEEY J, NAKANO I, LEE C, JOO KM, RICH JN, NAM DH & LEE J 2013. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell, 23, 839–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KIM J, LEE Y, LU X, SONG B, FONG KW, CAO Q, LICHT JD, ZHAO JC & YU J 2018. Polycomb- and Methylation-Independent Roles of EZH2 as a Transcription Activator. Cell Rep, 25, 2808–2820 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KIM JJ & KINGSTON RE 2022. Context-specific Polycomb mechanisms in development. Nat Rev Genet, 23, 680–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KIM TD, JIN F, SHIN S, OH S, LIGHTFOOT SA, GRANDE JP, JOHNSON AJ, VAN DEURSEN JM, WREN JD & JANKNECHT R 2016. Histone demethylase JMJD2A drives prostate tumorigenesis through transcription factor ETV1. J Clin Invest, 126, 706–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KIRBY MK, RAMAKER RC, ROBERTS BS, LASSEIGNE BN, GUNTHER DS, BURWELL TC, DAVIS NS, GULZAR ZG, ABSHER DM, COOPER SJ, BROOKS JD & MYERS RM 2017. Genome-wide DNA methylation measurements in prostate tissues uncovers novel prostate cancer diagnostic biomarkers and transcription factor binding patterns. BMC Cancer, 17, 273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KLOSE RJ, YAMANE K, BAE Y, ZHANG D, ERDJUMENT-BROMAGE H, TEMPST P, WONG J & ZHANG Y 2006. The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36. Nature, 442, 312–6. [DOI] [PubMed] [Google Scholar]
- KOBAYASHI Y, ABSHER DM, GULZAR ZG, YOUNG SR, MCKENNEY JK, PEEHL DM, BROOKS JD, MYERS RM & SHERLOCK G 2011. DNA methylation profiling reveals novel biomarkers and important roles for DNA methyltransferases in prostate cancer. Genome Res, 21, 1017–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KOMURA K, JEONG SH, HINOHARA K, QU F, WANG X, HIRAKI M, AZUMA H, LEE GS, KANTOFF PW & SWEENEY CJ 2016. Resistance to docetaxel in prostate cancer is associated with androgen receptor activation and loss of KDM5D expression. Proc Natl Acad Sci U S A, 113, 6259–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KOMURA K, YOSHIKAWA Y, SHIMAMURA T, CHAKRABORTY G, GERKE TA, HINOHARA K, CHADALAVADA K, JEONG SH, ARMENIA J, DU SY, MAZZU YZ, TANIGUCHI K, IBUKI N, MEYER CA, NANJANGUD GJ, INAMOTO T, LEE GM, MUCCI LA, AZUMA H, SWEENEY CJ & KANTOFF PW 2018. ATR inhibition controls aggressive prostate tumors deficient in Y-linked histone demethylase KDM5D. J Clin Invest, 128, 2979–2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KREGEL S, MALIK R, ASANGANI IA, WILDER-ROMANS K, RAJENDIRAN T, XIAO L, VO JN, SONI T, CIESLIK M, FERNADEZ-SALAS E, ZHOU B, CAO X, SPEERS C, WANG S & CHINNAIYAN AM 2019. Functional and Mechanistic Interrogation of BET Bromodomain Degraders for the Treatment of Metastatic Castration-resistant Prostate Cancer. Clin Cancer Res, 25, 4038–4048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KU SY, ROSARIO S, WANG Y, MU P, SESHADRI M, GOODRICH ZW, GOODRICH MM, LABBE DP, GOMEZ EC, WANG J, LONG HW, XU B, BROWN M, LODA M, SAWYERS CL, ELLIS L & GOODRICH DW 2017. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science, 355, 78–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KUKKONEN K, TAAVITSAINEN S, HUHTALA L, UUSI-MAKELA J, GRANBERG KJ, NYKTER M & URBANUCCI A 2021. Chromatin and Epigenetic Dysregulation of Prostate Cancer Development, Progression, and Therapeutic Response. Cancers (Basel), 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KUMARASWAMY A, WELKER LENG KR, WESTBROOK TC, YATES JA, ZHAO SG, EVANS CP, FENG FY, MORGAN TM & ALUMKAL JJ 2021. Recent Advances in Epigenetic Biomarkers and Epigenetic Targeting in Prostate Cancer. Eur Urol, 80, 71–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KUZMIN I, GILLESPIE JW, PROTOPOPOV A, GEIL L, DREIJERINK K, YANG Y, VOCKE CD, DUH FM, ZABAROVSKY E, MINNA JD, RHIM JS, EMMERT-BUCK MR, LINEHAN WM & LERMAN MI 2002. The RASSF1A tumor suppressor gene is inactivated in prostate tumors and suppresses growth of prostate carcinoma cells. Cancer Res, 62, 3498–502. [PubMed] [Google Scholar]
- LAKSHMIKANTHAN V, KADDOUR-DJEBBAR I, LEWIS RW & KUMAR MV 2006. SAHA-sensitized prostate cancer cells to TNFalpha-related apoptosis-inducing ligand (TRAIL): mechanisms leading to synergistic apoptosis. Int J Cancer, 119, 221–8. [DOI] [PubMed] [Google Scholar]
- LASKO LM, JAKOB CG, EDALJI RP, QIU W, MONTGOMERY D, DIGIAMMARINO EL, HANSEN TM, RISI RM, FREY R, MANAVES V, SHAW B, ALGIRE M, HESSLER P, LAM LT, UZIEL T, FAIVRE E, FERGUSON D, BUCHANAN FG, MARTIN RL, TORRENT M, CHIANG GG, KARUKURICHI K, LANGSTON JW, WEINERT BT, CHOUDHARY C, DE VRIES P, VAN DRIE JH, MCELLIGOTT D, KESICKI E, MARMORSTEIN R, SUN C, COLE PA, ROSENBERG SH, MICHAELIDES MR, LAI A & BROMBERG KD 2017. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature, 550, 128–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LAU KM, LASPINA M, LONG J & HO SM 2000. Expression of estrogen receptor (ER)-alpha and ER-beta in normal and malignant prostatic epithelial cells: regulation by methylation and involvement in growth regulation. Cancer Res, 60, 3175–82. [PubMed] [Google Scholar]
- LAVERY DN & BEVAN CL 2011. Androgen receptor signalling in prostate cancer: the functional consequences of acetylation. J Biomed Biotechnol, 2011, 862125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LEE KH, HONG S, KANG M, JEONG CW, KU JH, KIM HH & KWAK C 2018. Histone demethylase KDM7A controls androgen receptor activity and tumor growth in prostate cancer. Int J Cancer, 143, 2849–2861. [DOI] [PubMed] [Google Scholar]
- LEE WH, MORTON RA, EPSTEIN JI, BROOKS JD, CAMPBELL PA, BOVA GS, HSIEH WS, ISAACS WB & NELSON WG 1994. Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A, 91, 11733–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LEMSTER AL, SIEVERS E, PASTERNACK H, LAZAR-KARSTEN P, KLUMPER N, SAILER V, OFFERMANN A, BRAGELMANN J, PERNER S & KIRFEL J 2022. Histone Demethylase KDM5C Drives Prostate Cancer Progression by Promoting EMT. Cancers (Basel), 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI C, WANG Y, GONG Y, ZHANG T, HUANG J, TAN Z & XUE L 2021. Finding an easy way to harmonize: a review of advances in clinical research and combination strategies of EZH2 inhibitors. Clin Epigenetics, 13, 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI G, KANAGASABAI T, LU W, ZOU MR, ZHANG SM, CELADA SI, IZBAN MG, LIU Q, LU T, BALLARD BR, ZHOU X, ADUNYAH SE, MATUSIK RJ, YAN Q & CHEN Z 2020. KDM5B Is Essential for the Hyperactivation of PI3K/AKT Signaling in Prostate Tumorigenesis. Cancer Res, 80, 4633–4643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI H, LIEFKE R, JIANG J, KURLAND JV, TIAN W, DENG P, ZHANG W, HE Q, PATEL DJ, BULYK ML, SHI Y & WANG Z 2017a. Polycomb-like proteins link the PRC2 complex to CpG islands. Nature, 549, 287–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI J, AHN JH & WANG GG 2019. Understanding histone H3 lysine 36 methylation and its deregulation in disease. Cell Mol Life Sci, 76, 2899–2916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI LC, ZHAO H, NAKAJIMA K, OH BR, RIBEIRO FILHO LA, CARROLL P & DAHIYA R 2001. Methylation of the E-cadherin gene promoter correlates with progression of prostate cancer. J Urol, 166, 705–9. [PubMed] [Google Scholar]
- LI N, LI Y, LV J, ZHENG X, WEN H, SHEN H, ZHU G, CHEN TY, DHAR SS, KAN PY, WANG Z, SHIEKHATTAR R, SHI X, LAN F, CHEN K, LI W, LI H & LEE MG 2016. ZMYND8 Reads the Dual Histone Mark H3K4me1-H3K14ac to Antagonize the Expression of Metastasis-Linked Genes. Mol Cell, 63, 470–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI N, LIU Q, HAN Y, PEI S, CHENG B, XU J, MIAO X, PAN Q, WANG H, GUO J, WANG X, ZHANG G, LIAN Y, ZHANG W, ZANG Y, TAN M, LI Q, WANG X, XIAO Y, HU G, JIANG J, HUANG H & QIN J 2022a. ARID1A loss induces polymorphonuclear myeloid-derived suppressor cell chemotaxis and promotes prostate cancer progression. Nat Commun, 13, 7281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI N, XUE W, YUAN H, DONG B, DING Y, LIU Y, JIANG M, KAN S, SUN T, REN J, PAN Q, LI X, ZHANG P, HU G, WANG Y, WANG X, LI Q & QIN J 2017b. AKT-mediated stabilization of histone methyltransferase WHSC1 promotes prostate cancer metastasis. J Clin Invest, 127, 1284–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI X, BAEK G, CARREIRA S, YUAN W, MA S, HOFSTAD M, LEE S, GAO Y, BERTAN C, FENOR DE LA MAZA MLD, ALLURI PG, BURMA S, CHEN BP, RAJ GV, DE BONO J, POMMIER Y & MANI RS 2022b. Targeting radioresistance and replication fork stability in prostate cancer. JCI Insight, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LI X, BAEK G, RAMANAND SG, SHARP A, GAO Y, YUAN W, WELTI J, RODRIGUES DN, DOLLING D, FIGUEIREDO I, SUMANASURIYA S, CRESPO M, ASLAM A, LI R, YIN Y, MUKHERJEE B, KANCHWALA M, HUGHES AM, HALSEY WS, CHIANG CM, XING C, RAJ GV, BURMA S, DE BONO J & MANI RS 2018. BRD4 Promotes DNA Repair and Mediates the Formation of TMPRSS2-ERG Gene Rearrangements in Prostate Cancer. Cell Rep, 22, 796–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LIANG Y, AHMED M, GUO H, SOARES F, HUA JT, GAO S, LU C, POON C, HAN W, LANGSTEIN J, EKRAM MB, LI B, DAVICIONI E, TAKHAR M, ERHO N, KARNES RJ, CHADWICK D, VAN DER KWAST T, BOUTROS PC, ARROWSMITH CH, FENG FY, JOSHUA AM, ZOUBEIDI A, CAI C & HE HH 2017. LSD1-Mediated Epigenetic Reprogramming Drives CENPE Expression and Prostate Cancer Progression. Cancer Res, 77, 5479–5490. [DOI] [PubMed] [Google Scholar]
- LIN J, WU Y, TIAN G, YU D, YANG E, LAM WH, LIU Z, JING Y, DANG S, BAO X, WONG JWH, ZHAI Y & LI XD 2023. Menin “reads” H3K79me2 mark in a nucleosomal context. Science, 379, 717–723. [DOI] [PubMed] [Google Scholar]
- LINK KA, BALASUBRAMANIAM S, SHARMA A, COMSTOCK CE, GODOY-TUNDIDOR S, POWERS N, CAO KH, HAELENS A, CLAESSENS F, REVELO MP & KNUDSEN KE 2008. Targeting the BAF57 SWI/SNF subunit in prostate cancer: a novel platform to control androgen receptor activity. Cancer Res, 68, 4551–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LIU C, WANG C, WANG K, LIU L, SHEN Q, YAN K, SUN X, CHEN J, LIU J, REN H, LIU H, XU Z, HU S, XU D & FAN Y 2013. SMYD3 as an oncogenic driver in prostate cancer by stimulation of androgen receptor transcription. J Natl Cancer Inst, 105, 1719–28. [DOI] [PubMed] [Google Scholar]
- LIU J, HE D, CHENG L, HUANG C, ZHANG Y, RAO X, KONG Y, LI C, ZHANG Z, LIU J, JONES K, NAPIER D, LEE EY, WANG C & LIU X 2020. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene, 39, 3939–3951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LIU L, YOON JH, DAMMANN R & PFEIFER GP 2002. Frequent hypermethylation of the RASSF1A gene in prostate cancer. Oncogene, 21, 6835–40. [DOI] [PubMed] [Google Scholar]
- LU X, PAN X, WU CJ, ZHAO D, FENG S, ZANG Y, LEE R, KHADKA S, AMIN SB, JIN EJ, SHANG X, DENG P, LUO Y, MORGENLANDER WR, WEINRICH J, LU X, JIANG S, CHANG Q, NAVONE NM, TRONCOSO P, DEPINHO RA & WANG YA 2018. An In Vivo Screen Identifies PYGO2 as a Driver for Metastatic Prostate Cancer. Cancer Res, 78, 3823–3833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LUCO RF, PAN Q, TOMINAGA K, BLENCOWE BJ, PEREIRA-SMITH OM & MISTELI T 2010. Regulation of alternative splicing by histone modifications. Science, 327, 996–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LV S, JI L, CHEN B, LIU S, LEI C, LIU X, QI X, WANG Y, LAI-HAN LEUNG E, WANG H, ZHANG L, YU X, LIU Z, WEI Q & LU L 2018. Histone methyltransferase KMT2D sustains prostate carcinogenesis and metastasis via epigenetically activating LIFR and KLF4. Oncogene, 37, 1354–1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MAES T, MASCARO C, TIRAPU I, ESTIARTE A, CICERI F, LUNARDI S, GUIBOURT N, PERDONES A, LUFINO MMP, SOMERVAILLE TCP, WISEMAN DH, DUY C, MELNICK A, WILLEKENS C, ORTEGA A, MARTINELL M, VALLS N, KURZ G, FYFE M, CASTRO-PALOMINO JC & BUESA C 2018. ORY-1001, a Potent and Selective Covalent KDM1A Inhibitor, for the Treatment of Acute Leukemia. Cancer Cell, 33, 495–511 e12. [DOI] [PubMed] [Google Scholar]
- MAHON KL, QU W, DEVANEY J, PAUL C, CASTILLO L, WYKES RJ, CHATFIELD MD, BOYER MJ, STOCKLER MR, MARX G, GURNEY H, MALLESARA G, MOLLOY PL, HORVATH LG, CLARK SJ & CONSORTIUM PR 2014. Methylated Glutathione S-transferase 1 (mGSTP1) is a potential plasma free DNA epigenetic marker of prognosis and response to chemotherapy in castrate-resistant prostate cancer. Br J Cancer, 111, 1802–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MALIK R, KHAN AP, ASANGANI IA, CIESLIK M, PRENSNER JR, WANG X, IYER MK, JIANG X, BORKIN D, ESCARA-WILKE J, STENDER R, WU YM, NIKNAFS YS, JING X, QIAO Y, PALANISAMY N, KUNJU LP, KRISHNAMURTHY PM, YOCUM AK, MELLACHERUVU D, NESVIZHSKII AI, CAO X, DHANASEKARAN SM, FENG FY, GREMBECKA J, CIERPICKI T & CHINNAIYAN AM 2015. Targeting the MLL complex in castration-resistant prostate cancer. Nat Med, 21, 344–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MARINO-RAMIREZ L, KANN MG, SHOEMAKER BA & LANDSMAN D 2005. Histone structure and nucleosome stability. Expert Rev Proteomics, 2, 719–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MCLEOD AB, STICE JP, WARDELL SE, ALLEY HM, CHANG CY & MCDONNELL DP 2018. Validation of histone deacetylase 3 as a therapeutic target in castration-resistant prostate cancer. Prostate, 78, 266–277. [DOI] [PubMed] [Google Scholar]
- METZGER E, WISSMANN M, YIN N, MULLER JM, SCHNEIDER R, PETERS AH, GUNTHER T, BUETTNER R & SCHULE R 2005. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature, 437, 436–9. [DOI] [PubMed] [Google Scholar]
- MITTAL P & ROBERTS CWM 2020. The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol, 17, 435–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MOHAMMAD HP, SMITHEMAN KN, KAMAT CD, SOONG D, FEDEROWICZ KE, VAN ALLER GS, SCHNECK JL, CARSON JD, LIU Y, BUTTICELLO M, BONNETTE WG, GORMAN SA, DEGENHARDT Y, BAI Y, MCCABE MT, PAPPALARDI MB, KASPAREC J, TIAN X, MCNULTY KC, ROUSE M, MCDEVITT P, HO T, CROUTHAMEL M, HART TK, CONCHA NO, MCHUGH CF, MILLER WH, DHANAK D, TUMMINO PJ, CARPENTER CL, JOHNSON NW, HANN CL & KRUGER RG 2015. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell, 28, 57–69. [DOI] [PubMed] [Google Scholar]
- MOREL KL, SHEAHAN AV, BURKHART DL, BACA SC, BOUFAIED N, LIU Y, QIU X, CANADAS I, ROEHLE K, HECKLER M, CALAGUA C, YE H, PANTELIDOU C, GALBO P, PANJA S, MITROFANOVA A, WILKINSON S, WHITLOCK NC, TROSTEL SY, HAMID AA, KIBEL AS, BARBIE DA, CHOUDHURY AD, POMERANTZ MM, SWEENEY CJ, LONG HW, EINSTEIN DJ, SHAPIRO GI, DOUGAN SK, SOWALSKY AG, HE HH, FREEDMAN ML, BALK SP, LODA M, LABBE DP, OLSON BM & ELLIS L 2021. EZH2 inhibition activates a dsRNA-STING-interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer. Nat Cancer, 2, 444–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MUTHUSWAMI R, BAILEY L, RAKESH R, IMBALZANO AN, NICKERSON JA & HOCKENSMITH JW 2019. BRG1 is a prognostic indicator and a potential therapeutic target for prostate cancer. J Cell Physiol, 234, 15194–15205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NAN X, NG HH, JOHNSON CA, LAHERTY CD, TURNER BM, EISENMAN RN & BIRD A 1998. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature, 393, 386–9. [DOI] [PubMed] [Google Scholar]
- NASSA G, SALVATI A, TARALLO R, GIGANTINO V, ALEXANDROVA E, MEMOLI D, SELLITTO A, RIZZO F, MALANGA D, MIRANTE T, MORELLI E, NEES M, AKERFELT M, KANGASPESKA S, NYMAN TA, MILANESI L, GIURATO G & WEISZ A 2019. Inhibition of histone methyltransferase DOT1L silences ERalpha gene and blocks proliferation of antiestrogen-resistant breast cancer cells. Sci Adv, 5, eaav5590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NATESAN R, ARAS S, EFFRON SS & ASANGANI IA 2019. Epigenetic Regulation of Chromatin in Prostate Cancer. Adv Exp Med Biol, 1210, 379–407. [DOI] [PubMed] [Google Scholar]
- NG HH, ROBERT F, YOUNG RA & STRUHL K 2003. Targeted recruitment of Set1 histone methylase by elongating Pol II provides a localized mark and memory of recent transcriptional activity. Mol Cell, 11, 709–19. [DOI] [PubMed] [Google Scholar]
- NGUYEN AT & ZHANG Y 2011. The diverse functions of Dot1 and H3K79 methylation. Genes Dev, 25, 1345–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NICKERSON ML, DAS S, IM KM, TURAN S, BERNDT SI, LI H, LOU H, BRODIE SA, BILLAUD JN, ZHANG T, BOUK AJ, BUTCHER D, WANG Z, SUN L, MISNER K, TAN W, ESNAKULA A, ESPOSITO D, HUANG WY, HOOVER RN, TUCKER MA, KELLER JR, BOLAND J, BROWN K, ANDERSON SK, MOORE LE, ISAACS WB, CHANOCK SJ, YEAGER M, DEAN M & ANDRESSON T 2017. TET2 binds the androgen receptor and loss is associated with prostate cancer. Oncogene, 36, 2172–2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NOWACKA-ZAWISZA M & WISNIK E 2017. DNA methylation and histone modifications as epigenetic regulation in prostate cancer (Review). Oncol Rep, 38, 2587–2596. [DOI] [PubMed] [Google Scholar]
- OKADA Y, FENG Q, LIN Y, JIANG Q, LI Y, COFFIELD VM, SU L, XU G & ZHANG Y 2005. hDOT1L links histone methylation to leukemogenesis. Cell, 121, 167–78. [DOI] [PubMed] [Google Scholar]
- PADEKEN J, METHOT SP & GASSER SM 2022. Establishment of H3K9-methylated heterochromatin and its functions in tissue differentiation and maintenance. Nat Rev Mol Cell Biol, 23, 623–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PAKNESHAN P, XING RH & RABBANI SA 2003. Methylation status of uPA promoter as a molecular mechanism regulating prostate cancer invasion and growth in vitro and in vivo. FASEB J, 17, 1081–8. [DOI] [PubMed] [Google Scholar]
- PARK SH, FONG KW, KIM J, WANG F, LU X, LEE Y, BREA LT, WADOSKY K, GUO C, ABDULKADIR SA, CRISPINO JD, FANG D, NTZIACHRISTOS P, LIU X, LI X, WAN Y, GOODRICH DW, ZHAO JC & YU J 2021a. Posttranslational regulation of FOXA1 by Polycomb and BUB3/USP7 deubiquitin complex in prostate cancer. Sci Adv, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PARK SH, FONG KW, MONG E, MARTIN MC, SCHILTZ GE & YU J 2021b. Going beyond Polycomb: EZH2 functions in prostate cancer. Oncogene, 40, 5788–5798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PARTIN AW, VAN NESTE L, KLEIN EA, MARKS LS, GEE JR, TROYER DA, RIEGER-CHRIST K, JONES JS, MAGI-GALLUZZI C, MANGOLD LA, TROCK BJ, LANCE RS, BIGLEY JW, VAN CRIEKINGE W & EPSTEIN JI 2014. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol, 192, 1081–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PATEL GK, CHUGH N & TRIPATHI M 2019a. Neuroendocrine Differentiation of Prostate Cancer-An Intriguing Example of Tumor Evolution at Play. Cancers (Basel), 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PATEL PG, WESSEL T, KAWASHIMA A, OKELLO JBA, JAMASPISHVILI T, GUERARD KP, LEE L, LEE AY, HOW NE, DION D, SCARLATA E, JACKSON CL, BOURSALIE S, SACK T, DUNN R, MOUSSA M, MACKIE K, ELLIS A, MARRA E, CHIN J, SIDDIQUI K, HETOU K, PICKARD LA, ARTHUR-HAYWARD V, BAUMAN G, CHEVALIER S, BRIMO F, BOUTROS PC, LAPOINTE PH DJ, BARTLETT JMS, GOODING RJ & BERMAN DM 2019b. A three-gene DNA methylation biomarker accurately classifies early stage prostate cancer. Prostate, 79, 1705–1714. [DOI] [PubMed] [Google Scholar]
- PEREZ-SALVIA M & ESTELLER M 2017. Bromodomain inhibitors and cancer therapy: From structures to applications. Epigenetics, 12, 323–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PIUNTI A & SHILATIFARD A 2021. The roles of Polycomb repressive complexes in mammalian development and cancer. Nat Rev Mol Cell Biol, 22, 326–345. [DOI] [PubMed] [Google Scholar]
- POKHOLOK DK, HARBISON CT, LEVINE S, COLE M, HANNETT NM, LEE TI, BELL GW, WALKER K, ROLFE PA, HERBOLSHEIMER E, ZEITLINGER J, LEWITTER F, GIFFORD DK & YOUNG RA 2005. Genome-wide map of nucleosome acetylation and methylation in yeast. Cell, 122, 517–27. [DOI] [PubMed] [Google Scholar]
- POMERANTZ MM, QIU X, ZHU Y, TAKEDA DY, PAN W, BACA SC, GUSEV A, KORTHAUER KD, SEVERSON TM, HA G, VISWANATHAN SR, SEO JH, NGUYEN HM, ZHANG B, PASANIUC B, GIAMBARTOLOMEI C, ALAIWI SA, BELL CA, O’CONNOR EP, CHABOT MS, STILLMAN DR, LIS R, FONT-TELLO A, LI L, CEJAS P, BERGMAN AM, SANDERS J, VAN DER POEL HG, GAYTHER SA, LAWRENSON K, FONSECA MAS, REDDY J, CORONA RI, MARTOVETSKY G, EGAN B, CHOUEIRI T, ELLIS L, GARRAWAY IP, LEE GM, COREY E, LONG HW, ZWART W & FREEDMAN ML 2020. Prostate cancer reactivates developmental epigenomic programs during metastatic progression. Nat Genet, 52, 790–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- POWELL MJ, CASIMIRO MC, CORDON-CARDO C, HE X, YEOW WS, WANG C, MCCUE PA, MCBURNEY MW & PESTELL RG 2011. Disruption of a Sirt1-dependent autophagy checkpoint in the prostate results in prostatic intraepithelial neoplasia lesion formation. Cancer Res, 71, 964–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PUCA L, BAREJA R, PRANDI D, SHAW R, BENELLI M, KARTHAUS WR, HESS J, SIGOUROS M, DONOGHUE A, KOSSAI M, GAO D, CYRTA J, SAILER V, VOSOUGHI A, PAULI C, CHURAKOVA Y, CHEUNG C, DEONARINE LD, MCNARY TJ, ROSATI R, TAGAWA ST, NANUS DM, MOSQUERA JM, SAWYERS CL, CHEN Y, INGHIRAMI G, RAO RA, GRANDORI C, ELEMENTO O, SBONER A, DEMICHELIS F, RUBIN MA & BELTRAN H 2018. Patient derived organoids to model rare prostate cancer phenotypes. Nat Commun, 9, 2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- QUIGLEY DA, DANG HX, ZHAO SG, LLOYD P, AGGARWAL R, ALUMKAL JJ, FOYE A, KOTHARI V, PERRY MD, BAILEY AM, PLAYDLE D, BARNARD TJ, ZHANG L, ZHANG J, YOUNGREN JF, CIESLIK MP, PAROLIA A, BEER TM, THOMAS G, CHI KN, GLEAVE M, LACK NA, ZOUBEIDI A, REITER RE, RETTIG MB, WITTE O, RYAN CJ, FONG L, KIM W, FRIEDLANDER T, CHOU J, LI H, DAS R, LI H, MOUSSAVI-BAYGI R, GOODARZI H, GILBERT LA, LARA PN JR., EVANS CP, GOLDSTEIN TC, STUART JM, TOMLINS SA, SPRATT DE, CHEETHAM RK, CHENG DT, FARH K, GEHRING JS, HAKENBERG J, LIAO A, FEBBO PG, SHON J, SICKLER B, BATZOGLOU S, KNUDSEN KE, HE HH, HUANG J, WYATT AW, DEHM SM, ASHWORTH A, CHINNAIYAN AM, MAHER CA, SMALL EJ & FENG FY 2018. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell, 174, 758–769 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RADA-IGLESIAS A, BAJPAI R, SWIGUT T, BRUGMANN SA, FLYNN RA & WYSOCKA J 2011. A unique chromatin signature uncovers early developmental enhancers in humans. Nature, 470, 279–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RAINA K, LU J, QIAN Y, ALTIERI M, GORDON D, ROSSI AM, WANG J, CHEN X, DONG H, SIU K, WINKLER JD, CREW AP, CREWS CM & COLEMAN KG 2016. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A, 113, 7124–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RANA Z, DIERMEIER S, HANIF M & ROSENGREN RJ 2020. Understanding Failure and Improving Treatment Using HDAC Inhibitors for Prostate Cancer. Biomedicines, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- REN R, HORTON JR, ZHANG X, BLUMENTHAL RM & CHENG X 2018. Detecting and interpreting DNA methylation marks. Curr Opin Struct Biol, 53, 88–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RICHIARDI L, FIANO V, VIZZINI L, DE MARCO L, DELSEDIME L, AKRE O, TOS AG & MERLETTI F 2009. Promoter methylation in APC, RUNX3, and GSTP1 and mortality in prostate cancer patients. J Clin Oncol, 27, 3161–8. [DOI] [PubMed] [Google Scholar]
- RIOS-COLON L, CAJIGAS-DU ROSS CK, BASU A, ELIX C, ALICEA-POLANCO I, SANCHEZ TW, RADHAKRISHNAN V, CHEN CS & CASIANO CA 2017. Targeting the stress oncoprotein LEDGF/p75 to sensitize chemoresistant prostate cancer cells to taxanes. Oncotarget, 8, 24915–24931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ROBINSON D, VAN ALLEN EM, WU YM, SCHULTZ N, LONIGRO RJ, MOSQUERA JM, MONTGOMERY B, TAPLIN ME, PRITCHARD CC, ATTARD G, BELTRAN H, ABIDA W, BRADLEY RK, VINSON J, CAO X, VATS P, KUNJU LP, HUSSAIN M, FENG FY, TOMLINS SA, COONEY KA, SMITH DC, BRENNAN C, SIDDIQUI J, MEHRA R, CHEN Y, RATHKOPF DE, MORRIS MJ, SOLOMON SB, DURACK JC, REUTER VE, GOPALAN A, GAO J, LODA M, LIS RT, BOWDEN M, BALK SP, GAVIOLA G, SOUGNEZ C, GUPTA M, YU EY, MOSTAGHEL EA, CHENG HH, MULCAHY H, TRUE LD, PLYMATE SR, DVINGE H, FERRALDESCHI R, FLOHR P, MIRANDA S, ZAFEIRIOU Z, TUNARIU N, MATEO J, PEREZ-LOPEZ R, DEMICHELIS F, ROBINSON BD, SCHIFFMAN M, NANUS DM, TAGAWA ST, SIGARAS A, ENG KW, ELEMENTO O, SBONER A, HEATH EI, SCHER HI, PIENTA KJ, KANTOFF P, DE BONO JS, RUBIN MA, NELSON PS, GARRAWAY LA, SAWYERS CL & CHINNAIYAN AM 2015. Integrative clinical genomics of advanced prostate cancer. Cell, 161, 1215–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RODRIGUES LU, RIDER L, NIETO C, ROMERO L, KARIMPOUR-FARD A, LODA M, LUCIA MS, WU M, SHI L, CIMIC A, SIRINTRAPUN SJ, NOLLEY R, PAC C, CHEN H, PEEHL DM, XU J, LIU W, COSTELLO JC & CRAMER SD 2015. Coordinate loss of MAP3K7 and CHD1 promotes aggressive prostate cancer. Cancer Res, 75, 1021–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ROPERO S & ESTELLER M 2007. The role of histone deacetylases (HDACs) in human cancer. Mol Oncol, 1, 19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ROTILI D & MAI A 2011. Targeting Histone Demethylases: A New Avenue for the Fight against Cancer. Genes Cancer, 2, 663–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RUGGERO K, FARRAN-MATAS S, MARTINEZ-TEBAR A & AYTES A 2018. Epigenetic Regulation in Prostate Cancer Progression. Curr Mol Biol Rep, 4, 101–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SANDOVAL GJ, PULICE JL, PAKULA H, SCHENONE M, TAKEDA DY, POP M, BOULAY G, WILLIAMSON KE, MCBRIDE MJ, PAN J, ST PIERRE R, HARTMAN E, GARRAWAY LA, CARR SA, RIVERA MN, LI Z, RONCO L, HAHN WC & KADOCH C 2018. Binding of TMPRSS2-ERG to BAF Chromatin Remodeling Complexes Mediates Prostate Oncogenesis. Mol Cell, 71, 554–566 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SANTOS-ROSA H, SCHNEIDER R, BANNISTER AJ, SHERRIFF J, BERNSTEIN BE, EMRE NC, SCHREIBER SL, MELLOR J & KOUZARIDES T 2002. Active genes are tri-methylated at K4 of histone H3. Nature, 419, 407–11. [DOI] [PubMed] [Google Scholar]
- SARAC H, MOROVA T, PIRES E, MCCULLAGH J, KAPLAN A, CINGOZ A, BAGCI-ONDER T, ONDER T, KAWAMURA A & LACK NA 2020. Systematic characterization of chromatin modifying enzymes identifies KDM3B as a critical regulator in castration resistant prostate cancer. Oncogene, 39, 2187–2201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SASAKI M, TANAKA Y, PERINCHERY G, DHARIA A, KOTCHERGUINA I, FUJIMOTO S & DAHIYA R 2002. Methylation and inactivation of estrogen, progesterone, and androgen receptors in prostate cancer. J Natl Cancer Inst, 94, 384–90. [DOI] [PubMed] [Google Scholar]
- SCHMITGES FW, PRUSTY AB, FATY M, STUTZER A, LINGARAJU GM, AIWAZIAN J, SACK R, HESS D, LI L, ZHOU S, BUNKER RD, WIRTH U, BOUWMEESTER T, BAUER A, LY-HARTIG N, ZHAO K, CHAN H, GU J, GUT H, FISCHLE W, MULLER J & THOMA NH 2011. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell, 42, 330–41. [DOI] [PubMed] [Google Scholar]
- SEHRAWAT A, GAO L, WANG Y, BANKHEAD A 3RD, MCWEENEY SK, KING CJ, SCHWARTZMAN J, URRUTIA J, BISSON WH, COLEMAN DJ, JOSHI SK, KIM DH, SAMPSON DA, WEINMANN S, KALLAKURY BVS, BERRY DL, HAQUE R, VAN DEN EEDEN SK, SHARMA S, BEARSS J, BEER TM, THOMAS GV, HEISER LM & ALUMKAL JJ 2018. LSD1 activates a lethal prostate cancer gene network independently of its demethylase function. Proc Natl Acad Sci U S A, 115, E4179–E4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SETO E & YOSHIDA M 2014. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol, 6, a018713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SEVERSON TM, ZHU Y, PREKOVIC S, SCHUURMAN K, NGUYEN HM, BROWN LG, HAKKOLA S, KIM Y, KNEPPERS J, LINDER S, STELLOO S, LIEFTINK C, VAN DER HEIJDEN M, NYKTER M, VAN DER NOORT V, SANDERS J, MORRIS B, JENSTER G, VAN LEENDERS GJ, POMERANTZ M, FREEDMAN ML, BEIJERSBERGEN RL, URBANUCCI A, WESSELS L, COREY E, ZWART W & BERGMAN AM 2023. Enhancer profiling identifies epigenetic markers of endocrine resistance and reveals therapeutic options for metastatic castration-resistant prostate cancer patients. medRxiv. [Google Scholar]
- SHAH N, WANG P, WONGVIPAT J, KARTHAUS WR, ABIDA W, ARMENIA J, ROCKOWITZ S, DRIER Y, BERNSTEIN BE, LONG HW, FREEDMAN ML, ARORA VK, ZHENG D & SAWYERS CL 2017. Regulation of the glucocorticoid receptor via a BET-dependent enhancer drives antiandrogen resistance in prostate cancer. Elife, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SHAIN AH & POLLACK JR 2013. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS One, 8, e55119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SHARMA S, KELLY TK & JONES PA 2010. Epigenetics in cancer. Carcinogenesis, 31, 27–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SHEN SY, SINGHANIA R, FEHRINGER G, CHAKRAVARTHY A, ROEHRL MHA, CHADWICK D, ZUZARTE PC, BORGIDA A, WANG TT, LI T, KIS O, ZHAO Z, SPREAFICO A, MEDINA TDS, WANG Y, ROULOIS D, ETTAYEBI I, CHEN Z, CHOW S, MURPHY T, ARRUDA A, O’KANE GM, LIU J, MANSOUR M, MCPHERSON JD, O’BRIEN C, LEIGHL N, BEDARD PL, FLESHNER N, LIU G, MINDEN MD, GALLINGER S, GOLDENBERG A, PUGH TJ, HOFFMAN MM, BRATMAN SV, HUNG RJ & DE CARVALHO DD 2018. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature, 563, 579–583. [DOI] [PubMed] [Google Scholar]
- SHI J, WANG E, MILAZZO JP, WANG Z, KINNEY JB & VAKOC CR 2015. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat Biotechnol, 33, 661–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SHI Y, LAN F, MATSON C, MULLIGAN P, WHETSTINE JR, COLE PA, CASERO RA & SHI Y 2004. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell, 119, 941–53. [DOI] [PubMed] [Google Scholar]
- SHIN S & JANKNECHT R 2007. Activation of androgen receptor by histone demethylases JMJD2A and JMJD2D. Biochem Biophys Res Commun, 359, 742–6. [DOI] [PubMed] [Google Scholar]
- SHVEDUNOVA M & AKHTAR A 2022. Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol, 23, 329–349. [DOI] [PubMed] [Google Scholar]
- SJOSTROM M, ZHAO SG, LEVY S, ZHANG M, NING Y, SHRESTHA R, LUNDBERG A, HERBERTS C, FOYE A, AGGARWAL R, HUA JT, LI H, BERGAMASCHI A, MAURICE-DROR C, MAHESHWARI A, CHEN S, NG SWS, YE W, PETRICCA J, FRASER M, CHESNER L, PERRY MD, MORENO-RODRIGUEZ T, CHEN WS, ALUMKAL JJ, CHOU J, MORGANS AK, BEER TM, THOMAS GV, GLEAVE M, LLOYD P, PHILLIPS T, MCCARTHY E, HAFFNER MC, ZOUBEIDI A, ANNALA M, REITER RE, RETTIG MB, WITTE ON, FONG L, BOSE R, HUANG FW, LUO J, BJARTELL A, LANG JM, MAHAJAN NP, LARA PN, EVANS CP, TRAN PT, POSADAS EM, HE C, CUI XL, HUANG J, ZWART W, GILBERT LA, MAHER CA, BOUTROS PC, CHI KN, ASHWORTH A, SMALL EJ, HE HH, WYATT AW, QUIGLEY DA & FENG FY 2022. The 5-Hydroxymethylcytosine Landscape of Prostate Cancer. Cancer Res, 82, 3888–3902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SOLDI R, GHOSH HALDER T, WESTON A, THODE T, DRENNER K, LEWIS R, KAADIGE MR, SRIVASTAVA S, DANIEL AMPANATTU S, RODRIGUEZ DEL VILLAR R, LANG J, VANKAYALAPATI H, WEISSMAN B, TRENT JM, HENDRICKS WPD & SHARMA S 2020. The novel reversible LSD1 inhibitor SP-2577 promotes anti-tumor immunity in SWItch/Sucrose-NonFermentable (SWI/SNF) complex mutated ovarian cancer. PLoS One, 15, e0235705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- STATHIS A & BERTONI F 2018. BET Proteins as Targets for Anticancer Treatment. Cancer Discov, 8, 24–36. [DOI] [PubMed] [Google Scholar]
- STEIN J, MAJORES M, ROHDE M, LIM S, SCHNEIDER S, KRAPPE E, ELLINGER J, DIETEL M, STEPHAN C, JUNG K, PERNER S, KRISTIANSEN G & KIRFEL J 2014. KDM5C is overexpressed in prostate cancer and is a prognostic marker for prostate-specific antigen-relapse following radical prostatectomy. Am J Pathol, 184, 2430–7. [DOI] [PubMed] [Google Scholar]
- STREPKOS D, MARKOULI M, KLONOU A, PAPAVASSILIOU AG & PIPERI C 2021. Histone Methyltransferase SETDB1: A Common Denominator of Tumorigenesis with Therapeutic Potential. Cancer Res, 81, 525–534. [DOI] [PubMed] [Google Scholar]
- STRUHL K 1998. Histone acetylation and transcriptional regulatory mechanisms. Genes Dev, 12, 599–606. [DOI] [PubMed] [Google Scholar]
- SUN A, TAWFIK O, GAYED B, THRASHER JB, HOESTJE S, LI C & LI B 2007. Aberrant expression of SWI/SNF catalytic subunits BRG1/BRM is associated with tumor development and increased invasiveness in prostate cancers. Prostate, 67, 203–13. [DOI] [PubMed] [Google Scholar]
- SUN L, KOKURA K, IZUMI V, KOOMEN JM, SETO E, CHEN J & FANG J 2015. MPP8 and SIRT1 crosstalk in E-cadherin gene silencing and epithelial-mesenchymal transition. EMBO Rep, 16, 689–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SUN Y, WEI M, REN SC, CHEN R, XU WD, WANG FB, LU J, SHEN J, YU YW, HOU JG, XU CL, HUANG JT & SUN YH 2014. Histone methyltransferase SETDB1 is required for prostate cancer cell proliferation, migration and invasion. Asian J Androl, 16, 319–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SUNG H, FERLAY J, SIEGEL RL, LAVERSANNE M, SOERJOMATARAM I, JEMAL A & BRAY F 2021. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 71, 209–249. [DOI] [PubMed] [Google Scholar]
- TANG DE, DAI Y, HE JX, LIN LW, LENG QX, GENG XY, FU DX, JIANG HW & XU SH 2020. Targeting the KDM4B-AR-c-Myc axis promotes sensitivity to androgen receptor-targeted therapy in advanced prostate cancer. J Pathol, 252, 101–113. [DOI] [PubMed] [Google Scholar]
- TAYLOR BS, SCHULTZ N, HIERONYMUS H, GOPALAN A, XIAO Y, CARVER BS, ARORA VK, KAUSHIK P, CERAMI E, REVA B, ANTIPIN Y, MITSIADES N, LANDERS T, DOLGALEV I, MAJOR JE, WILSON M, SOCCI ND, LASH AE, HEGUY A, EASTHAM JA, SCHER HI, REUTER VE, SCARDINO PT, SANDER C, SAWYERS CL & GERALD WL 2010. Integrative genomic profiling of human prostate cancer. Cancer Cell, 18, 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- THOMPSON D, CHOO N, BOLTON DM, LAWRENTSCHUK N, RISBRIDGER GP, LAWRENCE MG & TAYLOR RA 2022. New approaches to targeting epigenetic regulation in prostate cancer. Curr Opin Urol, 32, 472–480. [DOI] [PubMed] [Google Scholar]
- TOKIZANE T, SHIINA H, IGAWA M, ENOKIDA H, URAKAMI S, KAWAKAMI T, OGISHIMA T, OKINO ST, LI LC, TANAKA Y, NONOMURA N, OKUYAMA A & DAHIYA R 2005. Cytochrome P450 1B1 is overexpressed and regulated by hypomethylation in prostate cancer. Clin Cancer Res, 11, 5793–801. [DOI] [PubMed] [Google Scholar]
- TOMLINS SA, RHODES DR, PERNER S, DHANASEKARAN SM, MEHRA R, SUN XW, VARAMBALLY S, CAO X, TCHINDA J, KUEFER R, LEE C, MONTIE JE, SHAH RB, PIENTA KJ, RUBIN MA & CHINNAIYAN AM 2005. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science, 310, 644–8. [DOI] [PubMed] [Google Scholar]
- TOPALIAN SL, HODI FS, BRAHMER JR, GETTINGER SN, SMITH DC, MCDERMOTT DF, POWDERLY JD, CARVAJAL RD, SOSMAN JA, ATKINS MB, LEMING PD, SPIGEL DR, ANTONIA SJ, HORN L, DRAKE CG, PARDOLL DM, CHEN L, SHARFMAN WH, ANDERS RA, TAUBE JM, MCMILLER TL, XU H, KORMAN AJ, JURE-KUNKEL M, AGRAWAL S, MCDONALD D, KOLLIA GD, GUPTA A, WIGGINTON JM & SZNOL M 2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med, 366, 2443–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- TOPCHU I, PANGENI RP, BYCHKOV I, MILLER SA, IZUMCHENKO E, YU J, GOLEMIS E, KARANICOLAS J & BOUMBER Y 2022. The role of NSD1, NSD2, and NSD3 histone methyltransferases in solid tumors. Cell Mol Life Sci, 79, 285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- TSUZUKI S, PARK SH, EBER MR, PETERS CM & SHIOZAWA Y 2016. Skeletal complications in cancer patients with bone metastases. Int J Urol, 23, 825–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- URBANUCCI A, BARFELD SJ, KYTOLA V, ITKONEN HM, COLEMAN IM, VODAK D, SJOBLOM L, SHENG X, TOLONEN T, MINNER S, BURDELSKI C, KIVINUMMI KK, KOHVAKKA A, KREGEL S, TAKHAR M, ALSHALALFA M, DAVICIONI E, ERHO N, LLOYD P, KARNES RJ, ROSS AE, SCHAEFFER EM, VANDER GRIEND DJ, KNAPP S, COREY E, FENG FY, NELSON PS, SAATCIOGLU F, KNUDSEN KE, TAMMELA TLJ, SAUTER G, SCHLOMM T, NYKTER M, VISAKORPI T & MILLS IG 2017. Androgen Receptor Deregulation Drives Bromodomain-Mediated Chromatin Alterations in Prostate Cancer. Cell Rep, 19, 2045–2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- URBANUCCI A & MILLS IG 2018. Bromodomain-containing proteins in prostate cancer. Mol Cell Endocrinol, 462, 31–40. [DOI] [PubMed] [Google Scholar]
- VALDES-MORA F, GOULD CM, COLINO-SANGUINO Y, QU W, SONG JZ, TAYLOR KM, BUSKE FA, STATHAM AL, NAIR SS, ARMSTRONG NJ, KENCH JG, LEE KML, HORVATH LG, QIU M, ILINYKH A, YEO-TEH NS, GALLEGO-ORTEGA D, STIRZAKER C & CLARK SJ 2017. Acetylated histone variant H2A.Z is involved in the activation of neo-enhancers in prostate cancer. Nat Commun, 8, 1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- VALDEZ CD, KUNJU L, DAIGNAULT S, WOJNO KJ & DAY ML 2013. The E2F1/DNMT1 axis is associated with the development of AR negative castration resistant prostate cancer. Prostate, 73, 1776–85. [DOI] [PubMed] [Google Scholar]
- VANDEKERKHOVE G, STRUSS WJ, ANNALA M, KALLIO HML, KHALAF D, WARNER EW, HERBERTS C, RITCH E, BEJA K, LOKTIONOVA Y, HURTADO-COLL A, FAZLI L, SO A, BLACK PC, NYKTER M, TAMMELA T, CHI KN, GLEAVE ME & WYATT AW 2019. Circulating Tumor DNA Abundance and Potential Utility in De Novo Metastatic Prostate Cancer. Eur Urol, 75, 667–675. [DOI] [PubMed] [Google Scholar]
- VARAMBALLY S, CAO Q, MANI RS, SHANKAR S, WANG X, ATEEQ B, LAXMAN B, CAO X, JING X, RAMNARAYANAN K, BRENNER JC, YU J, KIM JH, HAN B, TAN P, KUMAR-SINHA C, LONIGRO RJ, PALANISAMY N, MAHER CA & CHINNAIYAN AM 2008. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science, 322, 1695–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- VARAMBALLY S, DHANASEKARAN SM, ZHOU M, BARRETTE TR, KUMAR-SINHA C, SANDA MG, GHOSH D, PIENTA KJ, SEWALT RG, OTTE AP, RUBIN MA & CHINNAIYAN AM 2002. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature, 419, 624–9. [DOI] [PubMed] [Google Scholar]
- VATAPALLI R, SAGAR V, RODRIGUEZ Y, ZHAO JC, UNNO K, PAMARTHY S, LYSY B, ANKER J, HAN H, YOO YA, TRUICA M, CHALMERS ZR, GILES F, YU J, CHAKRAVARTI D, CARNEIRO B & ABDULKADIR SA 2020. Histone methyltransferase DOT1L coordinates AR and MYC stability in prostate cancer. Nat Commun, 11, 4153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- VIEIRA FQ, COSTA-PINHEIRO P, RAMALHO-CARVALHO J, PEREIRA A, MENEZES FD, ANTUNES L, CARNEIRO I, OLIVEIRA J, HENRIQUE R & JERONIMO C 2014. Deregulated expression of selected histone methylases and demethylases in prostate carcinoma. Endocr Relat Cancer, 21, 51–61. [DOI] [PubMed] [Google Scholar]
- WANG J, PARK KS, YU X, GONG W, EARP HS, WANG GG, JIN J & CAI L 2022a. A cryptic transactivation domain of EZH2 binds AR and AR’s splice variant, promoting oncogene activation and tumorous transformation. Nucleic Acids Res. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG J & WANG GG 2020. No Easy Way Out for EZH2: Its Pleiotropic, Noncanonical Effects on Gene Regulation and Cellular Function. Int J Mol Sci, 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG J, YU X, GONG W, LIU X, PARK KS, MA A, TSAI YH, SHEN Y, ONIKUBO T, PI WC, ALLISON DF, LIU J, CHEN WY, CAI L, ROEDER RG, JIN J & WANG GG 2022b. EZH2 noncanonically binds cMyc and p300 through a cryptic transactivation domain to mediate gene activation and promote oncogenesis. Nat Cell Biol, 24, 384–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG LY, HUNG CL, CHEN YR, YANG JC, WANG J, CAMPBELL M, IZUMIYA Y, CHEN HW, WANG WC, ANN DK & KUNG HJ 2016a. KDM4A Coactivates E2F1 to Regulate the PDK-Dependent Metabolic Switch between Mitochondrial Oxidation and Glycolysis. Cell Rep, 16, 3016–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG Q, WILLIAMSON M, BOTT S, BROOKMAN-AMISSAH N, FREEMAN A, NARICULAM J, HUBANK MJ, AHMED A & MASTERS JR 2007. Hypomethylation of WNT5A, CRIP1 and S100P in prostate cancer. Oncogene, 26, 6560–5. [DOI] [PubMed] [Google Scholar]
- WANG X, YEH S, WU G, HSU CL, WANG L, CHIANG T, YANG Y, GUO Y & CHANG C 2001. Identification and characterization of a novel androgen receptor coregulator ARA267-alpha in prostate cancer cells. J Biol Chem, 276, 40417–23. [DOI] [PubMed] [Google Scholar]
- WANG Y, JADHAV RR, LIU J, WILSON D, CHEN Y, THOMPSON IM, TROYER DA, HERNANDEZ J, SHI H, LEACH RJ, HUANG TH & JIN VX 2016b. Roles of Distal and Genic Methylation in the Development of Prostate Tumorigenesis Revealed by Genome-wide DNA Methylation Analysis. Sci Rep, 6, 22051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANT MY, TSUJI T, SINGH PK, THORNE JL, MATSUZAKI J, KARASIK E, GILLARD B, CORTES GOMEZ E, KOYA RC, LUGADE A, ODUNSI K & BATTAGLIA S 2021. WHSC1/NSD2 regulates immune infiltration in prostate cancer. J Immunother Cancer, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WATSON PA, ARORA VK & SAWYERS CL 2015. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer, 15, 701–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WEICHERT W, ROSKE A, GEKELER V, BECKERS T, STEPHAN C, JUNG K, FRITZSCHE FR, NIESPOREK S, DENKERT C, DIETEL M & KRISTIANSEN G 2008. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer, 98, 604–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WELSBIE DS, XU J, CHEN Y, BORSU L, SCHER HI, ROSEN N & SAWYERS CL 2009. Histone deacetylases are required for androgen receptor function in hormone-sensitive and castrate-resistant prostate cancer. Cancer Res, 69, 958–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WELTI J, SHARP A, BROOKS N, YUAN W, MCNAIR C, CHAND SN, PAL A, FIGUEIREDO I, RIISNAES R, GUREL B, REKOWSKI J, BOGDAN D, WEST W, YOUNG B, RAJA M, PROSSER A, LANE J, THOMSON S, WORTHINGTON J, ONIONS S, SHANNON J, PAOLETTA S, BROWN R, SMYTH D, HARBOTTLE GW, GIL VS, MIRANDA S, CRESPO M, FERREIRA A, PEREIRA R, TUNARIU N, CARREIRA S, NEEB AJ, NING J, SWAIN A, TADDEI D, TEAM SCPIPCD, SCHIEWER MJ, KNUDSEN KE, PEGG N & DE BONO JS 2021. Targeting the p300/CBP Axis in Lethal Prostate Cancer. Cancer Discov, 11, 1118–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WELTI J, SHARP A, YUAN W, DOLLING D, NAVA RODRIGUES D, FIGUEIREDO I, GIL V, NEEB A, CLARKE M, SEED G, CRESPO M, SUMANASURIYA S, NING J, KNIGHT E, FRANCIS JC, HUGHES A, HALSEY WS, PASCHALIS A, MANI RS, RAJ GV, PLYMATE SR, CARREIRA S, BOYSEN G, CHINNAIYAN AM, SWAIN A, DE BONO JS & INTERNATIONAL SUCPCFPCDT 2018. Targeting Bromodomain and Extra-Terminal (BET) Family Proteins in Castration-Resistant Prostate Cancer (CRPC). Clin Cancer Res, 24, 3149–3162. [DOI] [PubMed] [Google Scholar]
- WHETSTINE JR, NOTTKE A, LAN F, HUARTE M, SMOLIKOV S, CHEN Z, SPOONER E, LI E, ZHANG G, COLAIACOVO M & SHI Y 2006. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell, 125, 467–81. [DOI] [PubMed] [Google Scholar]
- WHYTE WA, ORLANDO DA, HNISZ D, ABRAHAM BJ, LIN CY, KAGEY MH, RAHL PB, LEE TI & YOUNG RA 2013. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell, 153, 307–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WISSMANN M, YIN N, MULLER JM, GRESCHIK H, FODOR BD, JENUWEIN T, VOGLER C, SCHNEIDER R, GUNTHER T, BUETTNER R, METZGER E & SCHULE R 2007. Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat Cell Biol, 9, 347–53. [DOI] [PubMed] [Google Scholar]
- WU A, CREMASCHI P, WETTERSKOG D, CONTEDUCA V, FRANCESCHINI GM, KLEFTOGIANNIS D, JAYARAM A, SANDHU S, WONG SQ, BENELLI M, SALVI S, GURIOLI G, FEBER A, PEREIRA MB, WINGATE AM, GONZALEZ-BILLALEBEITIA E, DE GIORGI U, DEMICHELIS F, LISE S & ATTARD G 2020. Genome-wide plasma DNA methylation features of metastatic prostate cancer. J Clin Invest, 130, 1991–2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WU MJ, CHEN CJ, LIN TY, LIU YY, TSENG LL, CHENG ML, CHUU CP, TSAI HK, KUO WL, KUNG HJ & WANG WC 2021. Targeting KDM4B that coactivates c-Myc-regulated metabolism to suppress tumor growth in castration-resistant prostate cancer. Theranostics, 11, 7779–7796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WU Y, SARKISSYAN M & VADGAMA JV 2015. Epigenetics in breast and prostate cancer. Methods Mol Biol, 1238, 425–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XIANG Y, ZHU Z, HAN G, LIN H, XU L & CHEN CD 2007a. JMJD3 is a histone H3K27 demethylase. Cell Res, 17, 850–7. [DOI] [PubMed] [Google Scholar]
- XIANG Y, ZHU Z, HAN G, YE X, XU B, PENG Z, MA Y, YU Y, LIN H, CHEN AP & CHEN CD 2007b. JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc Natl Acad Sci U S A, 104, 19226–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XIAO L, PAROLIA A, QIAO Y, BAWA P, EYUNNI S, MANNAN R, CARSON SE, CHANG Y, WANG X, ZHANG Y, VO JN, KREGEL S, SIMKO SA, DELEKTA AD, JABER M, ZHENG H, APEL IJ, MCMURRY L, SU F, WANG R, ZELENKA-WANG S, SASMAL S, KHARE L, MUKHERJEE S, ABBINENI C, AITHAL K, BHAKTA MS, GHURYE J, CAO X, NAVONE NM, NESVIZHSKII AI, MEHRA R, VAISHAMPAYAN U, BLANCHETTE M, WANG Y, SAMAJDAR S, RAMACHANDRA M & CHINNAIYAN AM 2022. Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer. Nature, 601, 434–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XIAO L, TIEN JC, VO J, TAN M, PAROLIA A, ZHANG Y, WANG L, QIAO Y, SHUKLA S, WANG X, ZHENG H, SU F, JING X, LUO E, DELEKTA A, JUCKETTE KM, XU A, CAO X, ALVA AS, KIM Y, MACLEOD AR & CHINNAIYAN AM 2018. Epigenetic Reprogramming with Antisense Oligonucleotides Enhances the Effectiveness of Androgen Receptor Inhibition in Castration-Resistant Prostate Cancer. Cancer Res, 78, 5731–5740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XU K, WU ZJ, GRONER AC, HE HH, CAI C, LIS RT, WU X, STACK EC, LODA M, LIU T, XU H, CATO L, THORNTON JE, GREGORY RI, MORRISSEY C, VESSELLA RL, MONTIRONI R, MAGI-GALLUZZI C, KANTOFF PW, BALK SP, LIU XS & BROWN M 2012. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science, 338, 1465–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XU S, FAN L, JEON HY, ZHANG F, CUI X, MICKLE MB, PENG G, HUSSAIN A, FAZLI L, GLEAVE ME, DONG X & QI J 2020. p300-Mediated Acetylation of Histone Demethylase JMJD1A Prevents Its Degradation by Ubiquitin Ligase STUB1 and Enhances Its Activity in Prostate Cancer. Cancer Res, 80, 3074–3087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- XU Y, WU F, TAN L, KONG L, XIONG L, DENG J, BARBERA AJ, ZHENG L, ZHANG H, HUANG S, MIN J, NICHOLSON T, CHEN T, XU G, SHI Y, ZHANG K & SHI YG 2011. Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol Cell, 42, 451–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YANG L, JIN M, PARK SJ, SEO SY & JEONG KW 2020. SETD1A Promotes Proliferation of Castration-Resistant Prostate Cancer Cells via FOXM1 Transcription. Cancers (Basel), 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YANG M, CULHANE JC, SZEWCZUK LM, JALILI P, BALL HL, MACHIUS M, COLE PA & YU H 2007. Structural basis for the inhibition of the LSD1 histone demethylase by the antidepressant trans-2-phenylcyclopropylamine. Biochemistry, 46, 8058–65. [DOI] [PubMed] [Google Scholar]
- YANG P, GUO L, DUAN ZJ, TEPPER CG, XUE L, CHEN X, KUNG HJ, GAO AC, ZOU JX & CHEN HW 2012. Histone methyltransferase NSD2/MMSET mediates constitutive NF-kappaB signaling for cancer cell proliferation, survival, and tumor growth via a feed-forward loop. Mol Cell Biol, 32, 3121–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YEGNASUBRAMANIAN S, KOWALSKI J, GONZALGO ML, ZAHURAK M, PIANTADOSI S, WALSH PC, BOVA GS, DE MARZO AM, ISAACS WB & NELSON WG 2004. Hypermethylation of CpG islands in primary and metastatic human prostate cancer. Cancer Res, 64, 1975–86. [DOI] [PubMed] [Google Scholar]
- YI Y, LI Y, MENG Q, LI Q, LI F, LU B, SHEN J, FAZLI L, ZHAO D, LI C, JIANG W, WANG R, LIU Q, SZCZEPANSKI A, LI Q, QIN W, WEINER AB, LOTAN TL, JI Z, KALANTRY S, WANG L, SCHAEFFER EM, NIU H, DONG X, ZHAO W, CHEN K & CAO Q 2021. A PRC2-independent function for EZH2 in regulating rRNA 2’-O methylation and IRES-dependent translation. Nat Cell Biol, 23, 341–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YOSHIHAMA Y, LABELLA KA, KIM E, BERTOLET L, COLIC M, LI J, SHANG X, WU CJ, SPRING DJ, WANG YA, HART T & DEPINHO RA 2021. AR-negative prostate cancer is vulnerable to loss of JMJD1C demethylase. Proc Natl Acad Sci U S A, 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YOU JS & JONES PA 2012. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell, 22, 9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YUAN G, FLORES NM, HAUSMANN S, LOFGREN SM, KHARCHENKO V, ANGULO-IBANEZ M, SENGUPTA D, LU X, CZABAN I, AZHIBEK D, VICENT S, FISCHLE W, JAREMKO M, FANG B, WISTUBA II, CHUA KF, ROTH JA, MINNA JD, SHAO NY, JAREMKO L, MAZUR PK & GOZANI O 2021. Elevated NSD3 histone methylation activity drives squamous cell lung cancer. Nature, 590, 504–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YUAN H, HAN Y, WANG X, LI N, LIU Q, YIN Y, WANG H, PAN L, LI L, SONG K, QIU T, PAN Q, CHEN Q, ZHANG G, ZANG Y, TAN M, ZHANG J, LI Q, WANG X, JIANG J & QIN J 2020a. SETD2 Restricts Prostate Cancer Metastasis by Integrating EZH2 and AMPK Signaling Pathways. Cancer Cell, 38, 350–365 e7. [DOI] [PubMed] [Google Scholar]
- YUAN S, NATESAN R, SANCHEZ-RIVERA FJ, LI J, BHANU NV, YAMAZOE T, LIN JH, MERRELL AJ, SELA Y, THOMAS SK, JIANG Y, PLESSET JB, MILLER EM, SHI J, GARCIA BA, LOWE SW, ASANGANI IA & STANGER BZ 2020b. Global Regulation of the Histone Mark H3K36me2 Underlies Epithelial Plasticity and Metastatic Progression. Cancer Discov, 10, 854–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YUAN W, XU M, HUANG C, LIU N, CHEN S & ZHU B 2011. H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J Biol Chem, 286, 7983–7989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZACHAROPOULOU N, KALLERGI G, ALKAHTANI S, TSAPARA A, ALARIFI S, SCHMID E, SUKKAR B, KAMPRANIS S, LANG F & STOURNARAS C 2020. The histone demethylase KDM2B activates FAK and PI3K that control tumor cell motility. Cancer Biol Ther, 21, 533–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZELIC R, FIANO V, GRASSO C, ZUGNA D, PETTERSSON A, GILLIO-TOS A, MERLETTI F & RICHIARDI L 2015. Global DNA hypomethylation in prostate cancer development and progression: a systematic review. Prostate Cancer Prostatic Dis, 18, 1–12. [DOI] [PubMed] [Google Scholar]
- ZHANG P, WANG D, ZHAO Y, REN S, GAO K, YE Z, WANG S, PAN CW, ZHU Y, YAN Y, YANG Y, WU D, HE Y, ZHANG J, LU D, LIU X, YU L, ZHAO S, LI Y, LIN D, WANG Y, WANG L, CHEN Y, SUN Y, WANG C & HUANG H 2017. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med, 23, 1055–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHANG W, JIAO H, ZHANG X, ZHAO R, WANG F, HE W, ZONG H, FAN Q & WANG L 2015. Correlation between the expression of DNMT1, and GSTP1 and APC, and the methylation status of GSTP1 and APC in association with their clinical significance in prostate cancer. Mol Med Rep, 12, 141–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHANG W & XU J 2017. DNA methyltransferases and their roles in tumorigenesis. Biomark Res, 5, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHANG Y, ZHENG D, ZHOU T, SONG H, HULSURKAR M, SU N, LIU Y, WANG Z, SHAO L, ITTMANN M, GLEAVE M, HAN H, XU F, LIAO W, WANG H & LI W 2018. Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers. Nat Commun, 9, 4080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHANG Z, ZHOU C, LI X, BARNES SD, DENG S, HOOVER E, CHEN CC, LEE YS, ZHANG Y, WANG C, METANG LA, WU C, TIRADO CR, JOHNSON NA, WONGVIPAT J, NAVRAZHINA K, CAO Z, CHOI D, HUANG CH, LINTON E, CHEN X, LIANG Y, MASON CE, DE STANCHINA E, ABIDA W, LUJAMBIO A, LI S, LOWE SW, MENDELL JT, MALLADI VS, SAWYERS CL & MU P 2020. Loss of CHD1 Promotes Heterogeneous Mechanisms of Resistance to AR-Targeted Therapy via Chromatin Dysregulation. Cancer Cell, 37, 584–598 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHAO S, ALLIS CD & WANG GG 2021. The language of chromatin modification in human cancers. Nat Rev Cancer, 21, 413–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHAO SG, CHEN WS, LI H, FOYE A, ZHANG M, SJOSTROM M, AGGARWAL R, PLAYDLE D, LIAO A, ALUMKAL JJ, DAS R, CHOU J, HUA JT, BARNARD TJ, BAILEY AM, CHOW ED, PERRY MD, DANG HX, YANG R, MOUSSAVI-BAYGI R, ZHANG L, ALSHALALFA M, LAURA CHANG S, HOULAHAN KE, SHIAH YJ, BEER TM, THOMAS G, CHI KN, GLEAVE M, ZOUBEIDI A, REITER RE, RETTIG MB, WITTE O, YVONNE KIM M, FONG L, SPRATT DE, MORGAN TM, BOSE R, HUANG FW, LI H, CHESNER L, SHENOY T, GOODARZI H, ASANGANI IA, SANDHU S, LANG JM, MAHAJAN NP, LARA PN, EVANS CP, FEBBO P, BATZOGLOU S, KNUDSEN KE, HE HH, HUANG J, ZWART W, COSTELLO JF, LUO J, TOMLINS SA, WYATT AW, DEHM SM, ASHWORTH A, GILBERT LA, BOUTROS PC, FARH K, CHINNAIYAN AM, MAHER CA, SMALL EJ, QUIGLEY DA & FENG FY 2020. The DNA methylation landscape of advanced prostate cancer. Nat Genet, 52, 778–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHAO Y & GARCIA BA 2015. Comprehensive Catalog of Currently Documented Histone Modifications. Cold Spring Harb Perspect Biol, 7, a025064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHOU B, HU J, XU F, CHEN Z, BAI L, FERNANDEZ-SALAS E, LIN M, LIU L, YANG CY, ZHAO Y, MCEACHERN D, PRZYBRANOWSKI S, WEN B, SUN D & WANG S 2018. Discovery of a Small-Molecule Degrader of Bromodomain and Extra-Terminal (BET) Proteins with Picomolar Cellular Potencies and Capable of Achieving Tumor Regression. J Med Chem, 61, 462–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHU H, WANG G & QIAN J 2016. Transcription factors as readers and effectors of DNA methylation. Nat Rev Genet, 17, 551–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHU Y, ZHAO Y, WEN J, LIU S, HUANG T, HATIAL I, PENG X, AL JANABI H, HUANG G, MITTLESTEADT J, CHENG M, BHARDWAJ A, ASHFELD BL, KAO KR, MAEDA DY, DAI X, WIEST O, BLAGG BSJ, LU X, CHENG L, WAN J & LU X 2023. Targeting the chromatin effector Pygo2 promotes cytotoxic T cell responses and overcomes immunotherapy resistance in prostate cancer. Sci Immunol, 8, eade4656. [DOI] [PMC free article] [PubMed] [Google Scholar]