Cancer Treatment Reviews xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Cancer Treatment Reviews journal homepage: www.elsevierhealth.com/journals/ctrv
Tumour Review
The hallmarks of castration-resistant prostate cancers Maria Katsogiannou ⇑,1, Hajer Ziouziou 1, Sara Karaki, Claudia Andrieu, Marie Henry de Villeneuve, Palma Rocchi ⇑ Inserm, UMR1068, CRCM, Marseille F-13009, France Institut Paoli-Calmettes, Marseille F-13009, France Aix-Marseille Université, F-13284 Marseille, France CNRS, UMR7258, CRCM, Marseille F-13009, France
a r t i c l e
i n f o
Article history: Received 11 February 2015 Received in revised form 30 April 2015 Accepted 4 May 2015 Available online xxxx Keywords: Castration resistant mechanisms Prostate cancer Hallmarks Androgen receptor
a b s t r a c t Prostate cancer has become a real public health issue in industrialized countries, mainly due to patients’ relapse by castration-refractory disease after androgen ablation. Castration-resistant prostate cancer is an incurable and highly aggressive terminal stage of prostate cancer, seriously jeopardizing the patient’s quality of life and lifespan. The management of castration-resistant prostate cancer is complex and has opened new fields of research during the last decade leading to an improved understanding of the biology of the disease and the development of new therapies. Most advanced tumors resistant to therapy still maintain the androgen receptor-pathway, which plays a central role for survival and growth of most castration-resistant prostate cancers. Many mechanisms induce the emergence of the castration resistant phenotype through this pathway. However some non-related AR pathways like neuroendocrine cells or overexpression of anti-apoptotic proteins like Hsp27 are described to be involved in CRPC progression. More recently, loss of expression of tumor suppressor gene, post-transcriptional modification using miRNA, epigenetic alterations, alternatif splicing and gene fusion became also hallmarks of castration-resistant prostate cancer. This review presents an up-to-date overview of the androgen receptor-related mechanisms as well as the latest evidence of the non-AR-related mechanisms underlying castration-resistant prostate cancer progression. Ó 2015 Elsevier Ltd. All rights reserved.
Introduction Prostate cancer (PC) is the second leading cause of cancer-related death in men in the western world after lung cancer [1,2]. According to the American Cancer Society estimates in 2013, over 230,000 American men will be diagnosed with prostate cancer and nearly 29,720 will die of the disease [2]. PC progresses from diagnosis to death through a series of clinical states characterized by the extent of the disease, the hormonal status (castrate or non-castrate) and the presence or absence of metastases. Before choosing a PC treatment, some parameters are considered for each patient such as age, health, stage and grade of the disease, PSA level. . .etc. Treatments can be applied as mono- or combined ⇑ Corresponding authors at: Aix-Marseille Université, U105, F-13284 Marseille, France. Tel.: +33 486 977 266; fax: +33 486 977 499 (M. Katsogiannou). Tel.: +33 486977267; fax: +33 486977499 (P. Rocchi). E-mail addresses:
[email protected] (M. Katsogiannou), palma.
[email protected] (P. Rocchi). 1 Both authors contributed equally to this work.
therapies. Common options for PC treatments include: active surveillance, surgery (prostatectomy), radiation therapy, chemotherapy or hormone-therapy (androgen deprivation therapy ADT). Active surveillance is often used when an early stage, slow-growing PC is found in older men. It may also be suggested when the risks of surgery, radiation therapy, or hormonal-therapy outweigh the possible benefits. Most patients initially respond well to androgen deprivation (ADT) leading to disease regression. Unfortunately, PC will ultimately become unresponsive and recur within 1–3 years after ADT as a castration-resistant prostate cancer (CRPC) [3]. CRPC is an incurable and highly aggressive terminal stage of PC, seriously jeopardizing the patient’s quality of life and lifespan. The management of CRPC is complex and has opened new fields of research during the last decade leading to an improved understanding of the biology of the disease and the development of new therapies [4–7]. This review presents an up-to-date overview of the androgen receptor (AR)-related mechanisms as well as the latest evidence of the non-AR-related mechanisms underlying CRPC progression.
http://dx.doi.org/10.1016/j.ctrv.2015.05.003 0305-7372/Ó 2015 Elsevier Ltd. All rights reserved.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
2
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
From castration-sensitivity to castration-resistance Growth of the prostate gland is initially dependent on androgens. Under constant stimulation by androgens, the prostate gland gradually develops into PC, which is the rationale for androgen deprivation therapy (ADT). Tumors that relapse CT were first thought to be ‘‘an drogen-independent’’, but very low levels of androgens may still be detectable in the tissues and serum of advanced patients [8,9]. ADT provides selective pressure leading to outgrowths of CR cancers [10]. Most advanced tumors resistant to CT still maintain expression of a functional AR showing that the AR pathway plays a central role for survival and growth of most CRPC [11–15] and constitutes an attractive target for therapy. Hence, how AR promotes cell proliferation and tumor growth is an extensive area of research and several mechanisms induce the emergence of the castration resistant phenotype [16]. In addition to AR-related-pathways, more and more non-AR-related pathways like anti-apoptotic proteins overexpression, mRNA splicing events, gene fusions loss of expression of tumor suppressor gene, post-transcriptional modification using miRNA, epigenetic alterations, have also become the new hallmarks of CRPC. PC growth and progression are driven by the accumulation of genetic and epigenetic alterations, events that will be reviewed here. Discovery and understanding of these mechanisms has led to the development of new generation of therapies for the treatment of CRPC [17]. Thus, 6 novel therapies have been approved by the Food and Drug Administration (FDA) for the treatment of CRPC in just the last few years (sipuleucel-T, cabazitaxel, denosumab, abiraterone, enzalutamide and Ra-223) and a few others demonstrating promising results in late-phase clinical trials (for review [4,6,7]). AR-related pathways Hypersensitive pathway AR could become sensitive to low amounts of residual androgens through increased protein production. This theory of a hypersensitive AR pathway (Fig. 1, pathway A) is supported by the fact that many CR tumors show overproduction of AR resulting of a selective outgrowth following death of cells during CT [13,18,19]. Excess of AR production could result from AR locus amplification, increased mRNA transcription rates and/or stabilization of the mRNA or protein [20,21]. Regardless of the mechanism, AR overproduction is expected to contribute to PC growth by compensating for low androgens levels after ADT. Among latest therapeutic strategies developed and FDA-approved, Enzalutamide is a novel antiandrogen selected for novel clinical development with promising results [22], presenting great affinity for the AR, lacks agonists effects and inhibits not only ligand binding to the receptor in a competitive manner but also AR nuclear translocation and DNA fixation. Clinical results showed a slowing down in cancer cell growth, induction of cancer cell apoptosis and tumor regression [23]. Intracrine androgen metabolism PC cells may survive ADT by regulating intracrine androgen synthesis within the prostate (Fig. 1, pathway B). This local synthesis of androgens is due to increased testosterone conversion to DHT resulting from overproduction of the 5a-reductase enzyme [24,25]. Increased 5a-reductase enzyme level results itself from a germline variant substituting valine to leucine at codon 89 [26]. This variant is commonly observed in African men, indicating a genetic influence in PC [13,19,27]. Intraprostatic androgens can also be synthesized from cholesterol or other precursors such as DHEA (dehydroepiandrosterone). DHEA could be converted to androstenedione, a substrate for conversion to testosterone. The expression of all the genes necessary for synthesizing androgens
are described to be increased in CRPC compared to early PC analyzed from untreated patients [9,25,28,29]. Among the recently developed drugs, abiraterone acetate is an inhibitor of CYP17A, an enzyme involved in the intratumoral androgen biosynthesis. Abiraterone has been shown to be effective and very potent and represents the first hormonal therapy to be approved in the post-chemotherapy setting for CRPC [4,6]. Promiscuous pathway The promiscuous pathway results in the AR being receptive to ligands other than DHT (Fig. 1, pathway C/D). AR mutations enhance AR ligand binding specificity [30–32] and its activation by weak adrenal androgens and other steroid hormones including DHEA, progesterone, estrogens and cortisol [33–37]. AR substitutions are referenced in a database (http://androgendb.mcgill.ca/). These mutations have various consequences depending on their localization [33,38,39]. For instance, cells from the LNCaP (castration-sensitive) cell line display an AR missense mutation in the ligand-binding domain. At codon 877, the threonine is substituted to alanine, which opens the AR sensitivity to a wide range of steroid ligands [40,41]. This mutation may also convert AR antagonists (flutamide and bicalutamide) to AR agonists. AR antagonist treatments may in fact select tumors expressing AR mutants activated by the therapeutic agents [42]. Other mutations, which occur in the AR DNA-binding domain or in the N-terminal domain, modulate the binding specificity of its co-regulators and the transcriptional activation of its target genes [43–45]. Interestingly, two mutations (T877A and Q640X) constitutively activate AR [46]. Finally, AR splice variants (AR3, 4 and 5), which have lost their C-terminal protein domain (ligand-binding domain) including the canonical nuclear localization signal (NLS), can activate AR target genes [47–51]. Mutants truncated in the N-terminal domain (DNA-binding domain) are still able to translocate into the nucleus and have ligand-independent transcriptional activity. Importantly, the expression of certain AR variants such as AR-V7 is associated with a short time to disease recurrence following radical prostatectomy [48,50,52]. These AR variants are key mediators of persistent AR signaling and androgen withdrawal therapies [53]. They represent a clinical challenge depending on their sensitivity to AR antagonists designed to target the AR ligand-binding domain. A small molecule (EPI-001) targeting the AR N-terminal domain has been recently designed and its efficiency against tumors expressing AR splice variants is under study [54]. Numerous pre-clinical studies show the efficiency of AR-siRNA strategy in inducing CR tumor growth inhibition and regression [55,56]. Outlaw pathway The outlaw pathway (Fig. 1, pathway E) is functional when the AR pathway is activated by growth factors, cytokines and receptor tyrosine kinases. Growth factor pathways such as IGF1 (insulin-like growth factor) and EGF (epidermal growth factor) can bind and activate AR in the castrated state, when they are overexpressed [57]. IGF1 has been also described to be involved in CR evolution in an AR-independent fashion. IGF1 promotes cell growth, survival and differentiation of prostate cells and is increased in advanced cancers especially in metastases [58]. IGF1 can activate powerful oncogenic signaling pathways such as RAS, RAF, MAPK, or PKC leading to the transcription of target genes that promote cell growth and survival [59]. Cytokines like interleukin-6 (IL6) and interleukin-4 (IL4) are also activators of the AR pathway in CRPC [60,61]. Notably, IL6 could activate AR through the activation of MAPK (mitogen-activated protein kinase) and STAT3 pathways [62]. Moreover, tyrosine kinase receptors such as ERBB2 are overexpressed in CRPC, resulting in the increase of AR expression and activity via the activation of
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
3
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
A. Amplification and hypersensibility of AR Low levels of T
C. Mutations of AR Steroïdes non A: Cortison, Oestradiol…
T
T
Prostate cell cytoplasm
5α-reductase B. Local production of A DHT AR
AR
AR
AR
AR
AR
exon1
intron
exon2
AR AR
ERG
exon2
Exon 3
Proteolysis by Calpain
AR
AR
AR
intron exon1
Exon 3
AR
AR
Transcription of target genes
L. Alternative splicing survival PSA DNA K. Epigenetic alterations Biological responses ARNm miRNAs apoptosis J. Post-transcriptionnal Regulation by miRNAs BAD P53, RB1
proliferation
Bcl-2 TP53INP1
nucleus
AR
AR
ProCaspase 9
Inactive P PTEN
AR
AR
AR
F. Alteration of co-regulator expression
AR
Exon 3
Akt
MAPK
AR
M. Gene fusions exon1
RTK
D. Variants and truncated forms of AR
AR
AR
AR
TMPRRS2
E. Activation of AR Gowth factors by other signaling and cytokines pathways
Akt
Hsp27 Clusterin
Protein degradation
NPs secretion
H. Anti-apoptotic proteins overexpression
I. Tumor suppessor deregulation Cell stress (HT..)
G. Neuroendocrine cells
Fig. 1. Androgen receptor (AR)-related and alternative pathways contributing to castration-resistant emergence and progression on prostate cancer.
MAPK and phosphoinositide 3-kinase (PI3K)/AKT signaling pathways [63–66]. The PI3K/AKT pathway seems required for AR protein expression because AKT mutant or PI3K disruption reduce AR protein level [65]. These kinase pathways enhance AR activity indirectly by mediating the phosphorylation of co-activators rather than directly mediating the phosphorylation of AR itself [67]. Finally, a cross-talk between NF-jB and AR indicates the role of the NF-jB signaling pathway in CR progression [68]. Phase II clinical trials of mTOR inhibitors (ridaforolimus, temsirolimus and everolimus), targeting the PI3K pathway in CRPC have not been overwhelming [69– 71]. Nevertheless, combinatory trials of PI3K and AR inhibition have
shown more promising results but may encounter some toxicity issues [72]. Alterations in co-factors recruitment Co-activator levels are increased in CR cancers enhancing the sensitivity of AR to various ligands other than androgens (Fig. 1, pathway F). Co-activators such as ARA70, ARA55, SRC-1, P/CAF and GRIP1/TIF2 are overexpressed in PC [13,73–77]. Additionally, loss of AR co-repressor function may promote AR transcription activity in CRPC such as the exclusion of NCoR from the AR
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
4
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
complex. Recently, nucleophosmin (NPM/B23), a member of the histone chaperone family, has also been identified as an AR co-factor. NPM binds to AR and enhances AR binding to its binding sites (AREs) [78]. Because NPM stimulates histone acetyltransferase p300, it is suggested that NPM facilitates AR attachment to DNA via p300 action on the chromatin [79]. Hence, NPM enhances AR transcriptional activity and the relationship between these two proteins may play a crucial role in PC progression. Together, in a low-hormone environment, the gain of co-activators and loss of co-repressors provide a compensatory mechanism for CR tumors. Recently, potent inhibitors of AR Binding factor 3 (BF3) pocket were developed, specifically targeting the interaction with AR and show activity in enzalutamide-resistant preclinical models [80]. Moreover, a novel class of small molecules has been recently developed (LXXLL peptidomimetics) and constitutes promising candidates for clinical development, has shown to disrupt AR co-activator interactions [81]. Neuroendocrine cells Neuroendocrine cells are a minor cell population of the epithelial compartment of normal prostate glands (Fig. 1, pathway G). In PC, neuroendocrine cells could be well-represented in CR tumors because they resist to ADT due to their lack of AR expression. The neuroendocrine phenotype acquisition is associated with tumor progression and castration resistance. Increasing evidence shows that neuroendocrine IL6 factor, one crucial factor induced by castration, enhances the neuroendocrine phenotype, thereby amplifying the wave of survival factor release [60,82]. Neuroendocrine cells notably secrete neuropeptides (NPs) such as bombesin/Gastrin-Releasing Peptide (GRP) or adrenomedullin described to induce paracrine mitogenic effects on PC cells via AR pathway activation [83–86]. Furthermore, neuroendocrine cells could promote the growth of CS tumors in castrated mice even when injected into the opposing flank, suggesting a paracrine and endocrine mechanism [87]. Data support the up-regulation of the neuroendocrine pathway after AR suppression [88,89] but the precise role of neuroendocrine cells is still unclear as their precise role in CR progression still unknown. Accumulating data support the existence of predisposing aberrations in neuroendocrine prostate cancer (NEPC) including loss of RB1 and TP53 and gain of MYCN and AURKA (for review see [17]). AURKA inhibitor MLN8237 was used in phase II trial in NEPC (NCT01799278). Non-AR-related pathways Anti-apoptotic proteins overexpression Anti-apoptotic proteins overexpression circumvents the AR pathway and utilizes other pathways to stimulate prostate cells proliferation in a CR environment (Fig. 1, pathway H). CR progression is associated with activation of anti-apoptotic proteins such as BCL2 [90–92] and clusterin [93]. We previously described that Hsp27 [94,95] and TP53INP1 [96] are upregulated in CRPC and block apoptosis normally induced by androgen withdrawal and chemotherapies. Notably, Hsp27 belongs to the 1% of genes overexpressed during CR progression [94,95] and is associated with poor prognosis and treatment resistance [97]. We also recently demonstrated that Hsp27 is a multifunctional protein capable of interacting with a great number of partner proteins to protect cells from lethal conditions [98]. We showed that the Hsp27-interacting protein TCTP is involved in Hsp27 cytoprotection in CRPC [99]. Hsp27 stabilizes TCTP by inhibiting its stress-induced ubiquitination and proteasomal degradation [99]. We provided proof-of-principle that TCTP is an inhibitor of apoptosis in human CRPC, and a rational target for therapy of CRPC that could avoid
undesirable toxicity in normal tissues through TCTP inhibition (antisense oligonucleotide) [99,100]. Hsp27 cytoprotection is also induced through its interaction with eIF4E (eukaryotic translational initiation factor 4E). Hsp27 interaction protects eIF4E from its ubiquitin–proteasome-dependent degradation process, leading to apoptosis resistance induced by castration and chemotherapy [101]. In this context, two inhibitors for anti-apoptotic proteins Hsp27 and clusterin have been developed (OGX-427 and OGX-011 respectively) and are currently used in clinical phases II and III (http://clinicaltrials.gov/). Tumor suppressor deregulation PTEN deregulation in PC initiation and progression have been largely described, nevertheless, a role for the retinoblastoma (RB1) and P53 tumor suppressors (Fig. 1, pathway I). The tumor suppressor PTEN is inactivated in CRPC by genetic mutations [102,103]. PTEN expression is often lost and reduced in advanced metastatic tumors compared to primary tumors [102]. PTEN loss causes aberrant activation of the PI3K/AKT signaling pathway, which increases AR protein level and transcriptional activity. In PTEN-deleted PC tumors, mTORC2 (mammalian target of rapamycin complex 2) is required to phosphorylate AKT at another site [104]. Loss of the PTEN regulatory phosphatase activity results in AKT recruitment and activation. In addition to influencing AR activity, activated AKT has a number of AR-independent downstream effects such as apoptosis blockage, cell proliferation and protein synthesis/cell growth [105]. AKT prevents cells from apoptosis by phosphorylating and inactivating pro-apoptotic proteins BAD and pro-caspase 9 [64] and promotes cell growth by downregulating the cell cycle inhibitor P27 [106–108]. The transcription factor and AKT target fork-head box O1 (FOXO1) induces the transcription of target genes that induce apoptosis. AKT phosphorylation of FOXO1 excludes it from the nucleus preventing cell death [109]. Functional inactivation or loss of the RB1 tumor suppressor is frequent in CRPC. RB1 loss results in upregulation of AR protein level and aberrant AR-mediated gene transcription activity. Hence, RB1 downregulation contributes to CR progression and DNA damage checkpoints loss. P53 mutations or loss are commonly found in advanced PC and bone metastasis but are uncommon in primary PC [110–112]. Thus, P53 is thought to play a role in PC progression but the extent of its contribution is still unclear. Burchardt et al. were the first to suggest that P53 mutations may promote CRPC progression [113]. P53 inhibition or loss-of-function conferred to in castration-sensitive (CS) LNCaP cells but not to parental LNCaP cells the ability to form tumors in castrated male nude mice. Recent studies have suggested that wild-type P53 is necessary for AR signaling [114] and that P53 is required for stable binding of AR to certain chromatin regions. Furthermore, it has been reported that P53 elimination in CS cells leads to cell proliferation independently of androgens [115]. Interestingly, we previously demonstrated that CRPC progression correlates with translationally controlled tumor protein (TCTP) overexpression and loss of P53. TCTP knockdown restored P53 expression and function, suggesting that castration-sensitivity is directly linked to P53 expression [116]. Taken together, these recent data show the importance of RB1 and P53 loss of functions in the transition of CSPC to CRPC and provide new insights for the development of predictive markers of response to ADT that might guide therapeutic decisions. In addition to mutations of P53, PTEN, mutations have been described in ZFHX3, RB1, APC, OR5L1, CDK12 and MLL2 [117]. miRNAs MicroRNAs (miRNAs) are non-coding RNAs that can repress protein expression by inhibiting their translation or promoting
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
mRNA degradation by base-pairing interactions with the 30 -UTR. MiRNAs may play the role of broad-spectrum oncogenes (oncomirs) or tumor suppressors (anti-oncomirs) [118] (Fig. 1, pathway J and Fig. 2). Hence, their use as prognostic, diagnostic or therapeutic biomolecules is under study [119]. Many studies have defined miRNAs with oncogenic properties in CR progression [120]. For instance, miR-221/222 upregulation has been described in CRPC [118] and an inverse correlation between miRNA-221/222 expression [121] and P27 has been reported [121,122]. On the contrary, miRNA-15a/16 is downregulated in PC samples and it has been further suggested that, because miRNA-15a/miRNA-16 are able to post-transcriptionally repress the expression of BCL2 and WNT3A, their reduced levels may play an essential role in CRPC. BCL2 upregulation is a common event in CR progression that promotes cell survival after androgens deprivation [90,92]. WNT3A has been implicated in b-catenin stabilization, which acts as a co-activator of AR promoting its activity in the presence of low androgen levels and in AKT and MAPK, activation which also provide PC cell survival and proliferation in the absence of androgens [123]. Thus, several lines of evidence support the fact that miRNA-15a/16 loss contribute to CR progression. Finally, another interesting miRNA suggested to be involved in CRPC progression is miRNA-34. Recent studies have implicated miRNA-34 in the P53 network [124,125]. miRNA-34 expression is markedly induced in a P53-dependent manner and its activation results in cell cycle arrest and apoptosis through downregulation of proteins such as cyclin D1 or BCL2 [126]. Interestingly, miRNA-34 has been found to be absent in P53-defective CRPC PC-3 and DU-145 cells [118]. These observations suggest that the loss of miRNA-34 promotes CR progression by activating proteins that enhance cell proliferation and inhibit apoptosis. Therefore, miRNA-34 is implicated in a positive feedback loop with P53 and its loss recapitulates, at least in part, P53 loss-of-function. Interestingly, several miRNAs (miR-21, miR-141, miR-221/222 and many others) have been described to regulate AR or to be regulated by AR and then to affect various signaling pathways related to cellular functions and tumor processes (cell proliferation, cell cycle transition, apoptosis, angiogenesis, castration invasion and metastasis) [120]. Among them oncomirs are highly expressed in CRPC, while anti-oncomirs are underexpressed (Fig. 2). miRNA-based therapeutics (miRNA antagonists (siRNA) and miRNA mimics) are being rapidly developed and are in phase II clinical trials in the case of Hepatitis C virus. In prostate cancer, systemic delivery of miR-34 mimic blocked tumor growth in mouse models [127].
Epigenetic changes Alterations in chromatin structure are tightly related to changes in gene transcription (Fig. 1, pathway K). DNA and histone
5
modifications like methylation or acetylation are critical regulators of gene expression. DNA methylation is mediated by DNA methyltransferases that methylate cytosine residues. 80–90% of coding CpG (50 -cytosine phosphatase guanidine-30 ) dinucleotides are methylated. CpG hypermethylation in human gene promoters is associated with gene silencing while hypomethylation results in increased gene transcription [128]. Increasing evidence have suggested that aberrant DNA methylation resulting in gene expression alterations may contribute to CR progression by promoting genomic instability [129]. As described above, PTEN loss-of-function plays an important role in CR progression and PTEN silencing is often the consequence of CpG islands methylation located in its promoter region [102,130]. This aberrant PTEN promoter region methylation would have a role in CR progression via the loss of PTEN and the activation of PI3K/AKT signaling pathway. In 30% of CRPC, the AR promoter region is hypermethylated, resulting in the loss of AR expression in those tumors [131]. A recent study has used metastatic CRPC tumors obtained at autopsy to identify the most common gene copy number aberrations and their concordant methylation levels in metastatic CRPC [132]. Gene copy number and CpG methylation was found to work in concert, particularly for genes commonly deleted in CRPC and that this cooperation seems to be essential for inhibiting the expression of critical tumor suppressors like RB1. Thus, DNA methylation seems to be a key epigenetic change that contributes to CR progression, highlighting the possibility of targeting such alterations in CRPC. Hypomethylating agents (e.g. 5-azacytidine) are already available and their efficiency to delay the progression of CR disease is under pre-clinical study [132].
Alternative splicing Splicing events control gene expression and their alteration plays a role in human disease and cancer [133]. The spliceosome complex is characterized by a dynamic composition and conformation. It results from the ordered interactions of small nuclear ribonucleoprotein (snRNPs) and numerous splicing factors [134]. mRNA alternative splicing (AS) (Fig. 1, pathway L) is a gene-expression regulation strategy used by cells to increase protein variety and regulation. Modulation of protein functions by AS has been documented for several genes in PC including AR itself, indicating that this mechanism can significantly contribute to genotypic heterogeneity in cancer cells and among CRPC patients [135,136]. Growing evidence show that AS of AR is a key capability of cancer cells to evade the normal activation of the pathway [136]. Splicing is itself controlled by several mechanisms. Our discovery of direct interactions of Hsp27 with splicing factors, regulators and spliceosome ribonucleoproteins as well as experimental evidence for the alternative splicing of 1777 genes in PC cells in the
CRPC progression Oncomirs miR-221/222/P27/AR miR-221/222/ /P27 27/A / R miR-21/AR miR-141/AR
An-oncomirs miRNA-34/Cyclin miRNA-34/C /Cyclililin D1/BCL2/P53 /C D1/BC /BCLL2/P53 miRNA-15a/16/P53/BCL2/WNT3A Fig. 2. Oncomirs are highly expressed in castration resistant prostate cancer and anti-oncomirs are underexpressed. miRNAs are in black and their target proteins in red.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
6
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
absence of Hsp27, strongly supports the involvement of Hsp27 in differential splicing events [98]. High-throughput analyses of PC samples have revealed more than 200 genes whose AS is differentially regulated. The physiological consequence of the majority of these aberrant AS events is still unknown [137]. Two critical pathways in CRPC are subject to AS control, AR and PI3K/AKT/mTOR [138]. The majority of the alternatively spliced genes show a variation in the expression levels, the best known examples including AR, transcription factor KLF6 (Kruppel-like factor 6), cyclin D1 (CCDN1), BCL-X, FGFR2 and VEGFA (Fig. 3). Several studies suggest that the upregulation of selected splicing factors (SAM68, SRSF1, DDX5) directly alter the splicing profile of key genes [135,138] and therefore constitute potential therapeutic targets. However, inhibition of a splicing factor activity is not an easy task, despite some examples of success [139]. Spliceostatin A (SSA), targeting the splicing factor 3B subunit 1 (SF3B1) shows anti-oncogenic properties in a variety of cancer cell models [140]. Altogether, AS (de)regulation represents a relatively unexplored aspect of the of CRPC complexity that deserve further exploration. Furthermore, clinical investigation of applying antisense oligonucleotides to down-regulate mRNAs contributing to cancer cell survival has shown promising results [141] but no other therapeutic strategy has been proposed to date to target splicing.
Gene fusions Technological progress, notably the development of paired-end and whole transcriptome sequencing, or exon arrays, has allowed
CCND1 normal
AS
CCND1D1a
the discovery of an increasing number of chimeric RNAs in PC [142–145] (Fig. 1, pathway M). One of the earliest genetic alterations described to occur in 50% of PC but have also been observed in CR and metastatic PC [144,146], is the overexpression of the ERG oncogene, a member of the large family of ETS transcription factors involved in various biological processes such as cell proliferation, differentiation, apoptosis, metastasis and angiogenesis [147–149]. Because ERG fusion transcripts are so frequently observed in prostate tumors, their presence in tissue or urine represent potent diagnostic markers in a large group of patients. This deregulation results from the fusion of the ERG gene with the first exon(s) of transmembrane protease serine 2 (TMPRSS2), a prostate-specific and androgen-regulated gene located close to ERG (Fig. 4) on the same 21q22 chromosome region [147,150]. Fusion of these genes can occur either by interstitial deletion leading to loss of genomic region between the two genes (60% of fusion-positive tumors), or by complex genomic rearrangements involving various chromosomes [148]. This gene fusion is specific to PC as has never been detected in normal prostate [151,152]. It is frequently correlated with poor clinical outcome [153]. Interestingly, the AR has been implicated in the processes resulting in gene fusions by inducing the spatial proximity of genes involved in rearrangements, promoting the formation of double-strand DNA breaks (DSB), and facilitating the recruitment of proteins for non-homologous end-joining (NHEJ). Under androgens influence, changes in chromatin organization are induced that can juxtapose the transcription units of both genes, thereby facilitating the genesis of this gene fusion [154]. Interestingly, the presence of TMPRSS2-ERG
BCL-X cancer
normal
CCND1D1b
AS
BCL-xS
FGFR2 cancer
normal
FGFR2IIIb
BCL-xL
AR
AR
AS
cancer
FGFR2IIIc FGF8b
FGF7
SLUG
Growth, EMT/Metastasis
Apoptosis Chemoresistance
VEGFA normal
AS
VEGF165b
EMT/Metastasis
KLF6 cancer
VEGF165
VEGFR2
normal
AS
AR cancer
KLF6SV1
KLF6-FL
VEGFR2
normal
AS
AR-FL DHT
Cancer CRPC
AR-V7 ARv567es
P21
Angiogenesis
Cell cycle Growth, Invasion, Angiogenesis
Differenaon Drug resistance
Fig. 3. Alternative splicing leading to isoforms differentially regulated in prostate cancer (pink = up-regulated, green = down-regulated).
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
7
ERG TMPRRS2
ERG Target Gene
ERG
ERG
ERG
ERG Target Gene
ERG Target Gene
Prostate cancer
Fig. 4. Gene fusion events are recurrent in castration-resistant prostate cancer. TMPRSS2-ERG fusion results in the upregulation of ERG transcription which then exerts its effects by binding target gene promoter regions, which results in their activation or inhibition, and the generation of neoplastic phenotype.
fusion was proposed as a biomarker for the identification of abiraterone-sensitive tumors [6]. ETS genes are frequently involved in gene fusions, resulting in the formation of chimeric proteins or altered expression of ETS proteins. Less frequently, ERG overexpression is not caused by TMPRSS2 fusion, but by fusion to SLC45A3 or NDRG1, both androgen-regulated genes preferentially expressed in prostate but not located on the same chromosomes ERG. This indicates that chromosomal proximity is not essential for fusion events but long distance gene fusions seem to be facilitated by nuclear proximity of the fusion partners [155,156]. In a small percentage (6%) of prostate tumors, the ETS gene ELK4 is deregulated due to cis-splicing of the adjacent SLC45A3 gene and with no apparent chromosomal rearrangement [157,158]. Moreover, it has been shown that TMPRSS2-ETS rearrangements also occur in CRPC providing strong evidence that ERG fusions represent an aggressive molecular subtype of PC suggesting the existence of castration-resistant mechanisms of TMPRSS2 expression in prostate [159,160]. In CRPC, one function of TMPRSS2-ERG is proposed to be the expansion of self-renewing cells, which may serve as targets for subsequent mutations [161]. The precise functional role of ERG and other ETS proteins in CRPC is not fully understood, because ETS overexpression alone is insufficient to induce PC. Some pathways associated to ERG overexpression have been identified, such as the TGFb, TDRD1 and WNT pathways [162–165]. Gene fusions in CRPC demand further investigation in order to decipher the role and outcome of gene fusions at this stage of the disease. Interestingly, patients with CRPC will be stratified for the presence of TMPRSS2-ERG fusion and randomized to abiraterone protocols (see clinicaltrials.gov, identifier: NCT01576172). A recent clinical trial using ABT-888 (Veliparib), an oral PARP inhibitor targeting TMPRSS2-ERG fusions has recently been carried out (see clinicaltrials.gov, identifier: NCT01085422, NCT01576172). Numerous studies show that PARP inhibitors, when added to hormone therapy, contribute to tumor regression especially those expressing TMPRSS2-ERG and clinical trials with several PARP inhibitors are currently being conducted in various cancers to assess their toxicities, efficacies and benefit as monotherapies or combined with radiation or other chemotherapeutic agents [166,167].
Conclusion and future directions Mechanisms underlying CRPC progression have been identified as early as the 1940s and many probably remain to be uncovered to fully understand the disease. Among the mechanisms, recent studies have identified a wide variety of recurrent mutations in proteins interacting with AR [117]. Due to the increasing amount of knowledge of the mutational profile of PC, there is a need to further understand the consequences of these recurrent mutations in CRPC progression. While there have been significant efforts to help understand CRPC biology over the past decades, some aspects of
genetic and epigenetic alterations deserve deeper exploration, such as alternative splicing, chimeric transcripts, miRNA interactions with target genes. Current knowledge of CRPC mechanisms have contributed to the development and FDA approval of various molecules which have allowed overall survival gains that were inevitably countered by resistance and emergence of a more aggressive and heterogeneous disease. We believe that further elucidation of CRPC mechanisms will increase the chances of finding new targets and designing new therapeutic options for CRPC, mainly cancer-specific combinatory therapies as well as siRNA and ASO therapeutic strategies as treatment alternatives. Translational research and close collaboration between physicians and researchers is indispensable for therapeutic strategies for this highly heterogeneous disease. Knowing a patient’s genetic and biological background of their tumor will be useful for identification of more effective prevention, screening and treatment strategies with fewer side effects allowing to customize and personalize therapy to each patient. In the future, optimal therapeutic choices will rely on targeting pathway combinations with the AR specific to patient sub-groups. Conflict of interest statement The authors declare they have no competing interests as defined by Nature Publishing Group, or other interests that might be perceived to influence the interpretation of the article. Acknowledgements The authors received financial support from: French Cancer Institute (InCa, PAIR prostate program #R10111AA), ITMO Cancer (BioSys call, #A12171AS), Institut national de la santé et de la recherche médicale (Inserm), Association pour la Recherche sur le Cancer (ARC), Agence Nationale pour la Recherche (ANR, Emergence Program #ANR-11-EMMA-0022), Aix-Marseille University and competitivity pole Eurobiomed, ARTP (Association pour la Recherche sur les Tumeurs de la Prostate, N° R14107AA). References [1] Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin 2010;60:277–300. [2] Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11–30. [3] Fusi A, Procopio G, Della Torre S, Ricotta R, Bianchini G, Salvioni R, et al. Treatment options in hormone-refractory metastatic prostate carcinoma. Tumori 2004;90:535–46. [4] Galsky MD, Small AC, Tsao CK, Oh WK. Clinical development of novel therapeutics for castration-resistant prostate cancer: historic challenges and recent successes. CA Cancer J Clin 2012;62:299–308. [5] Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JM, Papaemmanuil E, et al. The evolutionary history of lethal metastatic prostate cancer. Nature 2015;520:353–7. [6] Suzman DL, Antonarakis ES. Castration-resistant prostate cancer: latest evidence and therapeutic implications. Ther Adv Med Oncol 2014;6:167–79.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
8
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
[7] MacVicar GR, Hussain MH. Emerging therapies in metastatic castrationsensitive and castration-resistant prostate cancer. Curr Opin Oncol 2013;25:252–60. [8] Geller J. Prolonging survival in metastatic prostate cancer: the case for adrenal androgens–overview and summary of therapeutic controversies in prostatic cancer. J Clin Endocrinol Metab 1995;80:1074–8. [9] Mohler JL. Castration-recurrent prostate cancer is not androgen-independent. Adv Exp Med Biol 2008;617:223–34. [10] Craft N, Chhor C, Tran C, Belldegrun A, DeKernion J, Witte ON, et al. Evidence for clonal outgrowth of androgen-independent prostate cancer cells from androgen-dependent tumors through a two-step process. Cancer Res 1999;59:5030–6. [11] Debes JD, Tindall DJ. The role of androgens and the androgen receptor in prostate cancer. Cancer Lett 2002;187:1–7. [12] Devlin HL, Mudryj M. Progression of prostate cancer: multiple pathways to androgen independence. Cancer Lett 2009;274:177–86. [13] Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer 2001;1:34–45. [14] Lamont KR, Tindall DJ. Minireview: alternative activation pathways for the androgen receptor in prostate cancer. Mol Endocrinol 2011;25:897–907. [15] Saraon P, Jarvi K, Diamandis EP. Molecular alterations during progression of prostate cancer to androgen independence. Clin Chem 2011;57:1366–75. [16] Rocchi P, Muracciole X, Fina F, Mulholland DJ, Karsenty G, Palmari J, et al. Molecular analysis integrating different pathways associated with androgenindependent progression in LuCaP 23.1 xenograft. Oncogene 2004;23:9111–9. [17] Wyatt AW, Gleave ME. Targeting the adaptive molecular landscape of castration-resistant prostate cancer. EMBO Mol Med 2015. [18] Arnold JT, Isaacs JT. Mechanisms involved in the progression of androgenindependent prostate cancers: it is not only the cancer cell’s fault. Endocr Relat Cancer 2002;9:61–73. [19] Pienta KJ, Bradley D. Mechanisms underlying the development of androgenindependent prostate cancer. Clin Cancer Res 2006;12:1665–71. [20] Bubendorf L, Kononen J, Koivisto P, Schraml P, Moch H, Gasser TC, et al. Survey of gene amplifications during prostate cancer progression by highthroughout fluorescence in situ hybridization on tissue microarrays. Cancer Res 1999;59:803–6. [21] Edwards J, Krishna NS, Witton CJ, Bartlett JM. Gene amplifications associated with the development of hormone-resistant prostate cancer. Clin Cancer Res 2003;9:5271–81. [22] Scher HI, Beer TM, Higano CS, Anand A, Taplin ME, Efstathiou E, et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study. Lancet 2010;375:1437–46. [23] Schalken J, Fitzpatrick JM. Enzalutamide: targeting the androgen signalling pathway in metastatic castration-resistant prostate cancer. BJU Int 2015. [24] Holzbeierlein J, Lal P, LaTulippe E, Smith A, Satagopan J, Zhang L, et al. Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am J Pathol 2004;164:217–27. [25] Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res 2008;68:4447–54. [26] Dutt SS, Gao AC. Molecular mechanisms of castration-resistant prostate cancer progression. Future Oncol 2009;5:1403–13. [27] Makridakis N, Ross RK, Pike MC, Chang L, Stanczyk FZ, Kolonel LN, et al. A prevalent missense substitution that modulates activity of prostatic steroid 5alpha-reductase. Cancer Res 1997;57:1020–2. [28] Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, Wood CA, et al. Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res 2008;68:6407–15. [29] Mohler JL. A role for the androgen-receptor in clinically localized and advanced prostate cancer. Best Pract Res Clin Endocrinol Metab 2008;22:357–72. [30] Marcelli M, Ittmann M, Mariani S, Sutherland R, Nigam R, Murthy L, et al. Androgen receptor mutations in prostate cancer. Cancer Res 2000;60:944–9. [31] Taplin ME, Bubley GJ, Shuster TD, Frantz ME, Spooner AE, Ogata GK, et al. Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer. N Engl J Med 1995;332:1393–8. [32] Tilley WD, Buchanan G, Hickey TE, Bentel JM. Mutations in the androgen receptor gene are associated with progression of human prostate cancer to androgen independence. Clin Cancer Res 1996;2:277–85. [33] Brooke GN, Bevan CL. The role of androgen receptor mutations in prostate cancer progression. Curr Genomics 2009;10:18–25. [34] Culig Z, Hobisch A, Cronauer MV, Radmayr C, Trapman J, Hittmair A, et al. Androgen receptor activation in prostatic tumor cell lines by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth factor. Cancer Res 1994;54:5474–8. [35] Taplin ME. Drug insight: role of the androgen receptor in the development and progression of prostate cancer. Nat Clin Pract Oncol 2007;4:236–44. [36] Veldscholte J, Berrevoets CA, Brinkmann AO, Grootegoed JA, Mulder E. Antiandrogens and the mutated androgen receptor of LNCaP cells: differential effects on binding affinity, heat-shock protein interaction, and transcription activation. Biochemistry 1992;31:2393–9. [37] Zhao XY, Malloy PJ, Krishnan AV, Swami S, Navone NM, Peehl DM, et al. Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor. Nat Med 2000;6:703–6.
[38] Bergerat JP, Ceraline J. Pleiotropic functional properties of androgen receptor mutants in prostate cancer. Hum Mutat 2009;30:145–57. [39] Culig Z, Klocker H, Bartsch G, Hobisch A. Androgen receptor mutations in carcinoma of the prostate: significance for endocrine therapy. Am J Pharmacogenomics 2001;1:241–9. [40] Suzuki H, Sato N, Watabe Y, Masai M, Seino S, Shimazaki J. Androgen receptor gene mutations in human prostate cancer. J Steroid Biochem Mol Biol 1993;46:759–65. [41] Taplin ME, Bubley GJ, Ko YJ, Small EJ, Upton M, Rajeshkumar B, et al. Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Res 1999;59:2511–5. [42] Steinkamp MP, O’Mahony OA, Brogley M, Rehman H, Lapensee EW, Dhanasekaran S, et al. Treatment-dependent androgen receptor mutations in prostate cancer exploit multiple mechanisms to evade therapy. Cancer Res 2009;69:4434–42. [43] Black BE, Vitto MJ, Gioeli D, Spencer A, Afshar N, Conaway MR, et al. Transient, ligand-dependent arrest of the androgen receptor in subnuclear foci alters phosphorylation and coactivator interactions. Mol Endocrinol 2004;18:834–50. [44] Chmelar R, Buchanan G, Need EF, Tilley W, Greenberg NM. Androgen receptor coregulators and their involvement in the development and progression of prostate cancer. Int J Cancer 2007;120:719–33. [45] Li W, Cavasotto CN, Cardozo T, Ha S, Dang T, Taneja SS, et al. Androgen receptor mutations identified in prostate cancer and androgen insensitivity syndrome display aberrant ART-27 coactivator function. Mol Endocrinol 2005;19:2273–82. [46] Ceraline J, Cruchant MD, Erdmann E, Erbs P, Kurtz JE, Duclos B, et al. Constitutive activation of the androgen receptor by a point mutation in the hinge region: a new mechanism for androgen-independent growth in prostate cancer. Int J Cancer 2004;108:152–7. [47] Chan SC, Li Y, Dehm SM. Androgen receptor splice variants activate androgen receptor target genes and support aberrant prostate cancer cell growth independent of canonical androgen receptor nuclear localization signal. J Biol Chem 2012;287:19736–49. [48] Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res 2009;69:2305–13. [49] Marcias G, Erdmann E, Lapouge G, Siebert C, Barthelemy P, Duclos B, et al. Identification of novel truncated androgen receptor (AR) mutants including unreported pre-mRNA splicing variants in the 22Rv1 hormone-refractory prostate cancer (PCa) cell line. Hum Mutat 2009;31:74–80. [50] Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, et al. Ligandindependent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res 2009;69:16–22. [51] Sun S, Sprenger CC, Vessella RL, Haugk K, Soriano K, Mostaghel EA, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clin Invest 2010;120:2715–30. [52] Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, Widmark A, et al. Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival. PLoS ONE 2011;6:e19059. [53] Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KA, Dehm SM. Androgen receptor splice variants mediate enzalutamide resistance in castrationresistant prostate cancer cell lines. Cancer Res 2013;73:483–9. [54] Andersen RJ, Mawji NR, Wang J, Wang G, Haile S, Myung JK, et al. Regression of castrate-recurrent prostate cancer by a small-molecule inhibitor of the amino-terminus domain of the androgen receptor. Cancer Cell 2010;17:535–46. [55] Snoek R, Cheng H, Margiotti K, Wafa LA, Wong CA, Wong EC, et al. In vivo knockdown of the androgen receptor results in growth inhibition and regression of well-established, castration-resistant prostate tumors. Clin Cancer Res 2009;15:39–47. [56] Compagno D, Merle C, Morin A, Gilbert C, Mathieu JR, Bozec A, et al. SIRNAdirected in vivo silencing of androgen receptor inhibits the growth of castration-resistant prostate carcinomas. PLoS ONE 2007;2:e1006. [57] Manin M, Martinez A, Van Der Schueren B, Reynaert I, Jean C. Acquisition of androgen-mediated expression of mouse vas deferens protein (MVDP) gene in cultured epithelial cells and in vas deferens during postnatal development. J Androl 2000;21:641–50. [58] Wu JD, Haugk K, Woodke L, Nelson P, Coleman I, Plymate SR. Interaction of IGF signaling and the androgen receptor in prostate cancer progression. J Cell Biochem 2006;99:392–401. [59] Edwards J, Bartlett JM. The androgen receptor and signal-transduction pathways in hormone-refractory prostate cancer. Part 2: Androgen-receptor cofactors and bypass pathways. BJU Int 2005;95:1327–35. [60] Lee SO, Lou W, Hou M, de Miguel F, Gerber L, Gao AC. Interleukin-6 promotes androgen-independent growth in LNCaP human prostate cancer cells. Clin Cancer Res 2003;9:370–6. [61] Lee SO, Lou W, Hou M, Onate SA, Gao AC. Interleukin-4 enhances prostatespecific antigen expression by activation of the androgen receptor and Akt pathway. Oncogene 2003;22:7981–8. [62] Ueda T, Bruchovsky N, Sadar MD. Activation of the androgen receptor Nterminal domain by interleukin-6 via MAPK and STAT3 signal transduction pathways. J Biol Chem 2002;277:7076–85.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx [63] Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010;141:1117–34. [64] Wen Y, Hu MC, Makino K, Spohn B, Bartholomeusz G, Yan DH, et al. HER-2/ neu promotes androgen-independent survival and growth of prostate cancer cells through the Akt pathway. Cancer Res 2000;60:6841–5. [65] Manin M, Baron S, Goossens K, Beaudoin C, Jean C, Veyssiere G, et al. Androgen receptor expression is regulated by the phosphoinositide 3-kinase/ Akt pathway in normal and tumoral epithelial cells. Biochem J 2002;366: 729–36. [66] Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C. From HER2/Neu signal cascade to androgen receptor and its coactivators: a novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc Natl Acad Sci USA 1999;96:5458–63. [67] Gregory CW, Fei X, Ponguta LA, He B, Bill HM, French FS, et al. Epidermal growth factor increases coactivation of the androgen receptor in recurrent prostate cancer. J Biol Chem 2004;279:7119–30. [68] Nadiminty N, Lou W, Sun M, Chen J, Yue J, Kung HJ, et al. Aberrant activation of the androgen receptor by NF-kappaB2/p52 in prostate cancer cells. Cancer Res 2010;70:3309–19. [69] Amato RJ, Wilding G, Bubley G, Loewy J, Haluska F, Gross ME. Safety and preliminary efficacy analysis of the mTOR inhibitor ridaforolimus in patients with taxane-treated, castration-resistant prostate cancer. Clin Genitourin Cancer 2012;10:232–8. [70] Templeton AJ, Dutoit V, Cathomas R, Rothermundt C, Bartschi D, Droge C, et al. Phase 2 trial of single-agent everolimus in chemotherapy-naive patients with castration-resistant prostate cancer (SAKK 08/08). Eur Urol 2013;64: 150–8. [71] Kruczek K, Ratterman M, Tolzien K, Sulo S, Lestingi TM, Nabhan C. A phase II study evaluating the toxicity and efficacy of single-agent temsirolimus in chemotherapy-naive castration-resistant prostate cancer. Br J Cancer 2013;109:1711–6. [72] Thomas C, Lamoureux F, Crafter C, Davies BR, Beraldi E, Fazli L, et al. Synergistic targeting of PI3K/AKT pathway and androgen receptor axis significantly delays castration-resistant prostate cancer progression in vivo. Mol Cancer Ther 2013;12:2342–55. [73] Dehm SM, Tindall DJ. Regulation of androgen receptor signaling in prostate cancer. Expert Rev Anticancer Ther 2005;5:63–74. [74] Gregory CW, He B, Johnson RT, Ford OH, Mohler JL, French FS, et al. A mechanism for androgen receptor-mediated prostate cancer recurrence after androgen deprivation therapy. Cancer Res 2001;61:4315–9. [75] Halkidou K, Gnanapragasam VJ, Mehta PB, Logan IR, Brady ME, Cook S, et al. Expression of Tip60, an androgen receptor coactivator, and its role in prostate cancer development. Oncogene 2003;22:2466–77. [76] Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev 2007;28:778–808. [77] Mestayer C, Blanchere M, Jaubert F, Dufour B, Mowszowicz I. Expression of androgen receptor coactivators in normal and cancer prostate tissues and cultured cell lines. Prostate 2003;56:192–200. [78] Leotoing L, Meunier L, Manin M, Mauduit C, Decaussin M, Verrijdt G, et al. Influence of nucleophosmin/B23 on DNA binding and transcriptional activity of the androgen receptor in prostate cancer cell. Oncogene 2008;27: 2858–67. [79] Beaudoin C, Maquaire S, Leotoing L, Allemand J, Lours-Calet C, Veyssiere G, et al. NPM: a new player in prostate cancer? Med Sci (Paris) 2009;25: 19–21. [80] Munuganti RS, Hassona MD, Leblanc E, Frewin K, Singh K, Ma D, et al. Identification of a potent antiandrogen that targets the BF3 site of the androgen receptor and inhibits enzalutamide-resistant prostate cancer. Chem Biol 2014;21:1476–85. [81] Ravindranathan P, Lee TK, Yang L, Centenera MM, Butler L, Tilley WD, et al. Peptidomimetic targeting of critical androgen receptor-coregulator interactions in prostate cancer. Nat Commun. 2013;4:1923. [82] Qiu Y, Robinson D, Pretlow TG, Kung HJ. Etk/Bmx, a tyrosine kinase with a pleckstrin-homology domain, is an effector of phosphatidylinositol 30 -kinase and is involved in interleukin 6-induced neuroendocrine differentiation of prostate cancer cells. Proc Natl Acad Sci USA 1998;95:3644–9. [83] Aprikian AG, Cordon-Cardo C, Fair WR, Reuter VE. Characterization of neuroendocrine differentiation in human benign prostate and prostatic adenocarcinoma. Cancer 1993;71:3952–65. [84] Hirano D, Okada Y, Minei S, Takimoto Y, Nemoto N. Neuroendocrine differentiation in hormone refractory prostate cancer following androgen deprivation therapy. Eur Urol 2004;45:586–92. discussion 92. [85] Komiya A, Suzuki H, Imamoto T, Kamiya N, Nihei N, Naya Y, et al. Neuroendocrine differentiation in the progression of prostate cancer. Int J Urol 2009;16:37–44. [86] Rocchi P, Boudouresque F, Zamora AJ, Muracciole X, Lechevallier E, Martin PM, et al. Expression of adrenomedullin and peptide amidation activity in human prostate cancer and in human prostate cancer cell lines. Cancer Res 2001;61:1196–206. [87] Jin RJ, Wang Y, Masumori N, Ishii K, Tsukamoto T, Shappell SB, et al. NE-10 neuroendocrine cancer promotes the LNCaP xenograft growth in castrated mice. Cancer Res 2004;64:5489–95. [88] Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov 2011;1:487–95.
9
[89] Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res 2014;74:1272–83. [90] Colombel M, Symmans F, Gil S, O’Toole KM, Chopin D, Benson M, et al. Detection of the apoptosis-suppressing oncoprotein bc1-2 in hormonerefractory human prostate cancers. Am J Pathol 1993;143:390–400. [91] Gleave ME, Miayake H, Goldie J, Nelson C, Tolcher A. Targeting bcl-2 gene to delay androgen-independent progression and enhance chemosensitivity in prostate cancer using antisense bcl-2 oligodeoxynucleotides. Urology 1999;54:36–46. [92] McDonnell TJ, Troncoso P, Brisbay SM, Logothetis C, Chung LW, Hsieh JT, et al. Expression of the protooncogene bcl-2 in the prostate and its association with emergence of androgen-independent prostate cancer. Cancer Res 1992;52:6940–4. [93] July LV, Akbari M, Zellweger T, Jones EC, Goldenberg SL, Gleave ME. Clusterin expression is significantly enhanced in prostate cancer cells following androgen withdrawal therapy. Prostate 2002;50:179–88. [94] Rocchi P, Beraldi E, Ettinger S, Fazli L, Vessella RL, Nelson C, et al. Increased Hsp27 after androgen ablation facilitates androgen-independent progression in prostate cancer via signal transducers and activators of transcription 3-mediated suppression of apoptosis. Cancer Res 2005;65: 11083–93. [95] Rocchi P, So A, Kojima S, Signaevsky M, Beraldi E, Fazli L, et al. Heat shock protein 27 increases after androgen ablation and plays a cytoprotective role in hormone-refractory prostate cancer. Cancer Res 2004;64:6595–602. [96] Giusiano S, Baylot V, Andrieu C, Fazli L, Gleave M, Iovanna JL, et al. TP53INP1 as new therapeutic target in castration-resistant prostate cancer. Prostate 2012;72:1286–94. [97] Acunzo J, Andrieu C, Baylot V, So A, Rocchi P. Hsp27 as a therapeutic target in cancers. Curr Drug Targets 2014;15:423–31. [98] Katsogiannou M, Andrieu C, Baylot V, Baudot A, Dusetti NJ, Gayet O, et al. The functional landscape of Hsp27 reveals new cellular processes such as DNA repair and alternative splicing and proposes novel anticancer targets. Mol Cell Proteomics 2014;13:3585–601. [99] Baylot V, Katsogiannou M, Andrieu C, Taieb D, Acunzo J, Giusiano S, et al. Targeting TCTP as a new therapeutic strategy in castration-resistant prostate cancer. Mol Ther 2012;20:2244–56. [100] Acunzo J, Baylot V, So A, Rocchi P. TCTP as therapeutic target in cancers. Cancer Treat Rev 2014;40:760–9. [101] Andrieu C, Taieb D, Baylot V, Ettinger S, Soubeyran P, De-Thonel A, et al. Heat shock protein 27 confers resistance to androgen ablation and chemotherapy in prostate cancer cells through eIF4E. Oncogene 2010;29:1883–96. [102] Suzuki H, Freije D, Nusskern DR, Okami K, Cairns P, Sidransky D, et al. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res 1998;58:204–9. [103] Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 1997;275:1943–7. [104] Guertin DA, Stevens DM, Saitoh M, Kinkel S, Crosby K, Sheen JH, et al. MTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell 2009;15:148–59. [105] Sarker D, Reid AH, Yap TA, de Bono JS. Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clin Cancer Res 2009;15: 4799–805. [106] Graff JR, Konicek BW, McNulty AM, Wang Z, Houck K, Allen S, et al. Increased AKT activity contributes to prostate cancer progression by dramatically accelerating prostate tumor growth and diminishing p27Kip1 expression. J Biol Chem 2000;275:24500–5. [107] Li P, Lee H, Guo S, Unterman TG, Jenster G, Bai W. AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR. Mol Cell Biol 2003;23:104–18. [108] Medema RH, Kops GJ, Bos JL, Burgering BM. AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 2000;404:782–7. [109] Liu P, Li S, Gan L, Kao TP, Huang H. A transcription-independent function of FOXO1 in inhibition of androgen-independent activation of the androgen receptor in prostate cancer cells. Cancer Res 2008;68:10290–9. [110] Eastham JA, Hall SJ, Sehgal I, Wang J, Timme TL, Yang G, et al. In vivo gene therapy with p53 or p21 adenovirus for prostate cancer. Cancer Res 1995;55:5151–5. [111] Eastham JA, Stapleton AM, Gousse AE, Timme TL, Yang G, Slawin KM, et al. Association of p53 mutations with metastatic prostate cancer. Clin Cancer Res 1995;1:1111–8. [112] Navone NM, Labate ME, Troncoso P, Pisters LL, Conti CJ, von Eschenbach AC, et al. P53 mutations in prostate cancer bone metastases suggest that selected p53 mutants in the primary site define foci with metastatic potential. J Urol 1999;161:304–8. [113] Burchardt M, Burchardt T, Shabsigh A, Ghafar M, Chen MW, Anastasiadis A, et al. Reduction of wild type p53 function confers a hormone resistant phenotype on LNCaP prostate cancer cells. Prostate 2001;48:225–30. [114] Cronauer MV, Schulz WA, Burchardt T, Ackermann R, Burchardt M. Inhibition of p53 function diminishes androgen receptor-mediated signaling in prostate cancer cell lines. Oncogene 2004;23:3541–9. [115] Guseva NV, Rokhlin OW, Glover RA, Cohen MB. P53 and the proteasome regulate androgen receptor activity. Cancer Biol Ther 2012;13:553–8.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003
10
M. Katsogiannou et al. / Cancer Treatment Reviews xxx (2015) xxx–xxx
[116] Baylot V, Katsogiannou M, Andrieu C, Taieb D, Acunzo J, Giusiano S, et al. Targeting TCTP as a new therapeutic strategy in castration-resistant prostate. Cancer Mol Ther 2012. [117] Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239–43. [118] Coppola V, De Maria R, Bonci D. MicroRNAs and prostate cancer. Endocr Relat Cancer 2010;17. F1–17. [119] Maugeri-Sacca M, Coppola V, Bonci D, De Maria R. MicroRNAs and prostate cancer: from preclinical research to translational oncology. Cancer J 2012;18:253–61. [120] ChunJiao S, Huan C, ChaoYang X, GuoMei R. Uncovering the roles of miRNAs and their relationship with androgen receptor in prostate cancer. IUBMB Life 2014;66:379–86. [121] Mercatelli N, Coppola V, Bonci D, Miele F, Costantini A, Guadagnoli M, et al. The inhibition of the highly expressed miR-221 and miR-222 impairs the growth of prostate carcinoma xenografts in mice. PLoS ONE 2008;3:e4029. [122] Galardi S, Mercatelli N, Giorda E, Massalini S, Frajese GV, Ciafre SA, et al. MiR221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem 2007;282:23716–24. [123] Yun H, Lee M, Kim SS, Ha J. Glucose deprivation increases mRNA stability of vascular endothelial growth factor through activation of AMP-activated protein kinase in DU145 prostate carcinoma. J Biol Chem 2005;280:9963–72. [124] He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, et al. A microRNA component of the p53 tumour suppressor network. Nature 2007;447:1130–4. [125] Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE, et al. P53mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol 2007;17:1298–307. [126] Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007;26:745–52. [127] Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, et al. The microRNA miR34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 2011;17:211–5. [128] Jones PA, Gonzalgo ML, Tsutsumi M, Bender CM. DNA methylation in bladder cancer. Eur Urol 1998;33(Suppl. 4):7–8. [129] Chin SP, Dickinson JL, Holloway AF. Epigenetic regulation of prostate cancer. Clin Epigenetics 2011;2:151–69. [130] Cairns P, Okami K, Halachmi S, Halachmi N, Esteller M, Herman JG, et al. Frequent inactivation of PTEN/MMAC1 in primary prostate cancer. Cancer Res 1997;57:4997–5000. [131] Suzuki H, Ueda T, Ichikawa T, Ito H. Androgen receptor involvement in the progression of prostate cancer. Endocr Relat Cancer 2003;10:209–16. [132] Friedlander TW, Roy R, Tomlins SA, Ngo VT, Kobayashi Y, Azameera A, et al. Common structural and epigenetic changes in the genome of castrationresistant prostate cancer. Cancer Res 2012;72:616–25. [133] Germann S, Gratadou L, Dutertre M, Auboeuf D. Splicing programs and cancer. J Nucleic Acids 2012;2012:269570. [134] Matlin AJ, Moore MJ. Spliceosome assembly and composition. Adv Exp Med Biol 2007;623:14–35. [135] Sette C. Alternative splicing programs in prostate cancer. Int J Cell Biol 2013;2013:458727. [136] Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res 2008;68:5469–77. [137] Zhang C, Li HR, Fan JB, Wang-Rodriguez J, Downs T, Fu XD, et al. Profiling alternatively spliced mRNA isoforms for prostate cancer classification. BMC Bioinformatics 2006;7:202. [138] Lapuk AV, Volik SV, Wang Y, Collins CC. The role of mRNA splicing in prostate cancer. Asian J Androl 2014;16:515–21. [139] Pedrotti S, Bielli P, Paronetto MP, Ciccosanti F, Fimia GM, Stamm S, et al. The splicing regulator Sam68 binds to a novel exonic splicing silencer and functions in SMN2 alternative splicing in spinal muscular atrophy. EMBO J 2010;29:1235–47. [140] Bonnal S, Vigevani L, Valcarcel J. The spliceosome as a target of novel antitumour drugs. Nat Rev Drug Discov 2012;11:847–59. [141] Blair CA, Zi X. Potential molecular targeting of splice variants for cancer treatment. Indian J Exp Biol 2011;49:836–9. [142] Kannan K, Wang L, Wang J, Ittmann MM, Li W, Yen L. Recurrent chimeric RNAs enriched in human prostate cancer identified by deep sequencing. Proc Natl Acad Sci USA 2011;108:9172–7. [143] Teles Alves I, Hartjes T, McClellan E, Hiltemann S, Bottcher R, Dits N, et al. Next-generation sequencing reveals novel rare fusion events with functional implication in prostate cancer. Oncogene 2014.
[144] Smit FP, Salagierski M, Jannink S, Schalken JA. High-resolution ERGexpression profiling on GeneChip exon 1.0 ST arrays in primary and castration-resistant prostate cancer. BJU Int 2013;111:836–42. [145] Sowalsky AG, Xia Z, Wang L, Zhao H, Chen S, Bubley GJ, et al. Whole transcriptome sequencing reveals extensive unspliced mRNA in metastatic castration-resistant prostate cancer. Mol Cancer Res 2014. [146] Kumar-Sinha C, Tomlins SA, Chinnaiyan AM. Recurrent gene fusions in prostate cancer. Nat Rev Cancer 2008;8:497–511. [147] Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 2005;310:644–8. [148] Hermans KG, Boormans JL, Gasi D, van Leenders GJ, Jenster G, Verhagen PC, et al. Overexpression of prostate-specific TMPRSS2(exon 0)-ERG fusion transcripts corresponds with favorable prognosis of prostate cancer. Clin Cancer Res 2009;15:6398–403. [149] Hollenhorst PC, Paul L, Ferris MW, Graves BJ. The ETS gene ETV4 is required for anchorage-independent growth and a cell proliferation gene expression program in PC3 prostate cells. Genes Cancer 2011;1:1044–52. [150] Perner S, Demichelis F, Beroukhim R, Schmidt FH, Mosquera JM, Setlur S, et al. TMPRSS2:ERG fusion-associated deletions provide insight into the heterogeneity of prostate cancer. Cancer Res 2006;66:8337–41. [151] Park K, Tomlins SA, Mudaliar KM, Chiu YL, Esgueva R, Mehra R, et al. Antibody-based detection of ERG rearrangement-positive prostate cancer. Neoplasia 2010;12:590–8. [152] van Leenders GJ, Boormans JL, Vissers CJ, Hoogland AM, Bressers AA, Furusato B, et al. Antibody EPR3864 is specific for ERG genomic fusions in prostate cancer: implications for pathological practice. Mod Pathol 2011;24:1128–38. [153] Demichelis F, Fall K, Perner S, Andren O, Schmidt F, Setlur SR, et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 2007;26:4596–9. [154] Rickman DS, Soong TD, Moss B, Mosquera JM, Dlabal J, Terry S, et al. Oncogene-mediated alterations in chromatin conformation. Proc Natl Acad Sci USA 2012;109:9083–8. [155] Esgueva R, Perner S, J LaFargue C, Scheble V, Stephan C, Lein M, et al. Prevalence of TMPRSS2-ERG and SLC45A3-ERG gene fusions in a large prostatectomy cohort. Mod Pathol 2010;23:539–46. [156] Mani RS, Tomlins SA, Callahan K, Ghosh A, Nyati MK, Varambally S, et al. Induced chromosomal proximity and gene fusions in prostate cancer. Science 2009;326:1230. [157] Zhang Y, Gong M, Yuan H, Park HG, Frierson HF, Li H. Chimeric transcript generated by cis-splicing of adjacent genes regulates prostate cancer cell proliferation. Cancer Discov 2012;2:598–607. [158] Rickman DS, Pflueger D, Moss B, VanDoren VE, Chen CX, de la Taille A, et al. SLC45A3-ELK4 is a novel and frequent erythroblast transformation-specific fusion transcript in prostate cancer. Cancer Res 2009;69:2734–8. [159] Mehra R, Tomlins SA, Yu J, Cao X, Wang L, Menon A, et al. Characterization of TMPRSS2-ETS gene aberrations in androgen-independent metastatic prostate cancer. Cancer Res 2008;68:3584–90. [160] Gsponer JR, Braun M, Scheble VJ, Zellweger T, Bachmann A, Perner S, et al. ERG rearrangement and protein expression in the progression to castrationresistant prostate cancer. Prostate Cancer Prostatic Dis 2014;17:126–31. [161] Casey OM, Fang L, Hynes PG, Abou-Kheir WG, Martin PL, Tillman HS, et al. TMPRSS2-driven ERG expression in vivo increases self-renewal and maintains expression in a castration resistant subpopulation. PLoS ONE 2012;7:e41668. [162] Iljin K, Wolf M, Edgren H, Gupta S, Kilpinen S, Skotheim RI, et al. TMPRSS2 fusions with oncogenic ETS factors in prostate cancer involve unbalanced genomic rearrangements and are associated with HDAC1 and epigenetic reprogramming. Cancer Res 2006;66:10242–6. [163] Boormans JL, Korsten H, Ziel-van der Made AJ, van Leenders GJ, de Vos CV, Jenster G, et al. Identification of TDRD1 as a direct target gene of ERG in primary prostate cancer. Int J Cancer 2013;133:335–45. [164] Brase JC, Johannes M, Mannsperger H, Falth M, Metzger J, Kacprzyk LA, et al. TMPRSS2-ERG-specific transcriptional modulation is associated with prostate cancer biomarkers and TGF-beta signaling. BMC Cancer 2011;11:507. [165] Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 2010;18:11–22. [166] Burdova A, Bouchal J, Tavandzis S, Kolar Z. TMPRSS2-ERG gene fusion in prostate cancer. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014;158:502–10. [167] Tangutoori S, Baldwin P, Sridhar S. PARP inhibitors: a new era of targeted therapy. Maturitas 2015.
Please cite this article in press as: Katsogiannou M et al. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev (2015), http://dx.doi.org/ 10.1016/j.ctrv.2015.05.003