Abstract
Most prostate cancers initially respond to androgen deprivation therapy (ADT). With the long-term application of ADT, localized prostate cancer will progress to castration-resistant prostate cancer (CRPC), metastatic CRPC (mCRPC), and neuroendocrine prostate cancer (NEPC), and the transcriptional network shifted. Forkhead box protein A1 (FOXA1) may play a key role in this process through multiple mechanisms. To better understand the role of FOXA1 in prostate cancer, we review the interplay among FOXA1-targeted genes, modulators of FOXA1, and FOXA1 with a particular emphasis on androgen receptor (AR) function. Furthermore, we discuss the distinct role of FOXA1 mutations in prostate cancer and clinical significance of FOXA1. We summarize possible regulation pathways of FOXA1 in different stages of prostate cancer. We focus on links between FOXA1 and AR, which may play different roles in various types of prostate cancer. Finally, we discuss FOXA1 mutation and its clinical significance in prostate cancer. FOXA1 regulates the development of prostate cancer through various pathways, and it could be a biomarker for mCRPC and NEPC. Future efforts need to focus on mechanisms underlying mutation of FOXA1 in advanced prostate cancer. We believe that FOXA1 would be a prognostic marker and therapeutic target in prostate cancer.
Keywords: androgen receptor, forkhead box protein A1, mutation, prostate cancer
INTRODUCTION
Prostate cancer is the second leading cause of estimated cancer deaths and accounted for 21% of all cases among men in 2020.1 Prostate cancer has the highest 5-year survival rate for all stages combined among all cancers.1 Although the overall incidence of prostate cancer has declined, distant-stage diagnoses have increased.2 Men diagnosed with primary prostate cancer are usually treated with curative treatment, and more than 30% of patients progress to metastatic disease within 10 years.3,4 Androgen deprivation therapies (ADTs) are then used to treat metastatic disease and have an initial response rate of approximately 80%.5,6 Despite the efficacy of ADT, patients ultimately develop resistance and progress to metastatic castration-resistant prostate cancer (mCRPC).5,7 Notably, Asian patients usually present with higher tumor grades in prostate cancer at diagnosis compared with Western populations.8 Therefore, there remains a need to understand the genomic underpinnings of prostate cancer development.
Forkhead box protein A1 (FOXA1), also known as hepatocyte nuclear factor 3-alpha (HNF3A), was first identified as a transcription factor for the expression of transthyretin and α1-antitrypsin in the liver.9 FOXA1 was found to collaborate with NK2 homeobox 1 (NKX2-1) and functioned as an oncogene in lung cancer.10 Metastasis of pancreatic ductal adenocarcinoma was linked with FOXA1-dependent enhancer reprogramming.11 In the past few years, FOXA1 expression has been reported to be related to several human cancers, as both an oncogene and a tumor suppressor gene depending on the specific cancer types.12,13 Even in the same type of cancer, FOXA1 can play different roles in different subtypes.14,15
Recent findings have shown that FOXA1 plays an important role in regulating steroid receptor functions.16 Prostate cancer and breast cancer are the two most prominent hormone-related tumors, so they are not surprisingly related to FOXA1. As FOXA1 is obligatory for androgen receptor (AR)-mediated genes for CRPC, it is thought to play an important role in prostate cancer progression.17 Recent studies revealed that the genetic mutation frequency of FOXA1 is increased in mCRPC patients, suggesting that FOXA1 mutations may be a factor leading to aggressive prostate cancer.18,19 FOXA1 was also found to be the transcription factor (TF) motif most frequently enriched in prostate cancer-specific enhancer-promoter loops.20 Notably, FOXA1 mutations were found in 41% of a Chinese cohort, whereas FOXA1 was mutated in only 4% of prostate cancers in The Cancer Genome Atlas (TCGA) database.21,22 This review focuses on the possible roles of FOXA1 in prostate cancer initiation, progression, and drug resistance and as a new therapeutic target.
FOXA1 IN THE DEVELOPMENT OF THE PROSTATE
The secretory function of the prostate is dependent on epithelial AR expression, whereas morphogenesis depends on stromal AR expression.23,24 Considering the established role of FOXA1 in regulating AR transcriptional activity in the prostate, it is not surprising that FOXA1 is involved in prostate development.25 FOXA1, also known as HNF3A, was first identified as a necessary transcription factor for α1-antitrypsin and transthyretin in the liver.9 Interestingly, FOXA1 mRNA expression levels in the prostate are much higher than those in liver tissues.26 Expression studies on the prostate showed that FOXA1 is expressed in epithelial cells but not in the stromal cells.26,27 By analyzing Foxa1-deficient mouse prostates, Gao et al.28 found an altered ductal pattern, which indicated that Foxa1 promoted epithelial cell maturation and regulated early ductal pattern formation. As AR has separate and distinct functions in the stroma and epithelium, Foxa1 probably also plays different roles in different stages. In addition to its function in ductal morphogenesis and cytodifferentiation, Foxa1 has a secretory function as well.29 Biochemical analysis confirmed similar function of FOXA1 in humans.29 In summary, FOXA1 plays a distinct role in prostate morphogenesis, differentiation and secretion.
FOXA1 AND AR IN PROSTATE CANCER
Androgens play an important role in the development and maturation of the prostate, influencing the differentiation and proliferation of the epithelial cells.30 AR, activated by androgens, is a key mediator of targeted genes that regulate the development and differentiation of the luminal epithelium. AR plays a crucial role in the growth and maintenance of the prostate gland. Mice with AR function defects do develop prostate gland or prostate cancer.31,32 Clinical evidence has shown that AR signaling participates in all stages of prostatic disease, including advanced and metastatic prostate cancer.33,34 In addition, the development of CRPC also requires AR signaling.
Regulation of AR by FOXA1
Different AR-binding sites (ARBs) were identified in tumor tissues and benign/normal tissues.35 This difference between tumor and benign tissues in ARB preference is called the AR malignancy shift (AMS). FOXA1 was found at more than 60% of ARBs driving the transcriptional programs in prostate cancer cells and was considered one of the components of the AMS.36,37 Generally, FOXA1 serves as a pioneer factor for AR. It can bind to target sites in silent chromatin and trigger regulation in the genome before other transcription factors, which is why FOXA1 is called “pioneer factor”.38 FOXA1 is considered an auxiliary factor for AR to promote prostate cancer progression. However, genome-wide elucidation of the AR cistrome in prostate cancer cell lines showed approximately 70% overlap between FOXA1 and AR-binding sites, indicating an important role of FOXA1 in the transcriptional regulation of prostate cancer.39 Sahu et al.39 identified three distinct classes of ARBs and AR-regulated transcription programs: (1) the sites independent of FOXA1, (2) the sites pioneered by FOXA1 to recruit for AR, and (3) the sites that are masked by FOXA1 and activated by AR only when FOXA1 was depleted. Moreover, an in vitro experiment showed that not only did FOXA1 depletion not reduce the ARBs, but it could also result in an increased number of ARBs.12,39 Furthermore, FOXA1 was reported to inhibit the transcriptional activity of AR in prostate cancer cell lines possibly by competing with AR coactivators.40 These experiments showed the two-way regulation of AR by FOXA1, depending on the cellular context.12
Relationship between FOXA1 and AR in different types of prostate cancer
In primary prostate cancer tissue, FOXA1 was found to promote cell proliferation, and high FOXA1 expression was related to high AR expression.41 FOXA1 expression was also reported to be positively associated with AR and lymph node metastases in a cohort study of prostate cancer.42 In CRPC, FOXA1 was found to be a key regulator of the cell cycle, as it could promote G2 to M-phase transition and G1 to S-phase transition through two distinct pathways and its activity was reported to be essential for AR function.17,43 However, AR was also reported to continuously bind some specific genomic loci that are open to chromatin structures and independent of FOXA1 in CRPC.44 Furthermore, previous studies reported that FOXA1 was downregulated in neuroendocrine prostate cancer (NEPC) and CRPC and showed negative correlation with AR.12,45,46
Previous studies showed that FOXA1 silencing did not change the expression of prostate-specific antigen (PSA) and transmembrane protease serine 2 (TMPRSS2), which are key factors in androgen signaling, suggesting that FOXA1 may not play a crucial role in AR signaling of AR-dependent prostate cancer cells.38,47 Interestingly, another study showed a reduction in TMPRSS2 after suppressing FOXA1 expression in androgen-independent cell lines.48 FOXA1 is also required for chromatin looping and enhancer activity by regulating TMPRSS2 and long noncoding RNA (lncRNA) prostate cancer-associated transcript 38 (PRCAT38).49 Regulation of some AR-specific target genes is dependent on FOXA1, whereas regulation of AR-V7-specific target genes is independent of FOXA1.50,51 Paakinaho et al.52 found that androgens could induce the chromatin binding of FOXA1 in prostate cancer cell lines independent of AR. Taken together, these results illustrate multiple functions of FOXA1 regulating AR transcriptional activity through various mechanisms. It also reveals the possible different mechanisms of FOXA1 between AR-dependent and AR-independent prostate cancer.
THE FOXA1 REGULATORY CIRCUIT IN PROSTATE CANCER
Regulation of FOXA1 DNA binding by histone modification
FOXA1 is typically recruited in the epigenetic signature consisting of lysine 4 on histone 3 (H3K4) into functional regulatory elements.45 Removal of mono- and di-methylated H3K4 may lead to suppression of FOXA1 and AR recruitment.53,54 As FOXA1 has the ability to bind nucleosomes, different binding sites to chromatin of FOXA1 are dependent on the methylation and distribution of H3K4.53 Acetylation of lysine 27 on histone 3 (H3K27ac), another histone marker of active enhancers, could be downregulated by FOXA1.49 Moreover, FOXA1 binding at the enhancers could be impaired by inhibiting the transcription coactivator p300.49 Wang et al.12 detected three classes of AR-binding events after hormone stimulation, indicating that in the absence of FOXA1, histone acetyltransferase was needed for initiating AR transcriptional events. Collectively, these recent studies have begun to elucidate the specific control mechanisms that recruit FOXA1 to chromatin.
Targets of FOXA1
NK3 homeobox 1 (NKX3-1) is known as the earliest prostate marker.55 In prostate epithelial cells of mice, null Foxa1 prostate showed decreased NKX3-1 expression.28 As FOXA1, GATA binding protein 3 (GATA3), and estrogen receptor alpha (ERα) form a network regulating the development of luminal breast epithelial cells, GATA3 was thought to be required for FOXA1.56,57 GATA3 was necessary for prostate function in adult mice.58 However, GATA3 was not expressed in prostate cancer cell lines at the mRNA level.59 Moreover, GATA3 expression was reported to be completely negative in both primary and metastatic prostate cancer tissues tested by immunohistochemistry.42 FOXA1 loss was observed to lead to obviously high expression of transforming growth factor-beta 3 (TGFB3), which is a key factor in the TGF-β pathway in multiple prostate cancer cell lines.19 Chromatin immunoprecipitation followed by sequencing (ChIP-seq) analysis showed that FOXA1 inhibits TGFB3 expression by binding to its enhancer.19 FOXA1 depletion upregulated TGFB3, which was found to activate the TGF-β pathway and induce cell invasion and epithelial–mesenchymal transition (EMT).19 FOXA1 could also directly inhibit interleukin 8 (IL-8) transcription and snail family transcriptional repressor 2 (SNAI2) expression.46,60 In addition, FOXA1 controlled cell proliferation by inhibiting insulin-like growth factor binding protein 3 (IGFBP-3) through the mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) pathways.41 FOXA1 was significantly enriched in a chromatin region binding with N-Myc, and FOXA1 binding was related to androgen-dependent changes in N-Myc binding.61 It was also found that FOXA1 upregulated the expression of lncRNAs such as PRCAT38 and DSCAM antisense RNA 1 (DSCAM-AS1) by binding their promoters.37,62 Recently, FOXA1 was found to increase the production of proangiogenic factors, including endoglin, endothelin-1, and epidermal growth factor (EGF).63 In addition to being an oncogene, FOXA1 has also been found to enhance chemotherapy and immunotherapy resistance by suppressing signal transducer and activator of transcription 2 (STAT2) DNA-binding activity by binding the STAT2 DNA-binding domain.64
Regulation of FOXA1
It has been reported that nuclear factor I C (NFIC), TLE family member 3 (TLE3), and histone deacetylase (HDAC7) act as FOXA1 corepressor genes regulating breast cancer cell invasion.65,67,68 Wang et al.69 also found that knockdown of NFIC, TLE3, and HDAC7 promoted FOXA1-repressed invasion genes in LNCaP cell lines. Moreover, the small molecule JQ1 could promote prostate cancer cell invasion by directly interacting with FOXA1 and keeping FOXA1 from binding to its corepressor genes, such as NFIC, TLE3, and HDAC7, thereby activating invasive-related genes.69 In terms of posttranslational modification, histone lysine-specific demethylase 1 (LSD1) was found to increase the chromatin binding of FOXA1 by demethylating lysine 270.70 In addition, enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) could increase the stability of FOXA1 by methylating FOXA1 protein at lysine 295.71 Octamer-binding transcription factor 4 (OCT4) can activate the FOXA1/AR axis by promoting complex formation.72 Clinical evidence showed that hyperglycemia could induce high expression of FOXA1 and IGFBP-2.73 In addition, negative regulation between FOXA1 and IGFBP-2 was observed in normal epithelial cells but not in cancer cells.73 A positive association was observed between IGFBP-2 and FOXA1 in prostate cancer tissues, and the FOXA1/IGFBP-2 axis played an important role in high glucose-induced EMT in prostate cancer cells.73 Poly(ADP-ribose) polymerase-2 (PARP-2) is a member of the PARP family, which is responsible for multiple DNA damage repair pathways.74 As PARP-2 showed higher expression levels in prostate cancer tissue than in benign prostate tissue, PARP-2 was found to enhance AR-mediated transcription in prostate cancer by interacting with FOXA1.75 In addition, PARP-2 was found to have no interaction with AR by coimmunoprecipitation (Co-IP) in prostate cancer cell lines.75 Taken together, the inhibition of PARP-2 could disrupt the function of FOXA1 in AR transcription. Analysis of a TCGA cohort showed that fatty acid-binding protein 5 (FABP5) overexpression was related to FOXA1 mutations in e-twenty-six (ETS)-negative subtypes.76 The transcription factor ONECUT2 (OC2) was identified as a key factor in mCRPC.77 Endogenous OC2 was found to bind the FOXA1 promoter and gene body to inhibit its expression.77 The silencing of nuclear export protein exportin-1 (XPO1) inhibited the FOXA1 expression.78
Cis-regulatory elements (CREs) are regions of noncoding DNA that play a critical role in regulating gene expression by binding to transcription factors.79 Six CREs in the FOXA1 regulatory plexus were identified, and their repression led to significantly decreased FOXA1 expression.80 Notably, microRNA-194 (miR-194) was found to decrease the levels of FOXA1 protein and mRNA in prostate cancer cell lines and was also negatively correlated with FOXA1 in clinical specimens.81
A recent study showed that ectopic expression of achaete-scute family BHLH transcription factor 1 (ASCL1) and NKX2-1 in prostate adenocarcinoma cells could reprogram FOXA1 to bind to neuroendocrine elements and promote the development of NEPC.82 The FOXA1 regulatory circuit is summarized in Table 1.
Table 1.
Modulators of forkhead box protein A1 function in prostate cancer
| Gene | Function | Expression in prostate cancer | Reference |
|---|---|---|---|
| FOXA1-regulated genes | |||
| TMPRSS2 | Induced | Increased | 49 |
| PRCAT38 | Induced | Increased | 37 |
| NKX3-1 | Induced | Increased | 28 126 |
| DSCAM-AS1 | Induced | Increased | 62 |
| TGFB3 | Repressed | NA | 19 |
| IL-8 | Repressed | NA | 60 |
| SNAI2 | Repressed | NA | 46 |
| IGFBP-3 | Repressed | NA | 41 |
| AR | Repressed | NA | 40 |
| STAT2 | Repressed | NA | 64 |
| Genes involved in FOXA1 expression | |||
| TLE3 | Corepressor | NA | 69 |
| HDAC7 | Corepressor | NA | 69 |
| NFIC | Corepressor | NA | 69 |
| MYCN | Collaborator | NA | 61 |
| Genes/proteins that may control FOXA1 expression | |||
| H3K4me1 | Promoting | NA | 45 |
| H3K4me2 | Promoting | NA | 45 |
| H3K27ac | Promoting | NA | 82 |
| OC2 | Increased binding | Increased | 77 |
| LSD1 | Increased binding | Increased | 70 |
| OCT4 | Increased binding | NA | 72 |
| XPO1 | Repression | Increased | 78 |
| miR-194 | Repression | Increased | 81 |
| NKX2-1 | Reprogramming | NA | 82 |
| ASCL1 | Reprogramming | NA | 82 |
| EZH2 | Increased stability | NA | 71 |
FOXA1: forkhead box protein A1; TMPRSS2: transmembrane protease serine 2; PRCAT38: prostate cancer-associated transcript 38; NKX3-1: NK3 homeobox 1; DSCAM-AS1: DSCAM antisense RNA 1; TGFB3: transforming growth factor-beta 3; IL-8: interleukin 8; SNAI2: snail family transcriptional repressor 2; IGFBP-3: insulin-like growth factor binding protein 3; AR: androgen receptor; STAT2: signal transducer and activator of transcription 2; TLE3: TLE family member 3; HDAC7: histone deacetylase; NFIC: nuclear factor I C; OC2: ONECUT2; LSD1: histone lysine-specific demethylase 1; OCT4: octamer-binding transcription factor 4; XPO1: nuclear export protein exportin-1; miR-194: microRNA-194; NKX2-1: NK2 homeobox 1; ASCL1: achaete-scute family BHLH transcription factor 1; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; NA: not available
Mutation of FOXA1
Mutations, including losses of phosphatase and tensin homolog (PTEN), NKX3-1, and ETS fusion, were commonly identified.83,84,85 Recently, some new mutations were found including mediator complex subunit 12 (MED12), FOXA1, and speckle-type BTB/POZ protein (SPOP).86 FOXA1 mutation was found in 4%–9% of primary prostate tumors and in 12%–13% of mCRPCs in a Western cohort, suggesting that a high frequency of FOXA1 mutation may be related to more aggressive prostate cancer type.86,87 However, a recent study found that FOXA1 mutation was found in 41% of Chinese cohort in clinical localized prostate cancer, indicating the possibility of different mechanisms underlying the prostate cancer progression.21 Moreover, a cohort of African Americans showed an 8% mutation of FOXA1 in primary prostate tumors.88
Most FOXA1 mutations were found located in the Wing2 region of forkhead (FKHD). Genomic analysis showed that FOXA1 mutations around residue 253 in the FKHD domain caused prostate cancer cell growth under hormone-deprived conditions, leading to CRPC.89 Missense mutations at the winged-helix FKHD of FOXA1 reduced the dependency of prostate cancer cell on AR signaling and promoted prostate cancer progression.90 Moreover, TCGA studies revealed that FOXA1 mutations promoted AR-dependent transcriptional activities.22
Parolia et al.87 mapped FOXA1 mutations into two structural patterns: (1) indel, in-frame insertion, and missense mutations at FKHD; and (2) truncating frameshift mutations restricted to the regulatory domain. According to FOXA1 structure, FOXA1 mutations were categorized into three classes: class 1, which comprises all mutations within the FKHD in early prostate cancer; class 2, which includes mutations in the C-terminal domain of FKHD mainly in metastatic prostate cancers; and class 3, which comprises duplications and translocations within the FOXA1 locus mostly in mCRPC.87 Adams et al.91 defined two hotspots in the FKHD domain from 3086 human prostate cancers: (1) wing2 mutations that are detected mostly in adenocarcinomas at all stages; and (2) the highly conserved DNA-contact residue R219, which is enriched mostly in NEPC with metastatic tumors (Figure 1). Mutations of FOXA1 can increase, replace, and reprogram AR function to promote growth of prostate cancer (Figure 2).
Figure 1.

Structure of FOXA1 and classes of FOXA1 alterations. FOXA1: forkhead box protein A1; RD: regulatory domain.
Figure 2.

FOXA1 and AR-regulated pathway in AR-dependent prostate cancer and AR-independent prostate cancer. (a) Class 1 mutant FOXA1/wild-type FOXA1, opens the closed chromatin and recruits AR to promote oncogenic AR signaling. (b) In AR-independent prostate cancer, mutant FOXA1/wild-type FOXA1 reprograms or replaces AR function, and promotes AR-independent growth. FOXA1: forkhead box protein A1; Wnt: wingless-type MMTV integration site family; AR: androgen receptor; ARE: androgen-receptor response element; EMT: epithelial–mesenchymal transition.
Insertions and deletions (indels) in prostate cancer remain largely unexplored due to technical limitations.92 Notably, in the Chinese Prostate Cancer Genome and Epigenome Atlas (CPGEA), indels account for 61.8% of FOXA1 mutations.21 In-frame indel and missense of FOXA1 mutations were found to affect the nicotinate and nicotinamide metabolism pathways.21 With the application of new technologies, somatic FOXA1 untranslated region (UTR) mutations have been identified with high specificity.93
FOXA1 IN NEUROENDOCRINE PROSTATE CANCER
Advanced prostate cancer is commonly treated with ADT, and most patients finally develop androgen resistance and develop CRPC.94 New generation AR antagonists with higher affinity have been used for CRPC. Under treatment with these AR antagonists (e.g., abiraterone and enzalutamide), some CRPC patients develop into therapy-induced NEPC (t-NEPC).95,96,97
Studies have shown that lineage plasticity plays a critical role in prostate cancer therapy resistance.95 N-Myc (encoded by MYCN), a transcription factor, is necessary for neurodevelopment and is overexpressed in retinoblastoma, neuroblastoma, and glioblastoma multiforme.98,99,100,101,102,103 Interestingly, N-Myc is not found in the epithelial lineage of prostate tissue but is overexpressed in some CRPCs and a majority of NEPC.104,105 Genomic locus binding comparison revealed the nearby binding of N-Myc and FOXA1.61 Overlap between FOXA1 and N-Myc was also confirmed, and FOXA1 played an important role in regulating N-Myc binding.61 In addition, mutations of FOXA1 play an important role in the neuroendocrine process, and two different R219 mutants of FOXA1 blocked luminal differentiation and activated neuroendocrine transcription.91 More recently, miR-194 was reported to promote the neuroendocrine phenotype by decreasing FOXA1 binding.81 FOXA2, another member of the FOXA family, was found to be a NEPC TF.106,107 However, Baca et al.82 analyzed NEPC patient-derived xenografts (PDXs) and found that FOXA1 was a key factor for NEPC even if there was no FOXA1 mutation or no FOXA2 expression. FOXA1 plays a completely different role in prostate adenocarcinoma and NEPC. FOXA1 was reprogrammed to neuroendocrine-specific regulatory elements in NEPC. Possible regulation pathways are summarized in Figure 3.
Figure 3.
Pathways of FOXA1 involved in different types of prostate cancer. EMT: epithelial–mesenchymal transition; IFN: interferon; CRPC: castration-resistant prostate cancer; NEPC: neuroendocrine prostate cancer; FOXA1: forkhead box protein A1; TMPRSS2: transmembrane protease serine 2; PRCAT38: prostate cancer-associated transcript 38; NKX3-1: NK3 homeobox 1; DSCAM-AS1: DSCAM antisense RNA 1; TGFB3: transforming growth factor-beta 3; IL-8: interleukin 8; SNAI2: snail family transcriptional repressor 2; IGFBP-3: insulin-like growth factor binding protein 3; AR: androgen receptor; STAT2: signal transducer and activator of transcription 2; TLE3: TLE family member 3; HDAC7: histone deacetylase; NFIC: nuclear Factor I C; OC2: ONECUT2; LSD1: histone lysine-specific demethylase 1; OCT4: octamer-binding transcription factor 4; XPO1: nuclear export protein exportin-1; miR-194: microRNA-194; NKX2-1: NK2 homeobox 1; ASCL1: achaete-scute family BHLH transcription factor 1; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; EGF: epidermal growth factor; AR: androgen receptor; lncRNA: long noncoding RNA; miRNA: microRNA; CHGA: chromogranin A.
CLINICAL RELEVANCE OF FOXA1
The prognostic role of FOXA1 in many different cancers has been widely explored.86,108,109 Studies on the relationship of FOXA1 with prognosis in prostate cancer have shown controversial results.110 High FOXA1 expression in primary prostate cancer showed poor disease-specific survival in 350 patients with a hazard ratio (HR) of 2.89 (95% confidence interval [CI]: 1.02–8.21).39 In addition, FOXA1 expression levels were significantly associated with Gleason score and pT stage in primary prostate cancer.41,111 Notably, patients with high AR expression and low FOXA1 expression had better prognosis than patients with high expression of both AR and FOXA1.39 Another cohort study of 102 patients also showed that a high expression level of FOXA1 predicted poor biochemical recurrence (HR: 5.0, 95% CI: 1.2–21.1, P = 0.028).112 Other studies showed that high FOXA1 expression was related to poor disease-free survival, but FOXA1 expression was lower in metastatic tumors.12,113,114 Based on serum PSA, high FOXA1 expression was found to be related to shorter relapse-free survival.111 An overview of the different roles of FOXA1 in prostate cancer is provided in Table 2.
Table 2.
Overview of different roles of forkhead box protein A1 in prostate cancer
| Study | Cancer type | Expression relative to normal | Expression relative to primary cancer | Clinical relevance of high FOXA1 expression | Method | Activity |
|---|---|---|---|---|---|---|
| Mirosevich et al.25 2006 | Adenocarcinoma, neuroendocrine small cell carcinoma | No difference | NA | NA | IHC | Unclear |
| Sahu et al.39 2011 | Primary prostate cancer | Increased | NA | Poor cancer-specific survival | IHC | Oncogenic |
| Wang et al.12 2011 | Metastatic prostate cancer | NA | Decreased in metastatic tumor | Better cancer-specific survival | PCR | Tumor suppressive |
| Jain et al.42 2011 | Metastatic prostate cancer | NA | Increased in metastatic lesion | More metastasis, high Gleason score, high PSA, and more angiolymphatic invasion | IHC | Oncogenic |
| Imamura et al.41 2012 | Primary prostate cancer | Increased | NA | Poor biochemical recurrence-free survival | IHC | Oncogenic |
| Gerhardt et al.111 2012 | Primary prostate cancer | Increased | NA | Poor progression-free survival | IHC | Oncogenic |
| Metastatic prostate cancer | NA | Increased in metastatic tumor | Poor progression-free survival | IHC | Oncogenic | |
| CRPC | NA | Increased | Poor progression-free survival | IHC | Oncogenic | |
| Jin et al.46 2013 | Primary prostate cancer | Slightly increased | NA | NA | PCR | Unclear |
| Metastatic prostate cancer | Decreased | Decreased in metastatic tumor | Inhibited prostate cancer metastasis | PCR | Tumor suppressive | |
| Robinson et al.112 2014 | Primary prostate cancer | Increased | NA | Poor biochemical recurrence-free survival | IHC | Oncogenic |
| Tsourlakis et al.116 2017 | Primary prostate cancer | Increased | NA | Poor PSA recurrence-free survival | IHC | Oncogenic |
| Mansor et al.73 2020 | Primary prostate cancer | No difference | NA | NA | IHC | Unclear |
FOXA1: forkhead box protein A1; IHC: immunohistochemistry; PCR: polymerase chain reaction; CRPC: castration-resistant prostate cancer; PSA: prostate-specific antigen; NA: not available
ETS-related gene (ERG) status and TMPRSS2:ERG rearrangement are commonly observed in prostate cancer. In a small-sized cohort study, no significant association was found between ERG expression and prognosis of prostate cancer.115 High FOXA1 expression was found to be related to worse recurrence-free survival in ERG-negative prostate cancer but not in ERG-positive prostate cancer.116
Moreover, another cohort study from Spain found that FOXA1 or SPOP1 mutations were more common in Gleason grade group 5 and were associated with PSA recurrence in ERG wild-type tumors.117 However, a cohort study of African Americans failed to show an association between FOXA1 mutation and metastasis (HR: 0.34, 95% CI: 0.04–2.76, P = 0.310).88 The HRs and 95% CIs for tumor progression survival in association with FOXA1 mutation are listed in Table 3. Notably, FOXA1 3’-UTR mutations were found in early-stage prostate cancer.93 Combined with other databases, FOXA1 3’-UTR indel mutations were commonly seen in both primary and metastatic prostate cancers.118 A cohort study of 202 mCRPC patients also revealed that FOXA1 3’-UTR indel mutations did not influence OS from mCRPC treatment initiation and PFS on first-line abiraterone or enzalutamide therapy.119 Although FOXA1 mutations were also commonly observed in breast cancer and bladder cancer, the FOXA1 3’-UTR mutations were not detected in these cancers, suggesting that FOXA1 UTR indels could be a specific marker for the diagnosis and screening of prostate cancer.93,120
Table 3.
Clinical relevance of forkhead box protein A1 mutation
| Study | Cancer progression | Univariate analysis | Multivariate analysis | Clinical relevance | Main population | ||
|---|---|---|---|---|---|---|---|
|
|
|
||||||
| HR (95% CI) | P | HR (95% CI) | P | ||||
| Adams et al.91 2019 | Progression to biochemical recurrence | 2.92 (2.12–4.02) | 0.0013 | NA | NA | Significant worse in FOXA1 mutation group | Caucasian |
| Progression to metastatic disease | 4.59 (3.17–6.64) | 0.0001 | NA | NA | Significant worse in FOXA1 mutation group | Caucasian | |
| Hernández-Llodrà et al.117 2019 | Progression to biochemical recurrence | NA | 0.0009 | 5.76 (1.27–26.06) | 0.023 | Significant worse in FOXA1 mutation group | Caucasian |
| Faisal et al.88 2020 | Progression to metastatic disease | 0.57 (0.08–4.29) | 0.586 | 0.34 (0.04–2.76) | 0.31 | Better in FOXA1 mutation group but not significant | African American |
FOXA1: forkhead box protein A1; HR: hazard ratio; CI: confidence interval; NA: not available
CONCLUSIONS
While the role of AR in promoting prostate cancer progression has been established, recent studies have focused on the coregulators of AR such as FOXA1. As relationships between FOXA1 and AR have been widely researched, FOXA1 may play an independent role in the carcinogenesis of prostate cancer. The FOXA1 expression level in primary prostate cancer tissue is high and differs in various subtypes of prostate cancer. High FOXA1 expression usually predicts poor prognosis in prostate cancer. FOXA1 regulates the expression of multiple genes, especially AR in the development and progression of localized prostate cancer, metastatic prostate cancer, CRPC, and NEPC. Both primary and metastatic prostate cancers usually respond well to initial ADT, while treatment for mCRPC and NEPC patients is difficult. However, FOXA1 was found to be downregulated in NEPC and CRPC, indicating different mechanisms in different types of prostate cancers. In addition, the types of mutations in FOXA1 varied among early-stage prostate cancer, CRPC, and NEPC. Notably, the mutation rate of FOXA1 is higher in metastatic prostate cancer and NEPC than that in primary prostate cancer. Given the oncogenic activity of FOXA1 in both AR-independent and AR-dependent prostate cancers, FOXA1 could be a therapeutic target in prostate cancers, including mCRPC and NEPC.121 In addition, FOXA1 could be a biomarker for mCRPC and NEPC. Future efforts need to focus on the mechanisms underlying the mutation of FOXA1 in advanced prostate cancer.
Some significant barriers in the molecular mechanism should be considered. We usually thought that high-frequency mutations of FOXA1 were markers of metastatic prostate cancer or NEPC. However, a recent Chinese cohort study showed that FOXA1 was the most highly mutated gene (41%) in primary prostate cancer.21 Ethnic factors must be considered in the subsequent studies of FOXA1 as they may be involved in different molecular pathways. Moreover, it was reported that FOXA1 was observed in all types of prostate cancers, while FOXA2 was only detected in NEPC and advanced prostate cancer.25 In particular, FOXA2 may be a factor promoting androgen-independent prostate cancer.25 This result suggested that members of the FOXA family may play a crucial role in prostate cancer. Future studies should focus on the specific gene regulation between FOXA1 and tumor progression. Some drugs targeting gene mutations have been discovered. The mutation frequency of SPOP is even higher than that of FOXA1 in primary prostate cancer and advanced prostate cancer, and SPOP-mutated mCRPC appears to be highly sensitive to abiraterone.22,86,122 The PARP inhibitor olaparib has been used for prostate cancer patients with BRCA1 DNA repair-associated (BRCA1) and BRCA2 DNA repair-associated (BRCA2) mutations.123 Although there are currently no specific inhibitors against FOXA1, recent technologies have led to nucleotide-based inhibitors through antagonizing transcriptional programs by multiple pathways.124,125 In conclusion, we believe that FOXA1 could be a prognostic marker and therapeutic target in various stages of prostate cancer.
AUTHOR CONTRIBUTIONS
CL and JL conceived the structure of the manuscript and revised the manuscript. HYD, TRZ, and TY drafted the initial manuscript. HYD and LD made the figures and tables. All authors read and approved the final manuscript.
COMPETING INTERESTS
All authors declared no competing interests.
ACKNOWLEDGMENT
This work was supported by the National Natural Science Foundation of China (82002718); and the Jiangsu Natural Science Foundation (BK20191077).
REFERENCES
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30. doi: 10.3322/caac.21590. [DOI] [PubMed] [Google Scholar]
- 2.Negoita S, Feuer EJ, Mariotto A, Cronin KA, Petkov VI, et al. Annual report to the nation on the status of cancer, part II:recent changes in prostate cancer trends and disease characteristics. Cancer. 2018;124:2801–14. doi: 10.1002/cncr.31549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Boorjian SA, Thompson RH, Siddiqui S, Bagniewski S, Bergstralh EJ, et al. Long-term outcome after radical prostatectomy for patients with lymph node positive prostate cancer in the prostate specific antigen era. J Urol. 2007;178:864–70. doi: 10.1016/j.juro.2007.05.048. [DOI] [PubMed] [Google Scholar]
- 4.Litwin MS, Tan HJ. The diagnosis and treatment of prostate cancer:a review. JAMA. 2017;317:2532–42. doi: 10.1001/jama.2017.7248. [DOI] [PubMed] [Google Scholar]
- 5.Attard G, Parker C, Eeles RA, Schröder F, Tomlins SA, et al. Prostate cancer. Lancet. 2016;387:70–82. doi: 10.1016/S0140-6736(14)61947-4. [DOI] [PubMed] [Google Scholar]
- 6.Salonen AJ, Viitanen J, Lundstedt S, Ala-Opas M, Taari K, et al. Finnish multicenter study comparing intermittent to continuous androgen deprivation for advanced prostate cancer:interim analysis of prognostic markers affecting initial response to androgen deprivation. J Urol. 2008;180:915–9. doi: 10.1016/j.juro.2008.05.009. [DOI] [PubMed] [Google Scholar]
- 7.Aggarwal RR, Feng FY, Small EJ. Emerging categories of disease in advanced prostate cancer and their therapeutic implications. Oncology (Williston Park) 2017;31:467–74. [PubMed] [Google Scholar]
- 8.Kimura T. East meets West:ethnic differences in prostate cancer epidemiology between East Asians and Caucasians. Chin J Cancer. 2012;31:421–9. doi: 10.5732/cjc.011.10324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Costa RH, Grayson DR, Darnell JE., Jr Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and alpha 1-antitrypsin genes. Mol Cell Biol. 1989;9:1415–25. doi: 10.1128/mcb.9.4.1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hight SK, Mootz A, Kollipara RK, McMillan E, Yenerall P, et al. An in vivo functional genomics screen of nuclear receptors and their co-regulators identifies FOXA1 as an essential gene in lung tumorigenesis. Neoplasia. 2020;22:294–310. doi: 10.1016/j.neo.2020.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Roe JS, Hwang CI, Somerville TD, Milazzo JP, Lee EJ, et al. Enhancer reprogramming promotes pancreatic cancer metastasis. Cell. 2017;170:875–88.e20. doi: 10.1016/j.cell.2017.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wang D, Garcia-Bassets I, Benner C, Li W, Su X, et al. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature. 2011;474:390–4. doi: 10.1038/nature10006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Neben K, Schnittger S, Brors B, Tews B, Kokocinski F, et al. Distinct gene expression patterns associated with FLT3- and NRAS-activating mutations in acute myeloid leukemia with normal karyotype. Oncogene. 2005;24:1580–8. doi: 10.1038/sj.onc.1208344. [DOI] [PubMed] [Google Scholar]
- 14.Badve S, Turbin D, Thorat MA, Morimiya A, Nielsen TO, et al. FOXA1 expression in breast cancer - correlation with luminal subtype A and survival. Clin Cancer Res. 2007;13:4415–21. doi: 10.1158/1078-0432.CCR-07-0122. [DOI] [PubMed] [Google Scholar]
- 15.Hu X, Stern HM, Ge L, O’Brien C, Haydu L, et al. Genetic alterations and oncogenic pathways associated with breast cancer subtypes. Mol Cancer Res. 2009;7:511–22. doi: 10.1158/1541-7786.MCR-08-0107. [DOI] [PubMed] [Google Scholar]
- 16.Augello MA, Hickey TE, Knudsen KE. FOXA1:master of steroid receptor function in cancer. EMBO J. 2011;30:3885–94. doi: 10.1038/emboj.2011.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang Q, Li W, Zhang Y, Yuan X, Xu K, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell. 2009;138:245–56. doi: 10.1016/j.cell.2009.04.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jin HJ, Zhao JC, Wu L, Kim J, Yu J. Cooperativity and equilibrium with FOXA1 define the androgen receptor transcriptional program. Nat Commun. 2014;5:3972. doi: 10.1038/ncomms4972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Song B, Park SH, Zhao JC, Fong KW, Li S, et al. Targeting FOXA1-mediated repression of TGF-β signaling suppresses castration-resistant prostate cancer progression. J Clin Invest. 2019;129:569–82. doi: 10.1172/JCI122367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rhie SK, Perez AA, Lay FD, Schreiner S, Shi J, et al. A high-resolution 3D epigenomic map reveals insights into the creation of the prostate cancer transcriptome. Nat Commun. 2019;10:4154. doi: 10.1038/s41467-019-12079-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li J, Xu C, Lee HJ, Ren S, Zi X, et al. A genomic and epigenomic atlas of prostate cancer in Asian populations. Nature. 2020;580:93–9. doi: 10.1038/s41586-020-2135-x. [DOI] [PubMed] [Google Scholar]
- 22.Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell. 2015;163:1011–25. doi: 10.1016/j.cell.2015.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cunha GR, Donjacour AA, Cooke PS, Mee S, Bigsby RM, et al. The endocrinology and developmental biology of the prostate. Endocr Rev. 1987;8:338–62. doi: 10.1210/edrv-8-3-338. [DOI] [PubMed] [Google Scholar]
- 24.Donjacour AA, Cunha GR. Assessment of prostatic protein secretion in tissue recombinants made of urogenital sinus mesenchyme and urothelium from normal or androgen-insensitive mice. Endocrinology. 1993;132:2342–50. doi: 10.1210/endo.132.6.7684975. [DOI] [PubMed] [Google Scholar]
- 25.Mirosevich J, Gao N, Gupta A, Shappell SB, Jove R, et al. Expression and role of Foxa proteins in prostate cancer. Prostate. 2006;66:1013–28. doi: 10.1002/pros.20299. [DOI] [PubMed] [Google Scholar]
- 26.Kopachik W, Hayward SW, Cunha GR. Expression of hepatocyte nuclear factor-3α in rat prostate, seminal vesicle, and bladder. Dev Dyn. 1998;211:131–40. doi: 10.1002/(SICI)1097-0177(199802)211:2<131::AID-AJA2>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
- 27.Mirosevich J, Gao N, Matusik RJ. Expression of Foxa transcription factors in the developing and adult murine prostate. Prostate. 2005;62:339–52. doi: 10.1002/pros.20131. [DOI] [PubMed] [Google Scholar]
- 28.Gao N, Ishii K, Mirosevich J, Kuwajima S, Oppenheimer SR, et al. Forkhead box A1 regulates prostate ductal morphogenesis and promotes epithelial cell maturation. Development. 2005;132:3431–43. doi: 10.1242/dev.01917. [DOI] [PubMed] [Google Scholar]
- 29.Gao N, Zhang J, Rao MA, Case TC, Mirosevich J, et al. The role of hepatocyte nuclear factor-3 alpha (forkhead box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol Endocrinol. 2003;17:1484–507. doi: 10.1210/me.2003-0020. [DOI] [PubMed] [Google Scholar]
- 30.McDougal WS, Wein AJ, Kavoussi LR, Partin AW, Peters CA. 11th Edition Review. Philadelphia: Elsevier Health Sciences; 2015. Campbell-Walsh Urology; p. 2553. [Google Scholar]
- 31.McPhaul MJ. Androgen receptor mutations and androgen insensitivity. Mol Cell Endocrinol. 2002;198:61–7. doi: 10.1016/s0303-7207(02)00369-6. [DOI] [PubMed] [Google Scholar]
- 32.Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, et al. Generation and characterization of androgen receptor knockout (ARKO) mice:an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci U S A. 2002;99:13498–503. doi: 10.1073/pnas.212474399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Knudsen KE, Scher HI. Starving the addiction:new opportunities for durable suppression of AR signaling in prostate cancer. Clin Cancer Res. 2009;15:4792–8. doi: 10.1158/1078-0432.CCR-08-2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Knudsen KE, Penning TM. Partners in crime:deregulation of AR activity and androgen synthesis in prostate cancer. Trends Endocrinol Metab. 2010;21:315–24. doi: 10.1016/j.tem.2010.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pomerantz MM, Li F, Takeda DY, Lenci R, Chonkar A, et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat Genet. 2015;47:1346–51. doi: 10.1038/ng.3419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Krum SA, Miranda-Carboni GA, Lupien M, Eeckhoute J, Carroll JS, et al. Unique ERalpha cistromes control cell type-specific gene regulation. Mol Endocrinol. 2008;22:2393–406. doi: 10.1210/me.2008-0100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Copeland BT, Du J, Pal SK, Jones JO. Factors that influence the androgen receptor cistrome in benign and malignant prostate cells. Mol Oncol. 2019;13:2616–32. doi: 10.1002/1878-0261.12572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cirillo LA, Lin FR, Cuesta I, Friedman D, Jarnik M, et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol Cell. 2002;9:279–89. doi: 10.1016/s1097-2765(02)00459-8. [DOI] [PubMed] [Google Scholar]
- 39.Sahu B, Laakso M, Ovaska K, Mirtti T, Lundin J, et al. Dual role of FoxA1 in androgen receptor binding to chromatin, androgen signalling and prostate cancer. EMBO J. 2011;30:3962–76. doi: 10.1038/emboj.2011.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lee HJ, Hwang M, Chattopadhyay S, Choi HS, Lee K. Hepatocyte nuclear factor-3 alpha (HNF-3alpha) negatively regulates androgen receptor transactivation in prostate cancer cells. Biochem Biophys Res Commun. 2008;367:481–6. doi: 10.1016/j.bbrc.2007.12.162. [DOI] [PubMed] [Google Scholar]
- 41.Imamura Y, Sakamoto S, Endo T, Utsumi T, Fuse M, et al. FOXA1 promotes tumor progression in prostate cancer via the insulin-like growth factor binding protein 3 pathway. PLoS One. 2012;7:e42456. doi: 10.1371/journal.pone.0042456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jain RK, Mehta RJ, Nakshatri H, Idrees MT, Badve SS. High-level expression of forkhead-box protein A1 in metastatic prostate cancer. Histopathology. 2011;58:766–72. doi: 10.1111/j.1365-2559.2011.03796.x. [DOI] [PubMed] [Google Scholar]
- 43.Zhang C, Wang L, Wu D, Chen H, Chen Z, et al. Definition of a FoxA1 cistrome that is crucial for G1 to S-phase cell-cycle transit in castration-resistant prostate cancer. Cancer Res. 2011;71:6738–48. doi: 10.1158/0008-5472.CAN-11-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Decker KF, Zheng D, He Y, Bowman T, Edwards JR, et al. Persistent androgen receptor-mediated transcription in castration-resistant prostate cancer under androgen-deprived conditions. Nucleic Acids Res. 2012;40:10765–79. doi: 10.1093/nar/gks888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lupien M, Brown M. Cistromics of hormone-dependent cancer. Endocr Relat Cancer. 2009;16:381–9. doi: 10.1677/ERC-09-0038. [DOI] [PubMed] [Google Scholar]
- 46.Jin HJ, Zhao JC, Ogden I, Bergan RC, Yu J. Androgen receptor-independent function of FoxA1 in prostate cancer metastasis. Cancer Res. 2013;73:3725–36. doi: 10.1158/0008-5472.CAN-12-3468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang Q, Li W, Liu XS, Carroll JS, Jänne OA, et al. A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol Cell. 2007;27:380–92. doi: 10.1016/j.molcel.2007.05.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jia L, Berman BP, Jariwala U, Yan X, Cogan JP, et al. Genomic androgen receptor-occupied regions with different functions, defined by histone acetylation, coregulators and transcriptional capacity. PLoS One. 2008;3:e3645. doi: 10.1371/journal.pone.0003645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen Z, Song X, Li Q, Xie L, Guo T, et al. Androgen receptor-activated enhancers simultaneously regulate oncogene TMPRSS2 and lncRNA PRCAT38 in prostate cancer. Cells. 2019;8:864. doi: 10.3390/cells8080864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Krause WC, Shafi AA, Nakka M, Weigel NL. Androgen receptor and its splice variant, AR-V7, differentially regulate FOXA1 sensitive genes in LNCaP prostate cancer cells. Int J Biochem Cell Biol. 2014;54:49–59. doi: 10.1016/j.biocel.2014.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cai L, Tsai YH, Wang P, Wang J, Li D, et al. ZFX mediates non-canonical oncogenic functions of the androgen receptor splice variant 7 in castrate-resistant prostate cancer. Mol Cell. 2018;72:341–54.e6. doi: 10.1016/j.molcel.2018.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Paakinaho V, Swinstead EE, Presman DM, Grontved L, Hager GL. Meta-analysis of chromatin programming by steroid receptors. Cell Rep. 2019;28:3523–34.e2. doi: 10.1016/j.celrep.2019.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lupien M, Eeckhoute J, Meyer CA, Wang Q, Zhang Y, et al. FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription. Cell. 2008;132:958–70. doi: 10.1016/j.cell.2008.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet. 2007;39:311–8. doi: 10.1038/ng1966. [DOI] [PubMed] [Google Scholar]
- 55.Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, et al. Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 1999;13:966–77. doi: 10.1101/gad.13.8.966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Eeckhoute J, Keeton EK, Lupien M, Krum SA, Carroll JS, et al. Positive cross-regulatory loop ties GATA-3 to estrogen receptor alpha expression in breast cancer. Cancer Res. 2007;67:6477–83. doi: 10.1158/0008-5472.CAN-07-0746. [DOI] [PubMed] [Google Scholar]
- 57.Kouros-Mehr H, Slorach EM, Sternlicht MD, Werb Z. GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell. 2006;127:1041–55. doi: 10.1016/j.cell.2006.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Xiao L, Feng Q, Zhang Z, Wang F, Lydon JP, et al. The essential role of GATA transcription factors in adult murine prostate. Oncotarget. 2016;7:47891–903. doi: 10.18632/oncotarget.10294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang XD, Reeves K, Luo FR, Xu LA, Lee F, et al. Identification of candidate predictive and surrogate molecular markers for dasatinib in prostate cancer:rationale for patient selection and efficacy monitoring. Genome Biol. 2007;8:R255. doi: 10.1186/gb-2007-8-11-r255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kim J, Jin H, Zhao JC, Yang YA, Li Y, et al. FOXA1 inhibits prostate cancer neuroendocrine differentiation. Oncogene. 2017;36:4072–80. doi: 10.1038/onc.2017.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Berger A, Brady NJ, Bareja R, Robinson B, Conteduca V, et al. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest. 2019;129:3924–40. doi: 10.1172/JCI127961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang Y, Huang YX, Wang DL, Yang B, Yan HY, et al. LncRNA DSCAM-AS1 interacts with YBX1 to promote cancer progression by forming a positive feedback loop that activates FOXA1 transcription network. Theranostics. 2020;10:10823–37. doi: 10.7150/thno.47830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Su Y, Zhang Y, Zhao J, Zhou W, Wang W, et al. FOXA1 promotes prostate cancer angiogenesis by inducing multiple pro-angiogenic factors expression. J Cancer Res Clin Oncol. 2021;147:3225–43. doi: 10.1007/s00432-021-03730-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.He Y, Wang L, Wei T, Xiao YT, Sheng H, et al. FOXA1 overexpression suppresses interferon signaling and immune response in cancer. J Clin Invest. 2021;131:e147025. doi: 10.1172/JCI147025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sekiya T, Zaret KS. Repression by Groucho/TLE/Grg proteins:genomic site recruitment generates compacted chromatin in vitro and impairs activator binding in vivo. Mol Cell. 2007;28:291–303. doi: 10.1016/j.molcel.2007.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jangal M, Couture JP, Bianco S, Magnani L, Mohammed H, et al. The transcriptional co-repressor TLE3 suppresses basal signaling on a subset of estrogen receptor α target genes. Nucleic Acids Res. 2014;42:11339–48. doi: 10.1093/nar/gku791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Malik S, Jiang S, Garee JP, Verdin E, Lee AV, et al. Histone deacetylase 7 and FoxA1 in estrogen-mediated repression of RPRM. Mol Cell Biol. 2010;30:399–412. doi: 10.1128/MCB.00907-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Grabowska MM, Elliott AD, DeGraff DJ, Anderson PD, Anumanthan G, et al. NFI transcription factors interact with FOXA1 to regulate prostate-specific gene expression. Mol Endocrinol. 2014;28:949–64. doi: 10.1210/me.2013-1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wang L, Xu M, Kao CY, Tsai SY, Tsai MJ. Small molecule JQ1 promotes prostate cancer invasion via BET-independent inactivation of FOXA1. J Clin Invest. 2020;130:1782–92. doi: 10.1172/JCI126327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gao S, Chen S, Han D, Wang Z, Li M, et al. Chromatin binding of FOXA1 is promoted by LSD1-mediated demethylation in prostate cancer. Nat Genet. 2020;52:1011–7. doi: 10.1038/s41588-020-0681-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Park SH, Fong KW, Kim J, Wang F, Lu X, et al. Posttranslational regulation of FOXA1 by Polycomb and BUB3/USP7 deubiquitin complex in prostate cancer. Sci Adv. 2021;7:eabe2261. doi: 10.1126/sciadv.abe2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Takayama KI, Kosaka T, Suzuki T, Hongo H, Oya M, et al. Subtype-specific collaborative transcription factor networks are promoted by OCT4 in the progression of prostate cancer. Nat Commun. 2021;12:3766. doi: 10.1038/s41467-021-23974-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mansor R, Holly J, Barker R, Biernacka K, Zielinska H, et al. IGF-1 and hyperglycaemia-induced FOXA1 and IGFBP-2 affect epithelial to mesenchymal transition in prostate epithelial cells. Oncotarget. 2020;11:2543–59. doi: 10.18632/oncotarget.27650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–7. doi: 10.1038/nature03443. [DOI] [PubMed] [Google Scholar]
- 75.Gui B, Gui F, Takai T, Feng C, Bai X, et al. Selective targeting of PARP-2 inhibits androgen receptor signaling and prostate cancer growth through disruption of FOXA1 function. Proc Natl Acad Sci U S A. 2019;116:14573–82. doi: 10.1073/pnas.1908547116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Nitschke K, Erben P, Waldbillig F, Abdelhadi A, Weis CA, et al. Clinical relevance of gene expression in localized and metastatic prostate cancer exemplified by FABP5. World J Urol. 2020;38:637–45. doi: 10.1007/s00345-019-02651-8. [DOI] [PubMed] [Google Scholar]
- 77.Rotinen M, You S, Yang J, Coetzee SG, Reis-Sobreiro M, et al. ONECUT2 is a targetable master regulator of lethal prostate cancer that suppresses the androgen axis. Nat Med. 2018;24:1887–98. doi: 10.1038/s41591-018-0241-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Aboukameel A, Muqbil I, Baloglu E, Senapedis W, Landesman Y, et al. Down-regulation of AR splice variants through XPO1 suppression contributes to the inhibition of prostate cancer progression. Oncotarget. 2018;9:35327–42. doi: 10.18632/oncotarget.26239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rowley MJ, Corces VG. Organizational principles of 3D genome architecture. Nat Rev Genet. 2018;19:789–800. doi: 10.1038/s41576-018-0060-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhou S, Hawley JR, Soares F, Grillo G, Teng M, et al. Noncoding mutations target cis-regulatory elements of the FOXA1 plexus in prostate cancer. Nat Commun. 2020;11:441. doi: 10.1038/s41467-020-14318-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Fernandes RC, Toubia J, Townley S, Hanson AR, Dredge BK, et al. Post-transcriptional gene regulation by microRNA-194 promotes neuroendocrine transdifferentiation in prostate cancer. Cell Rep. 2021;34:108585. doi: 10.1016/j.celrep.2020.108585. [DOI] [PubMed] [Google Scholar]
- 82.Baca SC, Takeda DY, Seo JH, Hwang J, Ku SY, et al. Reprogramming of the FOXA1 cistrome in treatment-emergent neuroendocrine prostate cancer. Nat Commun. 2021;12:1979. doi: 10.1038/s41467-021-22139-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Daskivich TJ, Chamie K, Kwan L, Labo J, Palvolgyi R, et al. Overtreatment of men with low-risk prostate cancer and significant comorbidity. Cancer. 2011;117:2058–66. doi: 10.1002/cncr.25751. [DOI] [PubMed] [Google Scholar]
- 84.Li J, Yen C, Liaw D, Podsypanina K, Bose S, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275:1943–7. doi: 10.1126/science.275.5308.1943. [DOI] [PubMed] [Google Scholar]
- 85.Linja MJ, Visakorpi T. Alterations of androgen receptor in prostate cancer. J Steroid Biochem Mol Biol. 2004;92:255–64. doi: 10.1016/j.jsbmb.2004.10.012. [DOI] [PubMed] [Google Scholar]
- 86.Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 2012;44:685–9. doi: 10.1038/ng.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Parolia A, Cieslik M, Chu SC, Xiao L, Ouchi T, et al. Distinct structural classes of activating FOXA1 alterations in advanced prostate cancer. Nature. 2019;571:413–8. doi: 10.1038/s41586-019-1347-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Faisal FA, Murali S, Kaur H, Vidotto T, Guedes LB, et al. CDKN1B deletions are associated with metastasis in African American men with clinically localized, surgically treated prostate cancer. Clin Cancer Res. 2020;26:2595–602. doi: 10.1158/1078-0432.CCR-19-1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xu B, Song B, Lu X, Kim J, Hu M, et al. Altered chromatin recruitment by FOXA1 mutations promotes androgen independence and prostate cancer progression. Cell Res. 2019;29:773–5. doi: 10.1038/s41422-019-0204-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gao S, Chen S, Han D, Barrett D, Han W, et al. Forkhead domain mutations in FOXA1 drive prostate cancer progression. Cell Res. 2019;29:770–2. doi: 10.1038/s41422-019-0203-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Adams EJ, Karthaus WR, Hoover E, Liu D, Gruet A, et al. FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature. 2019;571:408–12. doi: 10.1038/s41586-019-1318-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Yang R, Van Etten JL, Dehm SM. Indel detection from DNA and RNA sequencing data with transIndel. BMC Genomics. 2018;19:270. doi: 10.1186/s12864-018-4671-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Annala M, Taavitsainen S, Vandekerkhove G, Bacon JV, Beja K, et al. Frequent mutation of the FOXA1 untranslated region in prostate cancer. Commun Biol. 2018;1:122. doi: 10.1038/s42003-018-0128-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
- 95.Beltran H, Hruszkewycz A, Scher HI, Hildesheim J, Isaacs J, et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin Cancer Res. 2019;25:6916–24. doi: 10.1158/1078-0432.CCR-19-1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.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. doi: 10.1016/j.eururo.2003.11.032. [DOI] [PubMed] [Google Scholar]
- 97.Beltran H, Tagawa ST, Park K, MacDonald T, Milowsky MI, et al. Challenges in recognizing treatment-related neuroendocrine prostate cancer. J Clin Oncol. 2012;30:e386–9. doi: 10.1200/JCO.2011.41.5166. [DOI] [PubMed] [Google Scholar]
- 98.Davis FG, McCarthy BJ, Freels S, Kupelian V, Bondy ML. The conditional probability of survival of patients with primary malignant brain tumors:surveillance, epidemiology, and end results (SEER) data. Cancer. 1999;85:485–91. [PubMed] [Google Scholar]
- 99.Garson JA, McIntyre PG, Kemshead JT. N-myc amplification in malignant astrocytoma. Lancet. 1985;2:718–9. doi: 10.1016/s0140-6736(85)92950-2. [DOI] [PubMed] [Google Scholar]
- 100.Lee WH, Murphree AL, Benedict WF. Expression and amplification of the N-myc gene in primary retinoblastoma. Nature. 1984;309:458–60. doi: 10.1038/309458a0. [DOI] [PubMed] [Google Scholar]
- 101.Marshall GM, Carter DR, Cheung BB, Liu T, Mateos MK, et al. The prenatal origins of cancer. Nat Rev Cancer. 2014;14:277–89. doi: 10.1038/nrc3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Northcott PA, Jones DT, Kool M, Robinson GW, Gilbertson RJ, et al. Medulloblastomics:the end of the beginning. Nat Rev Cancer. 2012;12:818–34. doi: 10.1038/nrc3410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Swartling FJ, Savov V, Persson AI, Chen J, Hackett CS, et al. Distinct neural stem cell populations give rise to disparate brain tumors in response to N-MYC. Cancer Cell. 2012;21:601–13. doi: 10.1016/j.ccr.2012.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60. doi: 10.1038/nature06008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chen Z, Wu D, Thomas-Ahner JM, Lu C, Zhao P, et al. Diverse AR-V7 cistromes in castration-resistant prostate cancer are governed by HoxB13. Proc Natl Acad Sci U S A. 2018;115:6810–5. doi: 10.1073/pnas.1718811115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Qi J, Nakayama K, Cardiff RD, Borowsky AD, Kaul K, et al. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell. 2010;18:23–38. doi: 10.1016/j.ccr.2010.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Liu Q, Pang J, Wang LA, Huang Z, Xu J, et al. Histone demethylase PHF8 drives neuroendocrine prostate cancer progression by epigenetically upregulating FOXA2. J Pathol. 2021;253:106–18. doi: 10.1002/path.5557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Nik-Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature. 2016;534:47–54. doi: 10.1038/nature17676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Melton C, Reuter JA, Spacek DV, Snyder M. Recurrent somatic mutations in regulatory regions of human cancer genomes. Nat Genet. 2015;47:710–6. doi: 10.1038/ng.3332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Robinson JL, Holmes KA, Carroll JS. FOXA1 mutations in hormone-dependent cancers. Front Oncol. 2013;3:20. doi: 10.3389/fonc.2013.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gerhardt J, Montani M, Wild P, Beer M, Huber F, et al. FOXA1 promotes tumor progression in prostate cancer and represents a novel hallmark of castration-resistant prostate cancer. Am J Pathol. 2012;180:848–61. doi: 10.1016/j.ajpath.2011.10.021. [DOI] [PubMed] [Google Scholar]
- 112.Robinson JL, Hickey TE, Warren AY, Vowler SL, Carroll T, et al. Elevated levels of FOXA1 facilitate androgen receptor chromatin binding resulting in a CRPC-like phenotype. Oncogene. 2014;33:5666–74. doi: 10.1038/onc.2013.508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Glinsky GV, Glinskii AB, Stephenson AJ, Hoffman RM, Gerald WL. Gene expression profiling predicts clinical outcome of prostate cancer. J Clin Invest. 2004;113:913–23. doi: 10.1172/JCI20032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. doi: 10.1016/j.ccr.2010.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Rezk M, Chandra A, Addis D, Møller H, Youssef M, et al. ETS-related gene (ERG) expression as a predictor of oncological outcomes in patients with high-grade prostate cancer treated with primary androgen deprivation therapy:a cohort study. BMJ Open. 2019;9:e025161. doi: 10.1136/bmjopen-2018-025161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Tsourlakis MC, Eleftheriadou A, Stender A, Weigand P, Grupp K, et al. FOXA1 expression is a strong independent predictor of early PSA recurrence in ERG negative prostate cancers treated by radical prostatectomy. Carcinogenesis. 2017;38:1180–7. doi: 10.1093/carcin/bgx105. [DOI] [PubMed] [Google Scholar]
- 117.Hernández-Llodrà S, Segalés L, Safont A, Juanpere N, Lorenzo M, et al. SPOP and FOXA1 mutations are associated with PSA recurrence in ERG wt tumors, and SPOP downregulation with ERG- rearranged prostate cancer. Prostate. 2019;79:1156–65. doi: 10.1002/pros.23830. [DOI] [PubMed] [Google Scholar]
- 118.Beltran H, Wyatt AW, Chedgy EC, Donoghue A, Annala M, et al. Impact of therapy on genomics and transcriptomics in high-risk prostate cancer treated with neoadjuvant docetaxel and androgen deprivation therapy. Clin Cancer Res. 2017;23:6802–11. doi: 10.1158/1078-0432.CCR-17-1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov. 2018;8:444–57. doi: 10.1158/2159-8290.CD-17-0937. [DOI] [PubMed] [Google Scholar]
- 120.Vandekerkhove G, Todenhöfer T, Annala M, Struss WJ, Wong A, et al. Circulating tumor DNA reveals clinically actionable somatic genome of metastatic bladder cancer. Clin Cancer Res. 2017;23:6487–97. doi: 10.1158/1078-0432.CCR-17-1140. [DOI] [PubMed] [Google Scholar]
- 121.Sunkel B, Wu D, Chen Z, Wang CM, Liu X, et al. Integrative analysis identifies targetable CREB1/FoxA1 transcriptional co-regulation as a predictor of prostate cancer recurrence. Nucleic Acids Res. 2017;45:6993. doi: 10.1093/nar/gkx282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Boysen G, Rodrigues DN, Rescigno P, Seed G, Dolling D, et al. SPOP-mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin Cancer Res. 2018;24:5585–93. doi: 10.1158/1078-0432.CCR-18-0937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ratta R, Guida A, Scotté F, Neuzillet Y, Teillet AB, et al. PARP inhibitors as a new therapeutic option in metastatic prostate cancer:a systematic review. Prostate Cancer Prostatic Dis. 2020;23:549–60. doi: 10.1038/s41391-020-0233-3. [DOI] [PubMed] [Google Scholar]
- 124.Chenoweth DM, Dervan PB. Allosteric modulation of DNA by small molecules. Proc Natl Acad Sci U S A. 2009;106:13175–9. doi: 10.1073/pnas.0906532106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chi KN, Gleave ME, Klasa R, Murray N, Bryce C, et al. A phase I dose-finding study of combined treatment with an antisense Bcl-2 oligonucleotide (Genasense) and mitoxantrone in patients with metastatic hormone-refractory prostate cancer. Clin Cancer Res. 2001;7:3920–7. [PubMed] [Google Scholar]
- 126.Tan PY, Chang CW, Chng KR, Wansa KD, Sung WK, et al. Integration of regulatory networks by NKX3-1 promotes androgen-dependent prostate cancer survival. Mol Cell Biol. 2012;32:399–414. doi: 10.1128/MCB.05958-11. [DOI] [PMC free article] [PubMed] [Google Scholar]

