Open Access

Oncogenic role of FOXM1 in human prostate cancer (Review)

  • Authors:
    • Da Young Lee
    • Jung Nyeo Chun
    • Insuk So
    • Ju-Hong Jeon
  • View Affiliations

  • Published online on: November 30, 2023     https://doi.org/10.3892/or.2023.8674
  • Article Number: 15
  • Copyright: © Lee et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer is the leading cause of cancer‑related mortality among men worldwide. In particular, castration‑resistant prostate cancer presents a formidable clinical challenge and emphasizes the need to develop novel therapeutic strategies. Forkhead box M1 (FOXM1) is a multifaceted transcription factor that is implicated in the acquisition of the multiple cancer hallmark capabilities in prostate cancer cells, including sustaining proliferative signaling, resisting cell death and the activation of invasion and metastasis. Elevated FOXM1 expression is frequently observed in prostate cancer, and in particular, FOXM1 overexpression is closely associated with poor clinical outcomes in patients with prostate cancer. In the present review, recent advances in the understanding of the oncogenic role of deregulated FOXM1 expression in prostate cancer were highlighted. In addition, the molecular mechanisms by which FOXM1 regulates prostate cancer development and progression were described, thereby providing knowledge and a conceptual framework for FOXM1. The present review also provided valuable insight into the inherent challenges associated with translating biomedical knowledge into effective therapeutic strategies for prostate cancer.

Introduction

Prostate cancer is a highly prevalent malignancy and the leading cause of cancer-related deaths among men worldwide (1,2). Localized or organ-confined prostate cancer can be effectively managed via surgical intervention or radiotherapy. Patients with de novo or recurrent metastatic prostate cancer initially exhibit favorable responses to androgen deprivation therapy, known as chemical castration. However, most patients eventually relapse and the disease progresses to an incurable or lethal castration-resistant state (35). The emergence of castration-resistant prostate cancer poses a substantial clinical challenge that highlights the need for the development of promising therapeutic strategies to overcome anti-cancer therapy resistance.

Forkhead box (FOX) proteins represent a superfamily of transcription factors that share an evolutionarily conserved DNA-binding domain called the ‘forkhead box’ or winged helix domain. FOX proteins are encoded by 50 genes in the human genome and may be categorized into 19 subfamilies (6) (Fig. 1). FOX proteins have crucial roles in regulating a wide spectrum of biological and developmental processes in response to environmental cues. The dysregulation of FOX alters cell fate and underlies various human diseases, particularly cancer (79). Furthermore, several FOX proteins, including FOXM1, have been shown to drive tumor initiation, progression, metastasis and drug resistance in various human cancers (9).

Among the FOX proteins, FOXM1 is ubiquitously expressed in various tissues during embryogenesis and its knockout leads to embryonic lethality owing to multiple developmental defects (10). FOXM1 expression is markedly decreased in adult tissues but is induced under various regenerative conditions (10). In addition, overexpression and mutation of FOXM1 are frequently observed in various human cancers, including prostate cancer (1115). It has also been confirmed that FOXM1 has a crucial role in tumorigenesis by regulating a multitude of biological processes, such as cell cycle, proliferation, migration, differentiation, apoptosis and metabolism (12,13,16). Therefore, FOXM1 has garnered attention as a promising target for the development of anti-cancer drugs. However, to the best of our knowledge, no comprehensive review that encompasses the oncogenic role and mechanisms of action of FOXM1 in prostate cancer has so far been compiled.

In the present review, recent advances in the understanding regarding the oncogenic role of FOXM1 and its underlying mechanisms in prostate cancer were presented. The challenges associated with FOXM1 were discussed and perspectives were provided for further related research and the application of the results obtained regarding the development of therapeutic strategies for prostate cancer.

Dysregulation of FOXM1 expression in prostate cancer

FOXM1 expression is frequently elevated in prostate cancer tissues (1727). It is regulated at multiple levels, including the transcription, post-transcription and protein stability (2143) (Fig. 2). FOXM1-activatory molecules are generally upregulated in prostate cancer tissues (21,24,25,3436), whereas FOXM1-inhibitory molecules are downregulated during prostate cancer progression (27,30,32,37). However, the molecular mechanisms associated with FOXM1 regulation in prostate cancer have remained to be fully elucidated, particularly regarding the nuclear localization and posttranslational modification of FOXM1. Therefore, further studies are required to elucidate the molecular mechanisms underlying dysregulated FOXM1 expression and accurately predict its transcriptional output in prostate cancer.

Overexpression of FOXM1 in prostate cancer tissues

Immunohistochemical, reverse transcription-PCR and transcriptomic analyses have consistently shown elevated FOXM1 expression levels in prostate cancer tissues compared to adjacent normal tissues (1727). A higher FOXM1 expression level has also been found to be significantly associated with tumor grade, disease severity or therapeutic resistance (1820,2224,28,29). In addition, FOXM1 overexpression is closely associated with poor prognosis in patients with prostate cancer (20,22,2528,3033) (Table I). Furthermore, several immunohistochemical studies have revealed that FOXM1 overexpression in the nucleus is strongly associated with tumor grade, disease severity and poor clinical outcomes (17,22,28). Therefore, targeting FOXM1 may be a clinically useful therapeutic approach for prostate cancer.

Table I.

Survival outcome of patients with prostate cancer based on forkhead box M1 expression level.

Table I.

Survival outcome of patients with prostate cancer based on forkhead box M1 expression level.

First author, yearData sourceHazard ratio; P-valueClinical outcome(Refs.)
Cheng, 2014GSE16560P=0.049Overall survival(30)
Ketola, 2017GSE21032 (Taylor dataset)P<0.001Disease-free survival(20)
Xu, 2022GEPIA1.9; P=0.0049Disease-free survival(27)
Tian, 2021RembrandtP=0.0107Overall survival(26)
Sharma, 2021TCGAP<0.01Recurrence-free survival(32)
Koo, 2023TCGA3.7; P=2.8×10-5Overall survival(33)
Kim, 2019Korea prostate bank10.524; P=0.022Biochemical recurrence-free survival(22)
Tian, 2021Private dataP=0.0125Overall survival(26)

[i] TCGA, The Cancer Genome Atlas.

FOXM1 overexpression via transcription

FOXM1 transcription is activated by various transcription factors, including chicken ovalbumin upstream promoter transcription factor II (COUP-TFII), cellular myelocytomatosis (c-Myc), hypoxia-inducible factor α (HIF1α) and E2 promoter binding factor 1 (E2F1) (21,24,34,35). These transcription factors induce FOXM1 expression by directly binding to the FOXM1 promoter. Specifically, E2F1 has been found to upregulate FOXM1 expression by recruiting su(var)3-9, enhancer-of-zeste and trithorax domain containing 1A (SETD1A), a histone H3K4 methyltransferase (25). Conversely, specificity protein 1, which is upregulated by G2 and S phase-expressed-1, also upregulates FOXM1 expression (36).

FOXM1 overexpression via derepression

Liver X receptor α (LXRα) knockdown was reported to upregulate FOXM1 expression (27). Indeed, a negative association has been observed between LXRα and FOXM1 in prostate tumor tissues (27). SAM-pointed domain-containing ETS transcription factor (SPDEF) also represses FOXM1 gene transcription by directly binding to the FOXM1 promoter (30). In addition, regucalcin and dachshund homolog 1 (DACH1), a winged helix/forkhead DNA-binding protein, suppresses FOXM1 expression (32,37). However, the expression levels of these repressive proteins are markedly reduced during prostate cancer progression and this leads to the derepression of FOXM1 expression.

FOXM1 overexpression via post-transcription

FOXM1 expression is increased by long noncoding RNAs, such as homeobox D cluster antisense RNA 1 (HOXD-AS1) and dipeptidyl peptidase-like 10-antisense RNA 1 (DPP10-AS1) (38,39). Furthermore, HOXD-AS1 mediates H3 lysine 4 (H3K4) trimethylation at the FOXM1 promoter by binding with tryptophan-aspartate repeat domain 5 (38). HOXD-AS1 also upregulates FOXM1 expression by sponging micro (mi)RNA miR-361-5p (40). By contrast, DPP10-AS1 induces cyclic AMP response element-binding protein-binding protein-mediated H3K27 acetylation at the FOXM1 promoter (39).

The post-transcriptional stability of FOXM1 mRNA is decreased by several miRNAs, including miR-31 and miR-193b (23,41). By contrast, miR-101 and miR-27a indirectly reduce FOXM1 expression by inhibiting COUP-TFII (34). MiR-877-5p also suppresses FOXM1 expression (42). These miRNAs are frequently downregulated in prostate cancer and this may partly explain the underlying mechanism of FOXM1 overexpression.

FOXM1 overexpression by protein stability

FOXM1 expression is regulated by altering O-linked β-N-acetylglucosamine transferase (OGT)-mediated protein stability (43). OGT upregulates FOXM1 expression by preventing its proteasomal degradation. However, as FOXM1 is not O-GlcNAcylated, OGT appears to indirectly regulate FOXM1 stability.

Other mechanisms

Transgenic adenocarcinoma of the mouse prostate (TRAMP) mice show upregulated FOXM1 expression, which is reversed via surgical castration (44). However, the synthetic androgen R1881 does not affect FOXM1 expression levels (45). This suggests that FOXM1 expression is upregulated in TRAMP mice through a mechanism that is independent of androgen receptor (AR) signaling. By contrast, p66Shc, an oxidative stress response protein, upregulates FOXM1 protein levels (46).

Summary of the regulation of FOXM1 expression

Several studies have contributed to the current knowledge regarding FOXM1 expression. However, the determinants of the expression levels of FOXM1 and its activity have remained to be fully elucidated. For instance, it remains unclear whether molecular signaling or nuclear transport pathways have crucial roles in the direct regulation of FOXM1 expression in prostate cancer. Therefore, efforts are required to further provide a comprehensive explanation and prediction of FOXM1-mediated biological outcomes.

Role of FOXM1 in prostate cancer

FOXM1 regulates various cancer hallmark-related biological processes, including cell cycle, survival, proliferation, apoptosis, autophagy, migration and invasion (Table II). In this section, the molecular mechanisms and biological roles of FOXM1 in prostate cancer were summarized.

Table II.

Summary of the biological roles of FOXM1 in human prostate cancer.

Table II.

Summary of the biological roles of FOXM1 in human prostate cancer.

First author, yearPhenomenonEffectPossible molecular mechanismCell modelAnimal model(Refs.)
Tian, 2021ApoptosisInhibitionIncrease in Bcl-2 and22Rv1, C4-2, siFOXM1-expressing(26)
RRM2 expressionDU145, DU145-DR,PC3-DR-
Xu, 2022 LNCaP, PC3, PC3-xenografted mouse(27)
Yu, 2020 DR, VCaP, VCaP- (47)
Yu, 2020 DR (48)
Lin, 2020 (49)
Mazzu, 2019 (50)
Wu, 2018 (54)
Lin, 2020AutophagyActivationIncrease in AMPKPC3, PC3-DR,/(49)
activity; Decrease inVCaP, VCaP-DR
mTOR activity
Kalin, 2006CellActivationIncrease in AR,22Rv1, C4-2, LNCaP-xenografted(17)
Pan, 2018proliferation EXO1,CDC6,DU145, DU145-DR,mouse; LNCaP-AI-
Mazzu, 2019and tumor 11β-HSD2,LNCaP, Myc-CaP,xenografted mouse;(21)
Tian, 2021growth KIF20A, RRM2,PC3, TRAMP C2, DU145-DR-xenografted
Xu, 2022 cyclin A2, cyclinVCaP, VCaP-DRmouse; FOXM1-(23)
Aytes, 2014 B1, cyclin B2, overexpressing PC3-(26)
Cheng, 2014 cyclin E1, cyclin D1, xenografted mouse;(27)
Lai, 2021 Cdc25b and CDK1 FOXM1-overexpressing(28)
Yu, 2020 expression LADY mouse; FOXM1-(30)
Yu, 2020 overexpressing; TRAMP
Mazzu, 2019 mouse; FOXM1-deleted(36)
Kim, 2021 TRAMP mouse;(47)
Wu, 2018 shFOXM1-expressing(48)
Li, 2011 DU145-xenografted(50)
Zhou, 2017 mouse(52)
Cai, 2013 (54)
Liu, 2014 (55)
(56)
(58)
(59)
Wang, 2014Invasion andActivationIncrease in EXO1,22Rv1, C4-2, 22Rv1-xenografted(19)
Pan, 2018metastasis KIF20A, RRM2,DU145, DU145-DR,mouse; FOXM1-(21)
Tang, 2019 vimentin, SLUG,LNCaP, PC3, PC3-overexpressing(24)
LOX, VCAN, ZEB2ML, TRAMP C2,TRAMP mouse; TRAMP
and VEGF expression;VCaP, VCaP-DRFOXM1-deleted mouse
Tian, 2021 Decrease in (26)
Cheng, 2014 E-cadherin expression (30)
Lin, 2016 (34)
Lynch, 2012 (43)
Yu, 2020 (48)
Mazzu, 2019 (50)
Kim, 2021 (52)
Li, 2011 (55)
Zhou, 2017 (56)
Cai, 2013 (58)
Qu, 2018 (60)
Yuan, 2018DrugActivationIncrease in UHRF1DU145-DR, LNCaP- siFOXM1-expressing(29)
Lin, 2016resistance and AR expressionER, PC3-DR, VCaP- PC3-DR-xenografted(34)
Gu, 2017 DRmouse(38)
Liu, 2017 (45)
Yu, 2020 (48)
Lin, 2020 (49)
Yuan, 2018CancerActivationIncrease in ALDH1,DU145, DU145-DR,/(29)
Koo, 2023stemness and NANOG, SOX2PC3 (30)
energy and SHH expression;
metabolism Increase in HK2, PKM2,
and LDHA expression

[i] DR, docetaxel-resistant; ER, enzalutamide resistance; AI, androgen-independent; FOX, forkhead box; RRM2, ribonucleotide reductase small subunit M2; EZH2, enhancer of zeste homolog 2; AMPK, adenosine monophosphate-activated protein kinase; mTOR, mammalian target of rapamycin; AR, androgen receptor; EXO1, exonuclease 1; CDC6, cell division cycle 6; 11β-HSD2, 11β-hydroxysteroid dehydrogenase 2; KIF20A, kinesin family member 20A; cdc25b, cell division cycle 25b; CDK1, cyclin dependent kinase 1; LOX, lysyl oxidase; VCAN, versican; ZEB2, zinc finger E-box binding homeobox 2; VEGF, vascular endothelial growth factor; ALDH1, aldehyde dehydrogenase 1; SOX2, sex-determining region of Y-related high mobility group-box; SHH, sonic hedgehog; HK2, hexokinase 2; PKM2, pyruvate kinase M2; LDHA, lactate dehydrogenase A.

Apoptosis and autophagy

In cell culture models, FOXM1 was observed to enable prostate cancer cells to acquire a cancer hallmark capability of resistance or evasion of apoptosis. Furthermore, FOMX1 overexpression suppresses apoptosis (4749) by inducing the ribonucleotide reductase small subunit M2 (RRM2) or enhancer of zeste homolog 2 (EZH2) (26,50). The expression levels of RRM2 and EZH2 are frequently elevated in prostate cancer and their upregulation is closely associated with poor clinical outcomes in patients with prostate cancer (50,51). By contrast, FOXM1 knockdown induces prostate cancer cell apoptosis (26,48). In addition, FOXM1 inhibition by miRNAs (e.g., miR-193b) or chemical compounds [e.g., forkhead domain inhibitory compound-6 (FDI-6), niclosamide, siomycin A, SR9009, morusin, cinnamaldehyde, cinnamic acid and eugenol] was reported to induce apoptosis (23,26,27,33,50,52,53).

It has also been observed that FOXM1 attenuates cell death by inducing protective autophagy (49). Furthermore, FOXM1 overexpression activates adenosine monophosphate-activated protein kinase (AMPK) and inhibits mammalian target of rapamycin (mTOR) activity, leading to autophagy. However, AMPK inhibitor compound C and mTOR activator MHY1485 were observed to abolish FOXM1-mediated autophagy and trigger apoptosis (49).

Cell proliferation and tumor growth

FOXM1 exerts oncogenic effects by sustaining proliferative signaling and evading growth suppressors in various experimental models, including cell cultures, xenografts and genetically engineered mouse models. Furthermore, FOXM1 knockdown suppresses cell cycle progression, cell viability, proliferation, colony formation or tumor growth (17,23,26,27,36,4850,52,54). In addition, FOXM1 inhibition by miRNAs (e.g., miR-877-5p) or chemical compounds [e.g., natura-α, tetramethylpyrazine (TMP), thiostrepton, monensin, FDI-6, thiostrepton, SR9009, morusin and baicalin] also reproduces knockdown phenotypes (20,27,33,36,42,47,50,55,56). By contrast, upregulation of FOXM1 by upstream molecules, such as c-Myc, DPP10-AS1, HOXD-AS1 and SETD1A, or dibutyl phthalate, was observed to promote cell proliferation, colony formation and tumor growth (21,25,38,39,57). Furthermore, FOXM1 upregulation following SPDEF inhibition also stimulated cell proliferation and tumor growth (30).

The possible mechanisms of cell proliferation and tumor growth mediated by FOXM1 include the induction of 11β-hydroxysteroid dehydrogenase 2, cell division cycle 6 (CDC6) and exonuclease 1 by FOXM1 (52,58,59), which also cooperates with other oncogenes, including AR and centromere protein F (CENPF) (28,31,34,59). The FOXM1-AR interaction has a crucial role in CDC6 upregulation (59), whereas the FOXM1-CENPF interaction activates various signaling pathways associated with prostate cancer malignancy, including the cell cycle and the PI3K and MAPK pathways (28). The combined inhibition of FOXM1 and AR or FOXM1 and CENPF using small interfering RNAs or chemical inhibitors significantly inhibit cell proliferation, colony formation and tumor growth (28,31,59).

Invasion and metastasis

FOXM1 has been shown to accelerate tumor malignancy by inducing angiogenesis and activating invasion and metastasis in cell cultures, xenografts and genetically engineered mouse models, while its knockdown inhibits cell migration and invasion (19,24,26,48,50). Furthermore, FOXM1 inhibition by miRNAs (miR-193b and miR-877-5p) or chemical compounds (FDI-6, TMP, thiostrepton, SR9009, natura-α and docetaxel plus anestat) also suppresses cell migration and invasion (23,27,42,46,55,56,60). In addition, FOXM1 inactivation via OGT depletion, regucalcin overexpression or FOXM1 gene ablation was reported to reduce angiogenesis, cell invasion and tumor metastasis (32,43,58). By contrast, ectopic FOXM1 expression or FOXM1 upregulation by COUP-TFII, c-Myc, HIF1α and exosomal HOXD-AS1 was observed to stimulate cell migration and invasion (21,24,34,40,48). Furthermore, FOXM1 upregulation following SPDEF inhibition also promoted cell migration and invasion (30).

The possible mechanisms of FOXM1-activated angiogenesis, invasion and metastasis include the upregulation of vascular endothelial growth factor, lysyl oxidase, versican and RRM2, as well as the stimulation of TGFβ-mediated epithelial-mesenchymal transition by FOXM1 (19,24,43,50,58). Of note, FOXM1 inhibition suppressed the expression of E-cadherin and upregulates the expression of vimentin, Slug and zinc finger E-box binding homeobox 2 (19,24).

Drug resistance

FOXM1 has been shown to confer resistance to chemical castration and nonselective chemotherapy in prostate cancer cells. FOXM1 knockdown enhances the efficacy of enzalutamide, an anti-androgen drug, as well as that of docetaxel (34,49). Furthermore, FOXM1 inhibition by miRNAs (e.g., miR-101 and miR-27a) or chemicals (e.g., thiostrepton) also increases sensitivity to docetaxel (34,48). HOXD-AS1 knockdown also represses resistance to bicalutamide and paclitaxel, possibly by suppressing FOXM1 expression (38). Furthermore, inhibition of FOXM1-induced autophagy via the knockdown of autophagy-related (ATG) protein 7 or beclin-1 or using chloroquine, compound C or MHY1485 restored sensitivity to docetaxel in FOXM1-overexpressing cells (49). Conversely, FOXM1 overexpression leads to enzalutamide and docetaxel resistance (34,48,49).

The upregulation of the plant homeodomain and an interesting new gene, finger domain-containing 1 is involved in FOXM1-mediated therapeutic resistance (29). In addition, FOXM1-mediated AR upregulation provides a possible explanation for resistance to chemical castration in prostate cancer (45).

Other biological processes

FOXM1 regulates cancer stemness and metabolic programs in cell culture and xenograft models. Specifically, inhibition of FOXM1 by thiostrepton or monensin suppresses cancer stemness (20), while its overexpression increases the expression levels of cancer stem cell-associated molecules, such as aldehyde dehydrogenase 1 (ALDH1), NANOG homeobox, sex-determining region of Y-related high mobility group-box (SOX) and sonic hedgehog (SHH) (29). Furthermore, FOXM1 inhibition using morusin suppresses glycolysis by reducing the expression of hexokinase 2 (HK2), pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA) (33), implicating FOXM1 in deregulating cellular energetics, a cancer hallmark.

Summary of the role of FOXM1 in prostate cancer

FOXM1 is a crucial determinant of tumor cell physiology in established prostate cancer cells, as evidenced by loss-of-function experiments, which showed reduced cell survival, proliferation, migration and invasion. However, the role of FOXM1 in driving prostate cancer remains inconclusive, as FOXM1 transgenic mice do not develop prostate tumors or hyperplasia (17,58). Furthermore, FOXM1 overexpression significantly accelerates tumor development and growth in TRAMP or LADY prostate cancer mouse models (17). These findings suggest that the precise role of FOXM1 and its underlying molecular mechanisms in prostate cancer development and progression are yet to be fully elucidated.

Therapeutic agents targeting FOXM1 in prostate cancer

Numerous synthetic and naturally occurring compounds have been shown to inhibit FOXM1 expression in prostate cancer cells. These compounds include siomycin A (61), thiostrepton (48,61), natura-α (55), metformin (19), TMP (39,56), monensin (20), FDI-6 (46), mocetinostat (23), baicalin (47), niclosamide (52), dilazep (62), MYCi975 (35), SR9009 (27) and morusin (33). In addition, combination therapies with rapamycin and PD0325901 (31) or docetaxel and aneustat (60) have also been shown to inhibit FOXM1 activity or expression.

These compounds exert anti-cancer effects by modulating various biological processes, including cell proliferation, migration, invasion or apoptosis (Table III). Among them, siomycin A has been found to potentiate the anti-cancer effects of bicalutamide (45,59), whereas thiostrepton has been shown to increase sensitivity to docetaxel (48). However, only a small number of compounds have been validated via FOXM1 rescue experiments, which demonstrated their ability to reverse phenotypic alterations. For instance, natura-α inhibits cell proliferation and invasion (55); tetramethylpyrazine suppresses cell proliferation, colony formation, migration and invasion (56), and combined treatment with docetaxel and aneustat reduces cell migration (60). It has also been noted that these alterations can be reversed via forced FOXM1 expression.

Table III.

Anti-tumor activity of the compounds inhibiting FOXM1 expression.

Table III.

Anti-tumor activity of the compounds inhibiting FOXM1 expression.

Effective concentration or dose for inhibiting FOXM1 expression

In vitro experimentIn vivo experiment


First author, yearCompound nameModelConcentrationModelDose, mg/kgAnti-tumor activity(Refs.)
Pandit, 2010Siomycin ADU145,5 µM//Apoptosis↑(61)
LNCaP, PC3 Proliferation↓
Pandit, 2010ThiostreptonDU145,5 µM//Apoptosis↑(61)
LNCaP, PC3 Proliferation↓
Yu, 2020 DU145-DR,3 µMDU145-DR-30Proliferation↓(48)
VCaP-DR xenografted mouse
Li, 2011Natura-αLNCaP,4-10 mmol/lLNCaP and100Proliferation↓(55)
LNCaP-AI LNCaP-AI- Invasion↓
xenografted mouse
Bu, 2023 C4-25.067 µM//Apoptosis↑(57)
PC34.523 µM//Proliferation↓
Wang, 2014MetforminDU14520 mM//Proliferation↓(19)
Zhou, 2017 TetramethylpyrazinePC3100-1,000 PC3-xenografted10-100Proliferation↓(56)
g/lmouse Invasion↓
Migration↓
Zhou, 2020 //DPP10-AS1-50Proliferation↓(39)
overexpressing
PC3-xenografted
mouse
Ketola, 2017Monensin42D-ER10-100 nM42D-ER-10Proliferation↓(20)
xenografted mouse
Ingersoll, 2018FDI-6LNCaP5 µM//Proliferation↓(46)
Migration↓
Mazzu, 2019MocetinostatLNCaP, 22Rv11 µM//Proliferation↓(23)
Yu, 2020BaicalinLNCaP, PC-310 µg/mlLNCaP-10-40Proliferation↓(47)
xenografted mouse
Kim, 2021Niclosamide22Rv1, PC30.25–10 µM22Rv1-20-50Apoptosis↑(52)
xenografted mouse Proliferation↓
Kaochar, 2021DilazepLNCaP,50 µMMDA PCa337A-50Proliferation↓(62)
LNCaP-Abl, xenografted mouse
LNCaP-ER
Holmes, 2022MYCi97522Rv110 µM22Rv1-100Proliferation↓(35)
xenografted mouse
Xu, 2022SR900922Rv1, PC320 µM22Rv1-100Apoptosis↑(27)
xenografted mouse Proliferation↓
Migration↓
Koo, 2023MorusinDU145, PC35-10 µM//Apoptosis↑(33)
Proliferation↓
Mitrofanova,Rapamycin +22Rv1,3 µM +//Proliferation↓(31)
2015PD0325901DU145, PC31 µM
Qu, 2018Aneustat +C4-2100 µg/ml +//Invasion↓(60)
Docetaxel 5 nM Migration↓

[i] DR, docetaxel-resistant; AI, androgen-independent; ER, enzalutamide-resistant; FDI-6, forkhead domain inhibitory compound-6; Abl, ablation of androgen; FOX, foxhead box.

Numerous compounds that suppress FOXM1 expression exhibit anti-tumor activity against prostate cancer, suggesting that therapeutic strategies targeting FOXM1 may be useful in the treatment of prostate cancer. However, further studies are necessary to establish whether these anti-tumor effects are solely attributable to FOXM1 inhibition. Moreover, the specific mechanisms by which these compounds inhibit FOXM1 expression need to be elucidated.

Summary and future direction

Altered transcriptional programs improve the biological fitness of prostate cancer cells under various stress conditions, providing survival benefits to these cells in a given microenvironment. FOXM1, a representative transcription factor, enhances prostate cancer cell survival by regulating transcription. Increased FOXM1 expression, which is frequently observed in prostate cancer cells, is associated with disease severity and a poor prognosis in patients. FOXM1 also mediates cancer hallmarks, including sustaining proliferative signaling, resisting cell death and activating invasion and metastasis. Furthermore, FOXM1 enhances sensitivity to anti-androgen therapy or nonselective chemotherapy. Therefore, these results suggest that FOXM1 holds promising potential as a therapeutic target in prostate cancer. In addition, FOXM1 has been indicated to have clinical utility as both a prognostic and predictive marker in prostate cancer.

However, several challenges still exist with respect to understanding the role of FOXM1 in prostate cancer. First, current research has mainly focused on identifying the molecular mechanisms that regulate FOXM1 expression. Therefore, the molecular signaling mechanisms that control FOXM1 activity in prostate cancer, such as post-translational modifications and subcellular localization, require further elucidation. Second, FOXM1 has four different splicing variants (11), and it remains unclear whether these different variants have specific oncogenic functions. Third, FOX proteins may act as monomers or dimers with other interacting partners (8). Therefore, additional studies are necessary to determine which transcription and chromatic remodeling factors cooperate with FOXM1 during prostate cancer progression. Furthermore, it is necessary to investigate changes in FOXM1-mediated transcription programs and outputs that drive tumor development and progression. Fourth, it is important to clarify the molecular mechanisms by which FOXM1 contributes to therapeutic resistance and androgen independence in prostate cancer. Finally, the tumor microenvironment (TME) has an important role in cancer evolution, with hypoxia leading to the selection of more malignant prostate cancer cells. However, the role of FOXM1 in the TME remains poorly understood. Therefore, further research should focus on answering these questions to improve our understanding of the role of FOXM1 in prostate cancer biology and treatment.

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Research Foundation of Korea funded by the Korean government (MSIT; grant nos. 2018R1A4A1023822 and 2020R1A2C1102574) and the Education and Research Encouragement Fund of Seoul National University Hospital.

Availability of data and materials

Not applicable.

Authors' contributions

DYL, JNC and JHJ conceived and designed the article. DYL, JNC and JHJ reviewed the literature and wrote the manuscript. DYL, JNC and IS surveyed the literature and provided suggestions. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Berenguer CV, Pereira F, Camara JS and Pereira JAM: Underlying features of prostate cancer-statistics, risk factors, and emerging methods for its diagnosis. Curr Oncol. 30:2300–2321. 2023. View Article : Google Scholar : PubMed/NCBI

3 

Huang Y, Jiang X, Liang X and Jiang G: Molecular and cellular mechanisms of castration resistant prostate cancer. Oncol Lett. 15:6063–6076. 2018.PubMed/NCBI

4 

Yanagisawa T, Kawada T, Rajwa P, Kimura T and Shariat SF: Emerging systemic treatment for metastatic castration-resistant prostate cancer: A review of recent randomized controlled trials. Curr Opin Urol. 33:219–229. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Cai M, Song XL, Li XA, Chen M, Guo J, Yang DH, Chen Z and Zhao SC: Current therapy and drug resistance in metastatic castration-resistant prostate cancer. Drug Resist Updat. 68:1009622023. View Article : Google Scholar : PubMed/NCBI

6 

Jackson BC, Carpenter C, Nebert DW and Vasiliou V: Update of human and mouse forkhead box (FOX) gene families. Hum Genomics. 4:345–352. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Myatt SS and Lam EW: The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer. 7:847–859. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Herman L, Todeschini AL and Veitia RA: Forkhead transcription factors in health and disease. Trends Genet. 37:460–475. 2021. View Article : Google Scholar : PubMed/NCBI

9 

Castaneda M, Hollander PD and Mani SA: Forkhead box transcription factors: Double-Edged swords in cancer. Cancer Res. 82:2057–2065. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Kalin TV, Ustiyan V and Kalinichenko VV: Multiple faces of FoxM1 transcription factor: Lessons from transgenic mouse models. Cell Cycle. 10:396–405. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Gartel AL: FOXM1 in Cancer: Interactions and vulnerabilities. Cancer Res. 77:3135–3139. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Liao GB, Li XZ, Zeng S, Liu C, Yang SM, Yang L, Hu CJ and Bai JY: Regulation of the master regulator FOXM1 in cancer. Cell Commun Signal. 16:572018. View Article : Google Scholar : PubMed/NCBI

13 

Kalathil D, John S and Nair AS: FOXM1 and cancer: Faulty cellular signaling derails homeostasis. Front Oncol. 10:6268362021. View Article : Google Scholar : PubMed/NCBI

14 

Liu C, Barger CJ and Karpf AR: FOXM1: A multifunctional oncoprotein and emerging therapeutic target in ovarian cancer. Cancers (Basel). 13:30652021. View Article : Google Scholar : PubMed/NCBI

15 

Katzenellenbogen BS, Guillen VS and Katzenellenbogen JA: Targeting the oncogenic transcription factor FOXM1 to improve outcomes in all subtypes of breast cancer. Breast Cancer Res. 25:762023. View Article : Google Scholar : PubMed/NCBI

16 

Khan MA, Khan P, Ahmad A, Fatima M and Nasser MW: FOXM1: A small fox that makes more tracks for cancer progression and metastasis. Semin Cancer Biol. 92:1–15. 2023. View Article : Google Scholar : PubMed/NCBI

17 

Kalin TV, Wang IC, Ackerson TJ, Major ML, Detrisac CJ, Kalinichenko VV, Lyubimov A and Costa RH: Increased levels of the FoxM1 transcription factor accelerate development and progression of prostate carcinomas in both TRAMP and LADY transgenic mice. Cancer Res. 66:1712–1720. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Chandran UR, Ma C, Dhir R, Bisceglia M, Lyons-Weiler M, Liang W, Michalopoulos G, Becich M and Monzon FA: Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer. 7:642007. View Article : Google Scholar : PubMed/NCBI

19 

Wang Y, Yao B, Wang Y, Zhang M, Fu S, Gao H, Peng R, Zhang L and Tang J: Increased FoxM1 expression is a target for metformin in the suppression of EMT in prostate cancer. Int J Mol Med. 33:1514–1522. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Ketola K, Munuganti RSN, Davies A, Nip KM, Bishop JL and Zoubeidi A: Targeting prostate cancer subtype 1 by forkhead box M1 pathway inhibition. Clin Cancer Res. 23:6923–6933. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Pan H, Zhu Y, Wei W, Shao S and Rui X: Transcription factor FoxM1 is the downstream target of c-Myc and contributes to the development of prostate cancer. World J Surg Oncol. 16:592018. View Article : Google Scholar : PubMed/NCBI

22 

Kim MY, Jung AR, Kim GE, Yang J, Ha US, Hong SH, Choi YJ, Moon MH, Kim SW, Lee JY and Park YH: High FOXM1 expression is a prognostic marker for poor clinical outcomes in prostate cancer. J Cancer. 10:749–756. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Mazzu YZ, Yoshikawa Y, Nandakumar S, Chakraborty G, Armenia J, Jehane LE, Lee GM and Kantoff PW: Methylation-associated miR-193b silencing activates master drivers of aggressive prostate cancer. Mol Oncol. 13:1944–1958. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Tang C, Liu T, Wang K, Wang X, Xu S, He D and Zeng J: Transcriptional regulation of FoxM1 by HIF-1α mediates hypoxia-induced EMT in prostate cancer. Oncol Rep. 42:1307–1318. 2019.PubMed/NCBI

25 

Yang L, Jin M, Park SJ, Seo SY and Jeong KW: SETD1A promotes proliferation of castration-resistant prostate cancer cells via FOXM1 transcription. Cancers (Basel). 12:17362020. View Article : Google Scholar : PubMed/NCBI

26 

Tian JH, Mu LJ, Wang MY, Zeng J, Long QZ, Bin-Guan Wang W, Jiang YM, Bai XJ and Du YF: FOXM1-Dependent transcriptional regulation of EZH2 induces proliferation and progression in prostate cancer. Anticancer Agents Med Chem. 21:1835–1841. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Xu H, Zhang J, Zheng X, Tan P, Xiong X, Yi X, Yang Y, Wang Y, Liao D, Li H, et al: SR9009 inhibits lethal prostate cancer subtype 1 by regulating the LXRα/FOXM1 pathway independently of REV-ERBs. Cell Death Dis. 13:9492022. View Article : Google Scholar : PubMed/NCBI

28 

Aytes A, Mitrofanova A, Lefebvre C, Alvarez MJ, Castillo-Martin M, Zheng T, Eastham JA, Gopalan A, Pienta KJ, Shen MM, et al: Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell. 25:638–651. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Yuan B, Liu Y, Yu X, Yin L, Peng Y, Gao Y, Zhu Q, Cao T, Yang Y, Fan X and Li X: FOXM1 contributes to taxane resistance by regulating UHRF1-controlled cancer cell stemness. Cell Death Dis. 9:5622018. View Article : Google Scholar : PubMed/NCBI

30 

Cheng XH, Black M, Ustiyan V, Le T, Fulford L, Sridharan A, Medvedovic M, Kalinichenko VV, Whitsett JA and Kalin TV: SPDEF inhibits prostate carcinogenesis by disrupting a positive feedback loop in regulation of the Foxm1 oncogene. PLoS Genet. 10:e10046562014. View Article : Google Scholar : PubMed/NCBI

31 

Mitrofanova A, Aytes A, Zou M, Shen MM, Abate-Shen C and Califano A: Predicting drug response in human prostate cancer from preclinical analysis of in vivo mouse models. Cell Rep. 12:2060–2071. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Sharma S, Pei X, Xing F, Wu SY, Wu K, Tyagi A, Zhao D, Deshpande R, Ruiz MG, Singh R, et al: Regucalcin promotes dormancy of prostate cancer. Oncogene. 40:1012–1026. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Koo JI, Sim DY, Lee HJ, Ahn CH, Park J, Park SY, Lee D, Shim BS, Kim B and Kim SH: Apoptotic and anti-Warburg effect of Morusin via ROS mediated inhibition of FOXM1/c-Myc signaling in prostate cancer cells. Phytother Res. 37:4473–4487. 2023. View Article : Google Scholar : PubMed/NCBI

34 

Lin SC, Kao CY, Lee HJ, Creighton CJ, Ittmann MM, Tsai SJ, Tsai SY and Tsai MJ: Dysregulation of miRNAs-COUP-TFII-FOXM1-CENPF axis contributes to the metastasis of prostate cancer. Nat Commun. 7:114182016. View Article : Google Scholar : PubMed/NCBI

35 

Holmes AG, Parker JB, Sagar V, Truica MI, Soni PN, Han H, Schiltz GE, Abdulkadir SA and Chakravarti D: A MYC inhibitor selectively alters the MYC and MAX cistromes and modulates the epigenomic landscape to regulate target gene expression. Sci Adv. 8:eabh36352022. View Article : Google Scholar : PubMed/NCBI

36 

Lai W, Zhu W, Li X, Han Y, Wang Y, Leng Q, Li M and Wen X: GTSE1 promotes prostate cancer cell proliferation via the SP1/FOXM1 signaling pathway. Lab Invest. 101:554–563. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Li Z, Jiao X, Robertson AG, Di Sante G, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, et al: The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. Oncogene. 42:1857–1873. 2023. View Article : Google Scholar : PubMed/NCBI

38 

Gu P, Chen X, Xie R, Han J, Xie W, Wang B, Dong W, Chen C, Yang M, Jiang J, et al: lncRNA HOXD-AS1 regulates proliferation and chemo-resistance of castration-resistant prostate cancer via recruiting WDR5. Mol Ther. 25:1959–1973. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Zhou Y, Zhou Z, Ji Z, Yan W, Li H and Yu X: Tetramethylpyrazine reduces prostate cancer malignancy through inactivation of the DPP10-AS1/CBP/FOXM1 signaling pathway. Int J Oncol. 57:314–324. 2020.PubMed/NCBI

40 

Jiang Y, Zhao H, Chen Y, Li K, Li T, Chen J, Zhang B, Guo C, Qing L, Shen J, et al: Exosomal long noncoding RNA HOXD-AS1 promotes prostate cancer metastasis via miR-361-5p/FOXM1 axis. Cell Death Dis. 12:11292021. View Article : Google Scholar : PubMed/NCBI

41 

Lin PC, Chiu YL, Banerjee S, Park K, Mosquera JM, Giannopoulou E, Alves P, Tewari AK, Gerstein MB, Beltran H, et al: Epigenetic repression of miR-31 disrupts androgen receptor homeostasis and contributes to prostate cancer progression. Cancer Res. 73:1232–1244. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Yang B, Diao H, Wang P, Guan F and Liu H: microRNA-877-5p exerts tumor-suppressive functions in prostate cancer through repressing transcription of forkhead box M1. Bioengineered. 12:9094–9102. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Lynch TP, Ferrer CM, Jackson SR, Shahriari KS, Vosseller K and Reginato MJ: Critical role of O-Linked β-N-acetylglucosamine transferase in prostate cancer invasion, angiogenesis, and metastasis. J Biol Chem. 287:11070–11081. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Wan L, Tan HL, Thomas-Ahner JM, Pearl DK, Erdman JW Jr, Moran NE and Clinton SK: Dietary tomato and lycopene impact androgen signaling- and carcinogenesis-related gene expression during early TRAMP prostate carcinogenesis. Cancer Prev Res (Phila). 7:1228–1239. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Liu Y, Liu Y, Yuan B, Yin L, Peng Y, Yu X, Zhou W, Gong Z, Liu J, He L and Li X: FOXM1 promotes the progression of prostate cancer by regulating PSA gene transcription. Oncotarget. 8:17027–17037. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Ingersoll MA, Chou YW, Lin JS, Yuan TC, Miller DR, Xie Y, Tu Y, Oberley-Deegan RE, Batra SK and Lin MF: p66Shc regulates migration of castration-resistant prostate cancer cells. Cell Signal. 46:1–14. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Yu Z, Zhan C, Du H and Zhang L, Liang C and Zhang L: Baicalin suppresses the cell cycle progression and proliferation of prostate cancer cells through the CDK6/FOXM1 axis. Mol Cell Biochem. 469:169–178. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Yu H, Xu Z, Guo M, Wang W, Zhang W, Liang S, Xu Z, Ye J, Zhu G, Zhang C and Lin J: FOXM1 modulates docetaxel resistance in prostate cancer by regulating KIF20A. Cancer Cell Int. 20:5452020. View Article : Google Scholar : PubMed/NCBI

49 

Lin JZ, Wang WW, Hu TT, Zhu GY, Li LN, Zhang CY, Xu Z, Yu HB, Wu HF and Zhu JG: FOXM1 contributes to docetaxel resistance in castration-resistant prostate cancer by inducing AMPK/mTOR-mediated autophagy. Cancer Lett. 469:481–489. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Mazzu YZ, Armenia J, Chakraborty G, Yoshikawa Y, Coggins SA, Nandakumar S, Gerke TA, Pomerantz MM, Qiu X, Zhao H, et al: A novel mechanism driving poor-prognosis prostate cancer: Overexpression of the DNA repair gene, ribonucleotide reductase small subunit M2 (RRM2). Clin Cancer Res. 25:4480–4492. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG, Otte AP, et al: The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 419:624–629. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Kim MY, Jung AR, Shin D, Kwon H, Cho HJ, Ha US, Hong SH, Lee JY, Kim SW and Park YH: Niclosamide exerts anticancer effects through inhibition of the FOXM1-mediated DNA damage response in prostate cancer. Am J Cancer Res. 11:2944–2959. 2021.PubMed/NCBI

53 

Gopalakrishnan S and Ismail A: Aromatic monophenols from cinnamon bark act as proteasome inhibitors by upregulating ER stress, suppressing FoxM1 expression, and inducing apoptosis in prostate cancer cells. Phytother Res. 35:5781–5794. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Wu M, Zhao H, Guo L, Wang Y, Song J, Zhao X, Li C, Hao L, Wang D and Tang J: Ultrasound-mediated nanobubble destruction (UMND) facilitates the delivery of A10-3.2 aptamer targeted and siRNA-loaded cationic nanobubbles for therapy of prostate cancer. Drug Deliv. 25:226–240. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Li Y, Ligr M, McCarron JP, Daniels G, Zhang D, Zhao X, Ye F, Wang J, Liu X, Osman I, et al: Natura-alpha targets forkhead box m1 and inhibits androgen-dependent and -independent prostate cancer growth and invasion. Clin Cancer Res. 17:4414–4424. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Zhou Y, Ji Z, Yan W, Zhou Z, Li H and Xiao Y: Tetramethylpyrazine inhibits prostate cancer progression by downregulation of forkhead box M1. Oncol Rep. 38:837–842. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Bu H, Tang S, Liu G, Miao C, Zhou X, Yang H and Liu B: In silico, in vitro and in vivo studies: Dibutyl phthalate promotes prostate cancer cell proliferation by activating Forkhead Box M1 and remission after Natura-alpha pretreatment. Toxicology. 488:1534652023. View Article : Google Scholar : PubMed/NCBI

58 

Cai Y, Balli D, Ustiyan V, Fulford L, Hiller A, Misetic V, Zhang Y, Paluch AM, Waltz SE, Kasper S and Kalin TV: Foxm1 expression in prostate epithelial cells is essential for prostate carcinogenesis. J Biol Chem. 288:22527–22541. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Liu Y, Gong Z, Sun L and Li X: FOXM1 and androgen receptor co-regulate CDC6 gene transcription and DNA replication in prostate cancer cells. Biochim Biophys Acta. 1839:297–305. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Qu S, Ci X, Xue H, Dong X, Hao J, Lin D, Clermont PL, Wu R, Collins CC, Gout PW and Wang Y: Treatment with docetaxel in combination with Aneustat leads to potent inhibition of metastasis in a patient-derived xenograft model of advanced prostate cancer. Br J Cancer. 118:802–812. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Pandit B and Gartel AL: New potential anti-cancer agents synergize with bortezomib and ABT-737 against prostate cancer. Prostate. 70:825–833. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Kaochar S, Rusin A, Foley C, Rajapakshe K, Robertson M, Skapura D, Mason C, Berman De Ruiz K, Tyryshkin AM, Deng J, et al: Inhibition of GATA2 in prostate cancer by a clinically available small molecule. Endocr Relat Cancer. 29:15–31. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2024
Volume 51 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee DY, Chun JN, So I and Jeon J: Oncogenic role of FOXM1 in human prostate cancer (Review). Oncol Rep 51: 15, 2024
APA
Lee, D.Y., Chun, J.N., So, I., & Jeon, J. (2024). Oncogenic role of FOXM1 in human prostate cancer (Review). Oncology Reports, 51, 15. https://doi.org/10.3892/or.2023.8674
MLA
Lee, D. Y., Chun, J. N., So, I., Jeon, J."Oncogenic role of FOXM1 in human prostate cancer (Review)". Oncology Reports 51.1 (2024): 15.
Chicago
Lee, D. Y., Chun, J. N., So, I., Jeon, J."Oncogenic role of FOXM1 in human prostate cancer (Review)". Oncology Reports 51, no. 1 (2024): 15. https://doi.org/10.3892/or.2023.8674