Open Access

Widespread activation and critical role of EMT and stemness in the neuroendocrine differentiation of prostate cancer (Review)

  • Authors:
    • Yun-Fan Li
    • Shuai Su
    • Yu Luo
    • Chengcheng Wei
    • Jingke He
    • Liang-Dong Song
    • Kun Han
    • Jue Wang
    • Xiangzhi Gan
    • De-Lin Wang
  • View Affiliations

  • Published online on: July 4, 2025     https://doi.org/10.3892/or.2025.8942
  • Article Number: 109
  • Copyright: © Li 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

Neuroendocrine (NE) prostate cancer (NEPC) is an aggressive and lethal subtype of prostate cancer. It is typically characterized by the expression of NE markers and the loss of androgen receptor expression. De novo NEPC is rare, accounting for <2% of all prostate cancer cases at diagnosis. More commonly, NEPC arises from prostate adenocarcinoma following androgen deprivation therapy, with 20‑25% of metastatic castration‑resistant prostate cancers undergoing NE differentiation due to lineage plasticity. During this transition, pathways associated with epithelial‑mesenchymal transition (EMT) and stemness are broadly activated, which is considered to be a key driver of NEPC's high metastatic potential, resistance to chemotherapy and radiotherapy and poor prognosis. EMT facilitates metastasis by enhancing cellular motility and invasiveness, while stemness properties contribute to post‑metastatic colonization, immune evasion, therapy resistance and cellular dormancy. As manifestations of cellular plasticity, these processes share overlapping molecular mechanisms. Targeting key regulators within these pathways may offer promising therapeutic strategies for NEPC.

Introduction

Prostate cancer (PCa) remains the most common non-skin cancer among American men, accounting for 11% of cancer-related mortalities and ranking as the second leading cause of cancer mortality in men (1). Androgen deprivation therapy (ADT) is the primary treatment for PCa, but although patients may initially respond, the disease often recurs in a lethal metastatic form known as castration-resistant prostate cancer (CRPC) (2). Adenocarcinoma is the predominant histological variant of CRPC and continues to rely on AR signaling, which can be reactivated through acquired AR gene mutations, amplifications, or other mechanisms (3,4). In a significant subset of patients undergoing ADT, 20–25% of CRPC tumors eventually lose their dependence on AR signaling over the course of the disease, transitioning from AR-expressing PCa tissue to AR-negative, poorly differentiated small cell neuroendocrine (NE) carcinoma tissue (5). This form of PCa is commonly referred to as NEPCa, encompassing pure small cell carcinoma as well as mixed adenocarcinoma-NE tumor morphologies (6). However, newly developed NEPC is rare, observed in <2% of diagnosed PCa cases (7).

The process of NE differentiation (NED) involves extensive molecular reprogramming and is accompanied by a range of biological events. Among these, epithelial-mesenchymal transition (EMT) and stemness have attracted particular attention due to their significant roles. Stemness refers to the ability to differentiate into multiple lineages. It is evident that all these processes involve transitions in histological types, thereby introducing the concept of cellular plasticity. Cellular plasticity refers to the dynamic process by which cells transition from one state to another. The concept of ‘plasticity’ underscores the extensive reprogramming in stem cells that drives differentiation, highlighting the pivotal role of stemness in transforming cellular identity (8). The present study aimed not only to explore NED in prostate cancer but also to elucidate the origins, molecular mechanisms and clinical significance of EMT and stemness. It sought to view these three aspects as an interconnected whole within the context of prostate cancer. Although ADT therapy is the primary driver of NED transformation, the roles of EMT and stemness in this process are also significant and cannot be overlooked (Fig. 1). Prostate cancer serves as a critical platform that integrates EMT, stemness and NED. By investigating the important roles of EMT and stemness in prostate cancer, we can deepen our understanding of NED. The present study explored the extensive interconnections among EMT, stemness and NED to clarify the mechanisms of NEPC development, its clinical characteristics, molecular pathways and potential therapeutic strategies.

NEPC

Origin of NEPC

The origin of NEPC remains a subject of controversy. Some studies suggest that normal NE cells in the prostate may be selectively preserved and undergo malignant proliferation during ADT (9,10). However, the lineage plasticity hypothesis, which is supported by more substantial evidence, posits that prostate adenocarcinoma cells originating from luminal or basal cells may lose their adenocarcinoma characteristics and acquire a NE phenotype during prolonged ADT (9). This hypothesis suggests a similarity between the origins of NE cells and stem cells. Derivatives of therapy-resistant NE tumors consistently exhibit molecular characteristics of the primary adenocarcinoma, including specific somatic mutations (9). This supports the hypothesis that aggressive NE tumors evolve through lineage plasticity rather than originating as a second primary cancer. An open question remains as to whether lineage conversion involves a dedifferentiation phase, in which adenocarcinoma cells initially lose AR-specific gene expression and acquire stem-like properties before differentiating into NE cells, or whether transdifferentiation occurs directly, bypassing this intermediate stem-like stage (11). Based on a comprehensive analysis, the present study proposed that the development of NEPC, as illustrated in Fig. 1, may involve multiple potential origins.

The incidence of NEPC appears to be increasing, potentially linked to the use of more effective AR-targeting therapies. A rapid autopsy study found that 13.3% of patients had metastases with an AR-negative NE phenotype and an additional 23.3% had AR-negative metastases without NE features (6). These rates exceed those reported in autopsies conducted prior to the approval of potent AR signaling inhibitors such as enzalutamide and abiraterone acetate (5.4% AR-negative, 6.3% NE) (6). This suggests that with the increasing use of ADT, the progression of normal prostate adenocarcinomas to NEPC is likely to become more common.

Diagnosis of NEPC

NEPC can be somewhat imprecise. While a number of prostate adenocarcinomas contain occasional benign NE cells within the epithelium, only 5–10% of tumors exhibit distinct, large, multifocal clusters of NE cells, which are classified as adenocarcinomas NED (12). These tumors typically present as small, infiltrating glands with prominent nucleoli, hyperchromasia, absence of intraluminal secretions or pale blue mucin and isolated tumor cells, often associated with eosinophilic granules (13,14). Immunohistochemical staining for at least one NE marker, such as synaptophysin, chromogranin A, neuron-specific enolase (NSE), or CD56, is essential for confirming the diagnosis of NE differentiation (14).

Clinical characteristics of NEPC

The association between NED and poorer oncologic outcomes in prostate cancer remains controversial. While some studies suggest a link, NED in typical adenocarcinoma has shown only a weak correlation with prognosis, insufficient to be considered clinically significant (11,13). Most research indicates that NED does not affect outcomes, with each study examining NED in prostate cancer through needle biopsies and transurethral resections of the prostate (1518). Different pathological subtypes yield varying outcomes: NEPC tumors with well-differentiated Paneth cell-like and carcinoid differentiation tend to have an improved prognosis, while poorly differentiated NE tumors, particularly small cell carcinoma, exhibit greater aggressiveness (11).

One study compared 36 cases of pure small cell carcinoma with 51 cases (58.6%) of mixed small cell carcinoma and adenocarcinoma. The findings revealed that pure small cell carcinoma had significantly shorter overall survival compared with mixed histology (8.9 months vs. 26.1 months following NEPC diagnosis; P<0.001) (19). The transition from adenocarcinoma to NEPC is not abrupt but involves an intermediate state. In its final stage, prostate cancer with NE differentiation presents the following four clinical features (6,14,19): i) Highly aggressive with poor prognosis: A systematic review found that the median survival following an NEPC diagnosis is only 7 months (20). ii) Predominantly osteolytic/visceral metastasis: Tumors tend to be large with low PSA levels, with a median PSA level of less than 4 ng/ml. iii) Non-responsive to androgen deprivation therapy. iv) Short-term response to platinum-based chemotherapy. The factors driving well-prognosed adenocarcinoma and adenocarcinoma with NED to transform into the poorly prognosed NEPC during this lineage switch remain an open question.

The role of NED in prostate cancer is closely associated with the development of castration resistance, yet the high malignancy of NEPC is more critically attributed to the molecular events that occur during NED. The extensive activation of EMT and stemness during NED probably contributes to the aggressive nature of NEPC, which is characterized by high metastatic potential, resistance to radiotherapy and chemotherapy and poor prognosis. The activation of EMT and stemness may be an indispensable component of NEPC.

The procedure and role of EMT

EMT is a crucial process in embryonic development; however, it also plays a pivotal role in resistance to endocrine therapy and cancer progression. EMT involves the transformation of epithelial cells into a mesenchymal phenotype, during which malignant epithelial cells progressively lose adhesion molecules such as E-cadherin, EpCAM, syndecans and tight junction molecules. At the same time, gene expression factors such as SNAIL1, SNAI2/Slug and TWIST increase, along with mesenchymal markers such as vimentin, N-cadherin and metalloproteinases (21). Mesenchymal cells, compared with epithelial cells, exhibit enhanced migratory capacity, invasiveness, apoptosis resistance and the ability to integrate components of the Extracellular Matrix (ECM) (22). Tumors often harbor hybrid cell populations in a hybrid epithelial/mesenchymal (E/M) state, co-expressing both epithelial and mesenchymal traits, which enhances the metastatic potential of cancer cells, allowing them to migrate from primary to distant sites (23,24). During this reprogramming, cancer cells release enzymes that break their attachment to the basement membrane, accompanied by phenotypic alterations such as actin cytoskeleton reorganization, which promote migration and metastasis (25). While EMT facilitates the initial spread of tumor cells, the clinical emergence of metastases requires mesenchymal-to-epithelial transition (MET), which is essential for the successful seeding and colonization of disseminated cells at secondary sites (26). In NEPC, the activation of EMT may be a critical factor driving the high metastatic potential of tumors. Therefore, targeting EMT-related factors could play a pivotal role in treating NEPC. Molecules such as MCTP1 and Snail, which promote both EMT and NED, represent potential therapeutic targets (27,28).

Stemness

Prevalence of stem cells and tissue colonization

The most lethal malignancies, including lung, gastric, liver, breast and prostate cancers, as well as hematological cancers such as leukemia, contain cells with varying stem cell-like properties (29). As early as 1877, Cohnheim, a student of Virchow, observed this population of cells with embryonic characteristics (30). Today, these cells are referred to as cancer stem cells (CSCs) or tumor-initiating cells (TICs) and are recognized as the driving force behind tumor establishment and progression (31). Although CSCs comprise <1% of the primary tumor (32,33), their effect is profound. Stemness is characterized by the capacity for self-renewal, multilineage differentiation and extended cellular lifespan functions vital for normal physiology and tissue repair (34). As with normal stem cells, cancer stem cells have the ability to divide, grow, undergo clonal expansion and differentiate (35). This ability closely mirrors the uncontrolled proliferation observed in cancer cells, making CSCs genetically akin to cancer cells and more susceptible to mutations that drive oncogenesis. However, unlike normal stem cell niches, which possess tumor-suppressive functions and various signaling pathways to regulate growth, CSC niches provide a supportive microenvironment for cancer cells. This niche includes cancer-associated macrophages and cancer-associated fibroblasts, which promote cancer cell growth and differentiation through the secretion of various molecules, including growth factors, cytokines and vesicles such as exosomes (36,37). CSCs can adapt to stress conditions such as oxidative damage, hypoxia and exposure to exogenous substances by altering their gene expression programs, leading to phenotypes associated with migration, invasion and drug resistance (38,39).

Current evidence suggests that the primary reservoirs of normal stem cells in the prostate are luminal and basal cells (24). Under normal conditions, stem cells typically exhibit a basal cell phenotype (40). However, following ADT, stem-like cells with a luminal phenotype are more likely to serve as the origin of tumor re-initiation in CRPC (41). The cellular origin of NEPC is also luminal epithelial cells (9) and the stemness of these luminal epithelial cells may facilitate NED.

Acquisition of stemness and resistance to therapy

Tumors acquire stemness under the pressure of endocrine therapy, chemotherapy and radiotherapy through a series of related molecular mechanisms. During chemotherapy or radiotherapy, certain tumor cells may enter a state of senescence known as therapy-induced senescence (TIS), with the senescence-associated secretory phenotype (SASP) promoting the emergence of stem cell-like subpopulations. Over time, the antitumor effects of TIS may wane, allowing the cancer to regain stemness, which can contribute to tumor recurrence (39). Research has shown that the tumorigenic potential of cancer cells is linked to the increased expression of CD44 or other CSC-like characteristics, driven by SASP-related cytokines such as IL-8 and IL-6 (42). The acquisition of cancer stemness is driven not only by the evasion of senescence but also by the plasticity of non-CSCs and CSCs. This plasticity is influenced by endocrine therapy, where the loss of AR signaling can induce EMT and NED, processes that further promote stemness. The effects of SASP and plasticity are difficult to distinguish, with key interleukins involved in SASP being IL-1, IL-6 and IL-8, which can influence the plasticity phenotype of CSCs (43). Additionally, the development of CSCs remains a topic of ongoing debate, with various models proposed. Genetic and epigenetic alterations, along with changes in the ‘stem cell niche’, can result in unchecked stem cell proliferation, endowing these stem-like cells with oncogenic and drug-resistant potential (39). CSCs may arise from aberrant differentiation of stem cells, mutations and dedifferentiation of normal cells, or dedifferentiation of tumor cells.

CSCs exhibit resistance to androgen deprivation, pro-oxidants, radiation and chemotherapy (4446). In other studies, CSCs have demonstrated significant resistance to various treatments, including tyrosine kinase inhibitors, taxanes and topoisomerase inhibitors (47,48). The resistance of CSCs to chemotherapeutic agents arises from a combination of intrinsic and extrinsic factors (49). Intrinsic factors contributing to CSC therapy resistance include efficient slow cell cycle progression, DNA repair mechanisms, tumor cell vasculogenic mimicry, active anti-apoptotic pathways, poor immunogenicity and the maintenance of stemness characteristics. Extrinsic factors involve the high cell density of CSCs, lineage plasticity, the tumor microenvironment (TME) and epigenetic influences (31,5052). These features are also evident in NEPC. Although NEPC initially shows sensitivity to platinum-based chemotherapy, resistance and disease progression often develop rapidly and the response to other types of drugs is generally poor. Additionally, similar to stem cells, NEPC exhibits alterations in DNA repair mechanisms (53), tumor angiogenesis (54,55), anti-apoptotic pathways (56) and autophagy (57,58).

Stemness and tumor metastasis

The relationship between CSCs and tumor metastasis is complex, with evidence suggesting that the role of stemness in metastasis is not to promote the initiation of metastasis, but rather to facilitate post-migration colonization (38). Current research indicates that EMT promotes the emergence of stemness (59), while stemness may, in turn, facilitate MET, aiding in cellular colonization; a coherent process. Mesenchymal cells lack the ability to colonize; however, CSCs, which constitute a small subset of malignant cells within most tumors, are ultimately responsible for relapse and metastasis (38). Epithelial cell adhesion molecule (EpCAM) is one of the earliest identified CSC markers. The expression of EpCAM is associated with a CSC-like phenotype that promotes bone metastasis in mice and is often associated with the expression of other CSC markers, such as CD44 and CD166 (60). Additionally, as an epithelial cell marker, EpCAM is downregulated during EMT and upregulated during MET, with larger metastatic tumors (≥30 cells) and primary tumors showing high levels of EpCAM expression (61). Therefore, the expression of EpCAM in CSCs reflects their potential for adhesion and post-migration colonization, which is associated with MET. In a mouse model of basal-like breast cancer, continuous MET activation induces stem cell properties (62). In pancreatic and prostate cancers, both MET and hepatocyte growth factor are predominantly expressed in stem-like tumor cells (38). Studies have shown that hematopoietic stem cells undergo a phenotypic shift toward a more mesenchymal state when multiple stemness genes such as Sox2, Myc and Klf4 are knocked out (38,46). By using magnetic-activated cell sorting, CSC populations have been isolated from mesenchymal cancer cells (MCCs). Compared with MCCs, CSCs demonstrate slower proliferation, greater resistance to apoptosis, drug resistance and heightened clonogenic potential, although their invasiveness is reduced (38).

CSCs and disseminated tumor cells (DTCs) share several biological characteristics, such as dormancy (63). Research indicates that DTCs expressing a CSC phenotype constitute only a small fraction of the total DTC population (63). However, these CSC-like DTCs are more effective at displacing hematopoietic stem cells (HSCs) from their interaction with osteoblasts a mechanism HSCs use to localize to the bone niche (64). By displacing HSCs, CSCs bind to components of the HSC niche, enriching the dormant population of prostate cancer cells and enhancing their ability to resist various chemotherapeutic attacks (65,66). Dormant-enriched human PCa cell populations bind to osteoblasts within the bone marrow HSC niche, inducing the expression of TANK-binding kinase 1 (67). This, in turn, inhibits mTOR signaling, promoting dormancy, maintaining the CSC phenotype (CD44+/CD133+) and contributing to drug resistance (61). Increasing evidence suggests that CSCs, though a small population within most tumors, are largely responsible for relapse and metastasis (38,68). Therefore, the activation of stemness in NEPC may promote the survival and colonization of tumor cells after metastasis, leading to the development of visceral and osteolytic metastases (14).

Interactions between ADT, EMT, stemness and NED

ADT promotes EMT, stemness and NED in prostate cancer. It is important to determine whether these three processes occur simultaneously or follow a specific sequential order. After reviewing a large body of literature, the present study proposed that the relationship among these processes can be simplified as shown in Fig. 2. The findings suggested that EMT occurs earlier than stemness, which, in turn, precedes NED. The evolution from androgen receptor-positive prostate cancer (ARPC; AR+, NE-) to NEPC (NE+, AR-) often involves intermediate states, including AR-, NE- and AR+, NE+ subtypes. Studies indicate that mesenchymal and stem-like prostate cancer (MSPC) overlaps with the AR-, NE-subtype by 75–80%, suggesting that MSPC is an intermediate state in the progression from ARPC to NEPC (69). EMT may occur prior to the acquisition of stemness, as the EMT process can induce a stem-like state due to the partial overlap between EMT and stemness transcriptional programs (59). The role of EMT in conferring stemness mirrors its function in normal embryonic development, where EMT primarily controls the differentiation of stem cells into various lineages (70). Furthermore, molecules associated with EMT and stemness collectively promote the progression to NED.

ADT promotes EMT, stemness and NED

Induced by ADT and inversely correlated with AR signaling, features such as EMT, CSCs and NED are commonly observed (24,71,72). These features emerge under the stress of ADT treatment, with tumor cells undergoing transdifferentiation to evade drug-related pressures (73). For example, LuCaP 35 ×enografts derived from human PCa show concurrent EMT and increased expression of stem cell markers such as WNT 5a and WNT 5b upon ADT treatment (73). Androgen deprivation can also induce the upregulation of EMT-associated molecules, including vimentin (VIM), Zinc finger E-box binding homeobox 1 (ZEB1), N-cadherin (CDH2), Snail Family Zinc Finger 2 (SLUG) and Twist Family BHLH Transcription Factor 1 (TWIST1) (73). Notably, a bidirectional negative feedback loop exists between AR and ZEB1. After ADT treatment, stem-like cells reach their peak levels (74). PSA-/low cell populations exhibit gene expression profiles similar to those of stem cells, characterized by enhanced self-renewal capacity and resistance to ADT and chemotherapeutic agents (75). CSCs isolated from AR-negative DU145 cells show high expression of stem cell markers, including CD24, integrin α2β1, CD44 and the reprogramming factor SRY-box 2 (SOX2). These cells display tumor-initiating potential and self-renewal capabilities (76). These changes may represent adaptive mechanisms under the pressure of ADT treatment, driving the tumor cells toward similar pathways.

ADT suppresses AR signaling, leading to a transformation of epithelial cells into a NE phenotype, which is widely accepted as a key process in prostate cancer progression. The focus here is on the downstream molecular mechanisms through which ADT-induced AR suppression promotes NED. ADT enhances NED by increasing the expression of Forkhead transcription factor (Fox)a2 and knockout of Foxa2 reverses the conversion of the glandular to the NE lineage (77). Notably, ten days after enzalutamide treatment, significant increases in chromatin accessibility were observed compared with 3 days and untreated CRPC, with enrichment in stem cell and neuronal pathways. Knockout experiments further confirmed that ASCL1 is essential for establishing neuronal and stem cell-like lineages (78). ASCL1 and FOXA2 are both considered lineage-determining factors in NED, as they remodel chromatin into a permissive state, facilitating subsequent transcription factor binding and indirectly highlighting the critical role of ADT in NED (79). ADT alleviates AR-mediated suppression of H19, a long non-coding (lnc)RNA involved in cellular plasticity, driving either the NE phenotype (H19 overexpression) or the luminal phenotype (H19 knockdown) (80). Furthermore, ADT reduces EHF transcription by disrupting AR binding to androgen response elements, thus promoting the expression and enzymatic activity of Enhancer of Zeste Homolog 2 (EZH2), which catalyzes histone H3 trimethylation at lysine 27 to repress downstream genes and facilitate NED (81). Enzalutamide promotes lncRNA-p21 transcription through AR inhibition, altering EZH2 function from a histone methyltransferase to a non-histone methyltransferase, leading to STAT3 methylation and promoting NED (82). ADT also activates cAMP response element-binding protein (CREB) and the CREB-EZH2-TSP1 pathway is a newly identified mechanism driving therapy-induced NED in prostate cancer (55). Overexpression of DPYSL5 in prostate cancer cells induces a neuron-like phenotype, enhances invasion and proliferation, upregulates stemness and NE-related markers and negatively correlates with AR and PSA (83). Mechanistically, DPYSL5 promotes prostate cancer cell plasticity via EZH2-mediated polycomb repressive complex 2 (PRC2) activation (83). Depletion of DPYSL5 reduces proliferation, induces G1 phase cell cycle arrest, reverses the NE phenotype and upregulates luminal genes. Notably, EZH2 emerges as a key downstream molecule in these ADT-induced NED pathways. Enzalutamide also induces NED via the MAOA/mTOR/HIF-1α signaling axis (84). Additionally, AR inhibition depletes REST, leading to NED (85). BRN2 expression, directly suppressed by AR and negatively correlated with AR activity, drives the NE phenotype by binding to SOX2 upon release from suppression (86). AR binds directly to the cis-enhancer region of SOX2 and AR inhibition enhances SOX2 expression (87). Following AR pathway inhibition (ARPI), the expression of PEG10 is derepressed, regulated by both AR and the E2F-RB1 transcription factor pathway during the early and late stages of NEPC development (88). Targeting these downstream pathways associated with AR inhibition may provide opportunities to reverse EMT, stemness and NED in prostate cancer.

EMT promotes stemness

Various EMT-related molecules, including ZEB1, PDGF-D, Snail and E-cadherin, play pivotal roles in promoting stemness in cancer cells (24). The relationship between stemness and EMT is complex. For example, silencing E-cadherin in spheroids derived from the PC3 human prostate cancer cell line stimulates the EMT process (89), whereas its expression induces stemness gene expression and spheroid formation in DU145 prostate cancer cells (90,91). Elevated levels of the EMT regulator ZEB1 promote stem cell-like traits in prostate cancer cells, leading to increased SOX2 expression (92). Additionally, CD44-low cells (non-CSCs) can transition to a CD44-high phenotype (CSCs), forming mammospheres in response to TGF-β-driven ZEB1 expression (93). Overexpression of platelet-derived growth factor D in PC3 cells results in morphological changes characteristic of EMT, enhanced clonogenicity and increased spheroid formation, along with higher expression of stemness-related transcription factors, including Sox2, Oct4, Nanog, Lin28B and components of the PRC2 (94). Loss of E-cadherin is crucial in conferring oncogenic properties, particularly in enhancing both stemness and metastatic potential (95). Activation of TGF-β signaling or transcription factors such as Snail can convert non-cancer stem cells (CD133- or ALDH-low) into cancer stem cells/progenitor cells (CD133+ or ALDH-high) (96).

EMT promotes NED

Recent evidence underscores the crucial role of EMT as a driving force in NED. Key EMT inducers involved in NED, such as Twist, Snail, Slug and E-cadherin repressors such as Zeb1/2, are essential to this process (97). For instance, overexpression of Snail leads to the downregulation of E-cadherin and the upregulation of NE differentiation markers, including ENO2 and CHGA (98). The interplay between the TGF-β signaling network and the AR axis markedly influences EMT-related phenotypic changes. Disruption of epithelial homeostasis via EMT facilitates the transformation of epithelial-derived tumors into invasive forms, enabling them to acquire a NE phenotype and rapidly metastasize (99).

Stemness-related factors promote NED

Through extensive literature review, the present study identified that stemness-related factors play a crucial role in the process of NED. Particularly, molecules such as SOX2, MYC, EZH2 and ASCL1.

SOX2, a member of the SRY-related HMG box family, is essential for maintaining embryonic stem cell pluripotency and regulating neuronal differentiation (100102). Compared with patients with PCa or CRPC-adenocarcinoma, NEPC patients exhibit significant upregulation of SOX2 expression (103). In PCa cells, NED genes are upregulated by SOX2 overexpression and downregulated by its silencing (103). These findings suggest that the loss of PTEN, TP53 and RB1 drives the NE phenotype through SOX2 (104). The molecular mechanism by which SOX2 promotes NED likely involves transactivation of Serine Peptidase Inhibitor Kazal Type 1 (SPINK1) and upregulation of H19 and ASCL1 (105). Additionally, SOX2 regulates global epigenetics through LSD1, leading to reduced ARPC-specific gene expression in NEPC, representing a novel mechanism for SOX2 in NEPC progression (106). Several signals upregulate SOX2 in NEPC. AR chromatin immunoprecipitation (ChIP) studies in CRPC-adenocarcinoma cells (CWR-R1) show that AR directly binds to the enhancer region of SOX2 and ligand activation of AR inhibits SOX2 expression, which is relieved by ADT, eliminating AR ligand-mediated suppression of SOX2 expression (107). RB1 can interact with E2F in fibroblasts and be recruited to the promoter region of SOX2, marking it for repression (108). The core pluripotent stem cell gene LIN28 mediates the derepression of the transcription factor HMGA2, which in turn promotes SOX2 expression by inhibiting let-7 miRNA expression (109). Overexpression of Mucin 1-C-terminal subunit (MUC1-C) induces SOX2 expression through the MUC1-C-MYC-BRN2 axis (110). TBX2 binds to the SOX2 promoter and inhibits the expression of miR-200c-3p, leading to increased expression of its targets, SOX2 and N-MYC (111). Furthermore, SNAI2, TRIM59, TRIB2 and SRRM4 all promote NED by upregulating SOX2 expression (9295). Overall, SOX2 serves as a critical downstream mechanism in multiple molecular pathways leading to NED. Additionally, the pluripotency factor Oct4 is closely associated with AR-independent NE-like tumors (112).

The MYC protein family plays a significant role in cell reprogramming. As a major transcriptional regulator, MYC promotes a plasticity-permissive, stem-like state. Inhibition of CDC7 suppresses NE transdifferentiation by inducing the degradation of MYC protein, enhancing the response to targeted therapy in in vivo models of NE transformation. This process is linked to both stemness and histological transformation (113). Additionally, CDC7 inhibition markedly prolongs the response to standard cytotoxic agents, such as cisplatin and irinotecan, in small cell prostate cancer models (113). The N-Myc oncogene, encoded by the MYCN gene, is a neurodevelopmental-related transcription factor. N-Myc expression is higher in NEPC compared with non-NE CRPC and it plays an important role in NED (114). In the absence of androgens, upregulation of N-Myc is associated with an enrichment of stem cell characteristics and neural lineage differentiation markers. In castrated 22Rv1 ×enograft mice and GEM models, the N-Myc signature was enriched with genes related to neural lineage pathways, such as neural progenitor cell bivalent genes and genes involved in neural development (such as SOX11, SOX21, NTRK1 and NKX2-1), adult stem cells (such as HOXA2/A3/A9/A10 and WNT5A), ESCs (such as SOX2) and NEPC (such as CHGA), while epithelial lineage-associated genes were downregulated (115). Studies on mouse models with PTEN deficiency and MYCN overexpression have shown that N-Myc interacts with EZH2, AR and various AR co-factors (such as FOXA1 and HOXB13) both in vitro and in vivo (114116). N-Myc directly binds to the promoters of NSE, SYP and AR, suggesting its critical role in regulating the emergence of NEPC (116). Furthermore, N-Myc interacts with AR by directly binding to AR enhancers, driving NE differentiation through EZH2-mediated transcriptional programs (114). Aurora A stabilizes N-Myc (117) and the Aurora A inhibitor alisertib can disrupt this stabilization, preventing NED (118). Another member of the MYC family, c-MYC, collaborates with Pim1 kinase to drive NE differentiation in aggressive prostate tumors in prostate xenografts (119).

ASCL1, an early lineage determinant of NED, is enriched in regions associated with DNA-binding motifs involved in stem cell and neuronal lineage programming (78,120). Genome-wide mapping of ASCL1 occupancy in NEPC cell lines through chromatin immunoprecipitation sequencing revealed that ASCL1 directly regulates key CSC genes, including SOX2, NANOG and OCT4, as well as NE genes such as CHGA, ENO2 (NSE), NCAM1 and DLL1, all known ASCL1 targets (78). Further supporting this, knockdown of ASCL1 resulted in downregulation of these genes, while deletion of ASCL1 triggered a lineage switch from neuronal to luminal (78,120). Additionally, the ASCL1 cistrome is enriched with targets of PRC2 and deletion of ASCL1 reduces EZH2 activity in NEPC cell lines. Conversely, overexpression of ASCL1 enhances EZH2 activity (78).

EZH2, an epigenetic regulator and catalytic subunit of PRC2, catalyzes the trimethylation of histone H3 at lysine 27 (H3K27me3), promoting transcriptional silencing. Its catalytic activity contributes to lineage-specific cell fate determination by either repressing lineage-specific factors or activating transcription factors that drive stem cell and NE programs (121). Compared with adenocarcinoma and a number of progressing NEPC patients, EZH2 is overexpressed in NEPC samples (121,122). It interacts with miR708 to drive the NE phenotype in PCa (123). In an N-Myc-driven NEPC mouse model, EZH2 catalytic activity facilitates the formation of an N-Myc/AR/EZH2-PRC2 complex, downregulating AR signaling and promoting NEPC (114). Furthermore, AR and EZH2 co-occupy the reprogrammed AR cistrome to transcriptionally modulate stem cell and neuronal gene networks (124). EZH2 also plays a crucial role in promoting NE differentiation during ADT, as outlined in previous sections.

The stem cell marker CD44 is selectively associated with NE tumors, with CD44+ PSA-cells capable of initiating tumors, developing castration resistance and expressing NE markers (75). Recently, the FOXC2 has been identified as a unique marker of the stem cell phenotype in NEPC, where AR+ prostate cancer cells acquire FOXC2 expression, correlating with NE transdifferentiation and resistance to enzalutamide and docetaxel (125). Loss of FOXC2 function is sufficient to trigger a phenotypic switch from NE to luminal epithelial characteristics. O-linked N-acetylglucosamine transferase (OGT), associated with stemness and EMT, is crucial for KIF1A-induced NE phenotype and invasive growth (126). MYBL2, a pluripotency gene, serves as a chromatin-binding partner for key pluripotency factors such as OCT4, NANOG and SOX2. Its expression and activity are enriched in human NEPC and NE-like mouse models (127). Cyclin-dependent kinase 2, a transcriptional target of MYBL2, exhibits potent anti-tumor responses when inhibited in RB1-deficient NEPC (128). Furthermore, L1CAM knockdown in PC3 cells impairs their ability to form tumor spheres and reduces the expression of both cancer stemness and NE markers (129).

Shared molecular mechanisms

Numerous classical molecular pathways, genes, transcription factors and epigenetic abnormalities can simultaneously activate EMT, stemness and NED (Table I). These phenomena exhibit broad associations, demonstrating the consistency and synergy among the three processes. A number of studies have shown that blocking these key molecules can reverse NED differentiation and inhibit tumor progression in vivo, suggesting that targeting these molecular pathways may offer meaningful therapeutic opportunities for advanced NEPC patients and potentially lead to improved treatment outcomes (114,130133).

Table I.

Shared pathway and molecular of EMT, stemness and NED.

Table I.

Shared pathway and molecular of EMT, stemness and NED.

A, Signaling pathways

NameRoleMechanisms(Refs.)
JAK/STATEMT1. JAK-STAT3 phosphodimerization-EMT(154159)
(Twist, MMP2, 9 and 7)
2. IL-6-JAK/STAT3-EMT
3. CCL3-JAK/STAT3-EMT
4. TGF-β1-JAK/STAT3-Twist-E-Cadherin↓
5. TP53-/RB1-JAK/STAT3-EMT
6. snail-JAK/STAT3-EMT
stemness1. JAK/STAT3-Oct4 and SOX family(154,158,160)
transcription factor
2. TP53-/RB1-JAK/STAT3-stemness
3. AJAP1↓-JAK/STAT3-stemness
NED1.IL-6-JAK/STAT3-NE maker(CHGA and ENO 2)
2.IL-8-JAK/STAT3-NED
3. TP53-/RB1-JAK/STAT3-NED
4. IL-6-PEDF-RhoA-NF-κB-STAT3-NED
5.mTOR or MAPK pathway-STAT3
phosphodimerization-AR↓
6. IL-6-JAK/STAT3-AR↓(154,158,161,162)
PI3K/AKT/mTOREMT PI3K/AKT/mTOR-EMT(163165)
stemnesspAKT-H3K9 acetylation↓-CSCs(163,164,166)
NED PI3K/AKT/mTOR-AMPK-NED(167)
Hedgehog pathwayEMT Hedgehog-TGF-β1(168,169)
stemnessHedgehog-the transforms and maintains of CSCs(170172)
NED Hedgehog↓-PTCH-SMO↓-AR↓-NED(173)
Wnt pathwayEMT 1.Wnt/β-Catenin-EMT
2.PRKAR2B-Wnt-EMT(174,175)
stemness1.Wnt signaling enhances
CSC renewal and expansion
2.DAB 21 P↓-Wnt/β-Catenin-CSC
3.ESM1-β-catenin-tcf4-stemness
4. KIT-Wnt/β-catenin-Stemness(176179)
NED1.Wnt or AR↓-Wntless (WLS)-ROR2/
PKCδ/ERK-NED
2.AR↓-Wnt-11-NSE and ASCL1-NED
3.AR↓-YY1-FZD8-Wnt9A-FYN/STAT3-NED
4.Wnt/β-Catenin-TCF4-NED
5. ADT-TCF7L1-Wnt4-IL-8/CXCR2-NED
6. AR↓-PCDH-PC-Wnt-NED
7.Wnt-EZH2-microRNA-708↓-NED
8. ALK/N-Myc-Wnt/β-catenin-NED(180186)
NotchEMT1.Notch-EMT
2.Notch-HIF-1α-SNAI1(187,188)
stemnessNotch-stemness(189)
NEDDLL3-Notch 1 and Notch 2↓-Hes1 and Hey1↓-NED(190192)
TGF-βEMTTGF-β-PI3 K/AKT-EMT(193)
stemnessTGF-β-Smad-c-MYC and SPOP↓(194,195)
NED IL-6-TGF-β-SMAD2-p38MAPK-NED(196)
p38 MAPKEMTCAF-IL32-β3-p38 MAPK-EMT(197)
stemnessTPTEP1-MAPK14-P38 MAPK-stemness↓(198)
NEDp38 MAPK-FOXC2-NED(125)

B, Genes

NameRole Mechanisms(Refs.)

MAOAEMT CAFs-MAOA/mTOR/HIF-1α-EMT(199)
stemnessMAOA-stemness(84,200)
NED AR↓-MAOA-mTOR-HIF-1α-NED(84)
TP53EMT 1.TP53↓-EMT(E-cadherin, β-catenin, vimentin, fibronectin, slug, snail)
2. Wild-type p53-miR-145-EMT↓(201205)
stemness1. Wild-type p53-miR-145-stemness↓
2.TP53-Nanog, Sox2, Oct4 and c-Myc↓(135,136,204,205)
NED 1.LSD1-TP53↓-NED
2.TP53-/RB1-SOX2 or EZH2-NED(131,206)
RB1EMTRB1↓-EMT(207)
stemness RB1↓-E2F-Pluripotency factors (Oct4, Nanog, Sox2,(110)
KLF4, TCF3) and EZH2
NEDTP53-/RB1-SOX2 or EZH2-NED(131)
PTENEMT PTEN↓-PI3K/Akt-EMT(207,208)
stemness PTEN↓-PI3K/Akt-stemness(207,208)
NEDTP53-/PTEN- or PTEN-/RB1-NED(209)

C, Transcription factors

NameRole Mechanisms(Refs.)

FOXA2EMTFOXA2-EMT(27)
stemness EGFR-ERK-FOXA2-SOX9-stemness(210)
NED1.ZBTB46-FOXA2 and HIF1A-MCTP1-SNAI1-NED
2.PHF8-FOXA2-NED
3.Siah2-HIF-1α-FOXA2-Hes6, Sox9 and Jmjd1a-NED
4.FOXA2-KIT-NED(27,211214)
SOX2EMT SOX2-SPINK1-EMT(105)
stemness1.SOX2 is the cell maker of stem cell
2.SOX2-SPINK1-stemness(105)
NED 1.SOX2-SPINK1-NED
2.SOX2-LSD1-NED
3.SOX2-ASCL1-NED
4.ADT-SOX2-H19-NED
5.SRRM4-SOX2-NED
6.LIN28B-et-7-SOX2-HMGA2/HMGA2-NED
7.TRIM59-RB1-/P53-SOX2-NED
8.TRIB2-BRN2-SOX2-NED
9.TBX2-miR-200c-3p-SOX2-N-MYC-NED(80,87,105,106,109,111,215218)
PEG10EMT PEG10-TGF-β-Snail(88)
stemness-
NED 1.RB1-/TP53-PEG10-NE maker
2.ONECUT2-PEG10-NED(219221)
ASCL1EMT ASCL1-Wnt11-E-cadherin(222)
stemnessASCL1-SOX2, NANOG and OCT4-stemness(78)
NED1.ASCL1-CHGA, ENO2 (NSE), NCAM1, DLL1-NED
2.ASCL1-CEACAM5-NED
3.NOTCH-ASCL1-RB-p53(78,223,224)
MYCEMT TM4SF1-Wnt/β-catenin-c-MYC/SOX2-stemness and(225)
EMT
stemnessc-MYC and N-MYC are the cell maker of stem cell(45)
NED 1.c-MYC/Pim1-NED
2.N-MYC-EZH2-AR↓-NED
3.N-MYC-SYP and NSE(114,115,119)

D, Epigenetics

NameRole Mechanisms(Refs.)

EZH2EMTEZH2-EMT(226,227)
stemnessEZH2 is the cell maker of stem cell(45,228)
NED 1.Enz-AR-lncRNA-p21-EZH2-STAT3-NED(54,55,81,82,107,
2.EZH2-NMyc-AR↓116,141)
2.CREB-EZH2-TSP1-NED
3.E2F1-RACGAP1-EZH2-NED
4.ADT-AR-EHF-EZH2-NED
RESTEMT 1.KIT↓-REST-AR-SPINK1↓-EMT↓
2.REST-Twist1↓-EMT↓(105,148,179)
stemness 1.KIT↓-REST-AR-SPINK1↓-stemness
2.REST-CD44↓-stemness↓(105,148,179)
NED 1.KIT↓-REST-AR-SPINK1↓-NED↓(56,57,58,85,105,
2.PI3K/AKT↓-REST↓-NED150,179,221)
3.IL-6-REST-NED
4.REST↓-ONECUT2-NED
5.REST↓-MAOA-NED
6.HP1α-AR and REST↓-NED

E, Others

NameRole Mechanisms(Refs.)

MUC1-CEMTMUC1-C-NF-κB p65-ZEB1-EMT(143)
stemness 1.MUC1-C-E2F1-BAF-NOTCH1-HES1 and HEY1-
EMT and stemness(147,229)
2.MUC1-C-E2F1-BAF-NANOG-stemness
3.MUC1-C-MYC-BRN2-SOX2-stemness
NED1.MUC1-C-AR↓, p53↓, RB↓, N-Myc and EZH2-
NED
2.MUC1-C-MYC-BRN2-SOX2-NED
3.MUC1-C-NF-κB p65-EZH2-NED(110,229)

[i] Red text indicates core molecules. This table is not meant to be complete, as there are many other methods that simultaneously regulate EMT, stemness and NED. New ones are being discovered routinely. JAK/STAT, Janus Kinase/Signal Transducer and Activator of Transcription; EMT, Epithelial-to-Mesenchymal Transition; MMP, Matrix Metalloproteinase; IL-6, Interleukin 6; CCL3, C-C Motif Chemokine Ligand 3; TGF-β1, Transforming Growth Factor Beta 1; TP53, Tumor Protein p53; RB1, Retinoblastoma 1; NED, Neuroendocrine Differentiation; CHGA, Chromogranin A; ENO2, Enolase 2; PEDF, Pigment Epithelium-Derived Factor; RhoA, Ras Homolog Family Member A; NF-κB, Nuclear Factor Kappa B; mTOR, Mechanistic Target of Rapamycin; MAPK, Mitogen-Activated Protein Kinase; AR, Androgen Receptor; PI3K, Phosphoinositide 3-Kinase; AKT, Protein Kinase B; AMPK, AMP-Activated Protein Kinase; PTCH, Patched; SMO, Smoothened; PRKAR2B, Protein Kinase cAMP-Dependent Type II Regulatory Subunit Beta; DAB2IP, DAB2 Interacting Protein; ESM1, Endothelial Cell-Specific Molecule 1; TCF4, Transcription Factor 4; KIT, KIT Proto-Oncogene, Receptor Tyrosine Kinase; ROR2, Receptor Tyrosine Kinase Like Orphan Receptor 2; PKCδ, Protein Kinase C Delta; ERK, Extracellular Signal-Regulated Kinase; YY1, Yin Yang 1; FZD8, Frizzled Class Receptor 8; FYN, FYN Proto-Oncogene, Src Family Tyrosine Kinase; ADT, Androgen Deprivation Therapy; TCF7L1, Transcription Factor 7 Like 1; CXCR2, C-X-C Motif Chemokine Receptor 2; PCDH-PC, Protocadherin-PC; EZH2, Enhancer of Zeste Homolog 2; ALK, Anaplastic Lymphoma Kinase; N-Myc, N-Myc Proto-Oncogene; HIF-1α, Hypoxia-Inducible Factor 1 Alpha; SNAI1, Snail Family Transcriptional Repressor 1; DLL3, Delta Like Canonical Notch Ligand 3; Hes1, Hes Family BHLH Transcription Factor 1; Hey1, Hes Related Family BHLH Transcription Factor With YRPW Motif 1; Smad, Mothers Against Decapentaplegic Homolog; c-MYC, MYC Proto-Oncogene; SPOP, Speckle Type BTB/POZ Protein; CAF, Cancer-Associated Fibroblast; FOXC2, Forkhead Box C2; MAOA, Monoamine Oxidase A; LSD1, Lysine-Specific Demethylase 1; E2F, E2F Transcription Factor; KLF4, Kruppel-Like Factor 4; TCF3, Transcription Factor 3; PTEN, Phosphatase and Tensin Homolog; ZBTB46, Zinc Finger and BTB Domain Containing 46; PHF8, PHD Finger Protein 8; Siah2, Seven in Absentia Homolog 2; Jmjd1a, Jumonji Domain Containing 1A; SPINK1, Serine Peptidase Inhibitor Kazal Type 1; ASCL1, Achaete-Scute Family BHLH Transcription Factor 1; SRRM4, Serine/Arginine Repetitive Matrix 4; LIN28B, Lin-28 Homolog B; HMGA2, High Mobility Group AT-Hook 2; TRIM59, Tripartite Motif Containing 59; TRIB2, Tribbles Pseudokinase 2; BRN2, POU Class 3 Homeobox 2; TBX2, T-Box Transcription Factor 2; PEG10, Paternally Expressed 10; NCAM1, Neural Cell Adhesion Molecule 1; DLL1, Delta Like Canonical Notch Ligand 1; CEACAM5, Carcinoembryonic Antigen Related Cell Adhesion Molecule 5; TM4SF1, Transmembrane 4 L Six Family Member 1; SYP, Synaptophysin; lncRNA-p21, Long Non-Coding RNA p21; CREB, cAMP Response Element-Binding Protein; TSP1, Thrombospondin 1; RACGAP1, Rac GTPase Activating Protein 1; EHF, ETS Homologous Factor; CD44, CD44 Molecule; HP1α, Heterochromatin Protein 1 Alpha; BAF, BRG1/BRM-Associated Factor; HES1, Hes Family BHLH Transcription Factor 1; HEY1, Hes Related Family BHLH Transcription Factor With YRPW Motif 1.

Key genes such as PTEN, TP53 and RB1, which play pivotal roles in NED, are also closely associated with stemness and EMT. Mutations or deletions in the TP53 gene are present in 66.7% of CRPC-NE and 31.4% of CRPC-Adeno patients (134). TP53 acts as a suppressor of genes involved in stemness, including Sox2, Oct4, Nanog and c-Myc (135,136). Thus, mutations and deletions in TP53 can lead to the activation of stemness. PTEN also plays a critical role in regulating normal and CSC homeostasis, with its loss promoting CSC expansion in both solid and hematologic malignancies (137). The loss of TP53 and PTEN induces an increase in the NE phenotype, with NE differentiation in TP53- and PTEN-deficient CRPC being partially mediated by SOX11 (138).

RB1 loss is present in 70% of NEPC samples compared with 32% of CRPC-Adenocarcinoma samples (139). Rb, the protein encoded by RB1, can bind to E2F proteins to form the Rb/E2F complex and interactions through the E2F-binding pocket are essential for the role of Rb in silencing pluripotency genes. The Rb/E2F complex binds to gene promoter regions, directly repressing the expression of pluripotency factors, including SOX2, Oct4, Nanog, KLF4 and TCF3. Loss of Rb leads to dissociation and inactivation of the Rb/E2F complex, resulting in the loss of inhibition on pluripotency genes such as SOX2. ChIP analysis following Rb loss reveals significant changes in histone modifications across the genome, with increased levels of H3Ac and H4K4me3 and decreases levels of trimethylation of H3K27me3 in multiple pluripotency genes, such as Oct4, Nanog and SOX2, leading to enhanced chromatin accessibility (108,140). After dissociation, E2F becomes activated and promotes the expression of target genes. For example, E2F1 induces RACGAP1 expression and RACGAP1 stabilizes EZH2, a component of the PRC2 pathway, thereby promoting NED in prostate cancer (110,140141). In gene-modified mouse models, the individual loss or mutation of PTEN, TP53, or RB1 does not lead to NEPC (138,142). However, the simultaneous loss or mutation of any two of these tumor suppressor genes, RB1, TP53 and PTEN, markedly elevates the incidence of NEPC (130,131,138,142). Gene set enrichment analysis shows that the expression of E2F target genes and NE lineage genes is altered in tumors with dual-gene and triple-gene deletions, with increased expression of stemness-related genes such as SOX2 and EZH2. This may be closely related to the dissociation of the Rb/E2F protein complex or due to the loss of TP53 inhibitory effects. By inhibiting EZH2, AR expression in dual-gene mutant tumor cells can be increased and SYP expression can be decreased (130). Knocking out SOX2 can also reverse lineage conversion and restore resistance to enzalutamide (131). This highlights the important role of stemness-related programs in promoting NE differentiation in prostate cancer.

MUC1-C is a representative molecule that concurrently regulates EMT, stemness and NED. MUC1 is a transmembrane mucin glycoprotein expressed on the apical surface of normal epithelia, consisting of two subunits: MUC1-N and MUC1-C subunit complex (143). The upregulation of MUC1-C in AR-dependent prostate cancer cells (LNCaP and C4-2) drives the progression of NEPC by inhibiting the p53 pathway and activating MYC (144,145). Additionally, MUC1-C promotes EZH2 transcription by binding to NF-κB p65 at NF-κB consensus sites within the EZH2 intron 1 enhancer region (145). MUC1-C inhibits AR signaling through multiple mechanisms, including post-transcriptional regulation (involving miR-135b-mediated downregulation of AR mRNA levels) and competitive inhibition (MUC1-C directly interacts with the AR DNA-binding domain through its cytoplasmic domain to form a complex with AR) (146).

The MUC1-C-activated MYC-BRN2 pathway is associated with the induction of the pluripotency factor SOX2 and NED markers (ASCL1, AURKA and SYP), while this pathway operates independently of EZH2, KLF4, or OCT4 expression (110). Compared with the MUC1-C-MYC-BRN2 pathway, the function of the MUC1-C-E2F1-BAF network serves as a parallel and unique driver of NED. Furthermore, in addition to driving NE, the MUC1-C-E2F1-BAF complex (comprising BRG1, ARID1A, BAF60a, BAF155 and BAF170)-NOTCH1-NANOG pathway also promotes EMT, stemness and the self-renewal ability of CSCs (147). MUC1-C can directly activate the inflammatory NF-κB p65-ZEB1 pathway to induce EMT, which promotes cancer cell invasion and the acquisition of stem-like properties (143). SPINK1 induces EMT, stemness and cellular plasticity, playing a crucial role in maintaining the NE phenotype (105). Long-term loss of REST expression promotes EMT and stemness characteristics (148), while REST deficiency induces NED in prostate cancer cells by derepressing HOTAIR, MAOA and SPINK1 (149151). The mechanisms of other pathways and molecules inducing EMT, stemness and NED are summarized in Table I.

Conclusion and future perspectives

With the widespread adoption of ADT, NEPC has emerged as an increasingly critical clinical challenge. The malignancy progressively escalates from prostate adenocarcinoma to mixed types and ultimately to NEPC, with a corresponding decline in patient prognosis. Activated signaling pathways are also involved in EMT, CSC characteristics/stemness, DNA damage response, metabolic reprogramming, hypoxia, ferroptosis and other biological processes, all of which are closely related to NED. Among these, the widespread activation of EMT and stemness has attracted particular attention. Identifying and validating core EMT/stem cell signaling pathways could lead to actionable biology that may be translated into therapeutic interventions. Therefore, the present study revisited the origins, mechanisms and the biological significance of EMT and stemness. The poor prognosis, high metastatic potential and resistance to radiotherapy, chemotherapy and endocrine therapy observed in NEPC are closely associated with EMT and stemness. Specifically, EMT confers high metastatic potential in NEPC, while stemness enables post-metastatic colonization, resistance to radiotherapy and chemotherapy and lineage plasticity. NED represents the ability of NEPC to resist castration. The NE phenotype in prostate cancer does not necessarily imply high aggressiveness, as there are a number of pathological types with favorable prognosis. The NE molecular markers themselves hold limited clinical significance; rather, it is the activation of EMT and stemness during NED that endows NEPC with its distinct clinical characteristics. Studies have shown that mesenchymal and MSPC exhibits more advanced Gleason scores, pathological T stages and N stages compared with primary ARPC (69,152,153). A higher proportion of primary MSPC patients experience accelerated progression and die from the disease within 24 months of diagnosis (69).

EMT and stemness are critical characteristics of NEPC and represent essential aspects of studying highly aggressive prostate malignancies. In the future, EMT, stemness and NED should be considered as an integrated whole. Growing evidence indicates that conventional therapies frequently fail to eliminate cancer cells that have transitioned into a CSC state through the activation of EMT and NED programs, leading to CSC-mediated clinical relapse. The discovery and validation of potential core EMT/stemness signaling pathways in NEPC could uncover actionable biological mechanisms for therapeutic interventions.

Acknowledgements

Not applicable.

Funding

The present study was supported by the doctoral program of the first affiliated hospital of Chongqing Medical University (grant no. CYYY-79 BSYJSCXXM-202332).

Availability of data and materials

Not applicable.

Authors' contributions

YFL and DLW participated in conceptualization and study design. YFL, SS and LDS conducted the investigation and data collection. SS and JW drafted the initial manuscript. CW, JH, and YL contributed to reviewing and editing. KH and YL were responsible for visualization. DW and XG supervised the study and finalized the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

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, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Feldman BJ and Feldman D: The development of androgen-independent prostate cancer. Nat Rev Cancer. 1:34–45. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, et al: Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 371:424–433. 2014. View Article : Google Scholar : PubMed/NCBI

4 

de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, Chi KN, Jones RJ, Goodman OB Jr, Saad F, et al: Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 364:1995–2005. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Davies AH, Beltran H and Zoubeidi A: Cellular plasticity and the neuroendocrine phenotype in prostate cancer. Nat Rev Urol. 15:271–286. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Yamada Y and Beltran H: Clinical and biological features of neuroendocrine prostate cancer. Curr Oncol Rep. 23:152021. View Article : Google Scholar : PubMed/NCBI

7 

Zaffuto E, Pompe R, Zanaty M, Bondarenko HD, Leyh-Bannurah SR, Moschini M, Dell'Oglio P, Gandaglia G, Fossati N, Stabile A, et al: Contemporary incidence and cancer control outcomes of primary neuroend ocrine prostate cancer: A SEER database analysis. Clin Genitourin Cancer. 15:e793–e800. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Varga J and Greten FR: Cell plasticity in epithelial homeostasis and tumorigenesis. Nat Cell Biol. 19:1133–1141. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Wang Y and Wang Y, Ci X, Choi SYC, Crea F, Lin D and Wang Y: Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol. 18:581–596. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Garabedian EM, Humphrey PA and Gordon JI: A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells. Proc Natl Acad Sci USA. 95:15382–15387. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Merkens L, Sailer V, Lessel D, Janzen E, Greimeier S, Kirfel J, Perner S, Pantel K, Werner S and von Amsberg G: Aggressive variants of prostate cancer: Underlying mechanisms of neuroendocrine transdifferentiation. J Exp Clin Cancer Res. 41:462022. View Article : Google Scholar : PubMed/NCBI

12 

Varma M, Lee MW, Tamboli P, Zarbo RJ, Jimenez RE, Salles PGO and Amin MB: Morphologic criteria for the diagnosis of prostatic adenocarcinoma in needle biopsy specimens. A study of 250 consecutive cases in a routine surgical pathology practice. Arch Pathol Lab Med. 126:554–561. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Epstein JI, Amin MB, Beltran H, Lotan TL, Mosquera JM, Reuter VE, Robinson BD, Troncoso P and Rubin MA: Proposed morphologic classification of prostate cancer with neuroendocrine differentiation. Am J Surg Pathol. 38:756–767. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Spetsieris N, Boukovala M, Patsakis G, Alafis I and Efstathiou E: Neuroendocrine and aggressive-variant prostate cancer. Cancers. 12:37922014. View Article : Google Scholar

15 

Weinstein MH, Partin AW, Veltri RW and Epstein JI: Neuroendocrine differentiation in prostate cancer: Enhanced prediction of progression after radical prostatectomy. Hum Pathol. 27:683–687. 1996. View Article : Google Scholar : PubMed/NCBI

16 

Theodorescu D, Broder SR, Boyd JC, Mills SE and Frierson HF Jr: Cathepsin D and chromogranin A as predictors of long term disease specific survival after radical prostatectomy for localized carcinoma of the prostate. Cancer. 80:2109–2119. 1997. View Article : Google Scholar : PubMed/NCBI

17 

Berruti A, Mosca A, Tucci M, Terrone C, Torta M, Tarabuzzi R, Russo L, Cracco C, Bollito E, Scarpa RM, et al: Independent prognostic role of circulating chromogranin A in prostate cancer patients with hormone-refractory disease. Endocr Relat Cancer. 12:109–117. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Bostwick DG, Qian J, Pacelli A, Zincke H, Blute M, Bergstralh EJ, Slezak JM and Cheng L: Neuroendocrine expression in node positive prostate cancer: Correlation with systemic progression and patient survival. J Urol. 168:1204–1211. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Conteduca V, Oromendia C, Eng KW, Bareja R, Sigouros M, Molina A, Faltas BM, Sboner A, Mosquera JM, Elemento O, et al: Clinical features of neuroendocrine prostate cancer. Eur J Cancer. 121:7–18. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Wang HT, Yao YH, Li BG, Tang Y, Chang JW and Zhang J: Neuroendocrine Prostate Cancer (NEPC) progressing from conventional pr ostatic adenocarcinoma: Factors associated with time to development of NEPC and survival from NEPC diagnosis-a systematic review and pooled analysis. J Clin Oncol. 32:3383–3390. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Montanari M, Rossetti S, Cavaliere C, D'Aniello C, Malzone MG, Vanacore D, Di Franco R, La Mantia E, Iovane G, Piscitelli R, et al: Epithelial-mesenchymal transition in prostate cancer: An overview. Oncotarget. 8:35376–35389. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Kalluri R and Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 112:1776–1784. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Williams ED, Gao D, Redfern A and Thompson EW: Controversies around epithelial-mesenchymal plasticity in cancer metastasis. Nat Rev Cancer. 19:716–732. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Tiwari R, Manzar N and Ateeq B: Dynamics of cellular plasticity in prostate cancer progression. Front Mol Biosci. 7:1302020. View Article : Google Scholar : PubMed/NCBI

25 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Bakir B, Chiarella AM, Pitarresi JR and Rustgi AK: EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol. 30:764–776. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Liu YN, Chen WY, Yeh HL, Chen WH, Jiang KC, Li HR, Dung PVT, Chen ZQ, Lee WJ, Hsiao M, et al: MCTP1 increases the malignancy of androgen-deprived prostate cancer cells by inducing neuroendocrine differentiation and EMT. Sci Signal. 17:eadc91422024. View Article : Google Scholar : PubMed/NCBI

28 

Conteduca V, Aieta M, Amadori D and De Giorgi U: Neuroendocrine differentiation in prostate cancer: Current and emerging therapy strategies. Crit Rev Oncol Hematol. 92:11–24. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

30 

Capp JP: Cancer stem cells: From historical roots to a new perspective. J Oncol. 2019:51892322019. View Article : Google Scholar : PubMed/NCBI

31 

Phi LTH, Sari IN, Yang YG, Lee SH, Jun N, Kim KS, Lee YK and Kwon HY: Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 2018:54169232018. View Article : Google Scholar : PubMed/NCBI

32 

Ayob AZ and Ramasamy TS: Cancer stem cells as key drivers of tumour progression. J Biomed Sci. 25:202018. View Article : Google Scholar : PubMed/NCBI

33 

Nguyen LV, Vanner R, Dirks P and Eaves CJ: Cancer stem cells: An evolving concept. Nat Rev Cancer. 12:133–143. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Pekovic V and Hutchison CJ: Adult stem cell maintenance and tissue regeneration in the ageing context: The role for A-type lamins as intrinsic modulators of ageing in adult stem cells and their niches. J Anat. 213:5–25. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Ojo D, Lin X, Wong N, Gu Y and Tang D: Prostate cancer stem-like cells contribute to the development of castration-resistant prostate cancer. Cancers (Basel). 7:2290–2308. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Brown TJ and James V: The role of extracellular vesicles in the development of a cancer stem cell microenvironment niche and potential therapeutic targets: A systematic review. Cancers (Basel). 13:24352021. View Article : Google Scholar : PubMed/NCBI

37 

Plaks V, Kong N and Werb Z: The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 16:225–238. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Castellón EA, Indo S and Contreras HR: Cancer Stemness/epithelial-mesenchymal transition axis influences metastasis and castration resistance in prostate cancer: Potential therapeutic target. Int J Mol Sci. 23:149172022. View Article : Google Scholar : PubMed/NCBI

39 

Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11:12802020. View Article : Google Scholar : PubMed/NCBI

40 

Xin L, Lawson DA and Witte ON: The Sca-1 cell surface marker enriches for a prostate-regenerating cell subpopulation that can initiate prostate tumorigenesis. Proc Natl Acad Sci USA. 102:6942–6947. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Germann M, Wetterwald A, Guzmán-Ramirez N, van der Pluijm G, Culig Z, Cecchini MG, Williams ED and Thalmann GN: Stem-like cells with luminal progenitor phenotype survive castration in human prostate cancer. Stem Cells. 30:1076–1086. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Ma F, Chen D, Chen F, Chi Y and Han Z, Feng X, Li X and Han Z: Human umbilical cord mesenchymal stem cells promote breast cancer metastasis by Interleukin-8- and Interleukin-6-Dependent Induction of CD44(+)/CD24(−) cells. Cell Transplant. 24:2585–2599. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Ji Y, Liu B, Chen L, Li A, Shen K, Su R, Zhang W, Zhu Y, Wang Q and Xue W: Repurposing ketotifen as a therapeutic strategy for neuroendocrine prostate cancer by targeting the IL-6/STAT3 pathway. Cell Oncol (Dordr). 46:1445–1456. 2023. View Article : Google Scholar : PubMed/NCBI

44 

Kopczyńska E: Role of microRNAs in the resistance of prostate cancer to docetaxel and paclitaxel. Contemp Oncol (Pozn). 19:423–427. 2015.PubMed/NCBI

45 

Verma P, Shukla N, Kumari S, Ansari MS, Gautam NK and Patel GK: Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer. 1878:1888872023. View Article : Google Scholar : PubMed/NCBI

46 

Castillo V, Valenzuela R, Huidobro C, Contreras HR and Castellon EA: Functional characteristics of cancer stem cells and their role in drug resistance of prostate cancer. Int J Oncol. 45:985–994. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Biserova K, Jakovlevs A, Uljanovs R and Strumfa I: Cancer stem cells: Significance in origin, pathogenesis and treatment of glioblastoma. Cells. 10:6212021. View Article : Google Scholar : PubMed/NCBI

48 

Garcia-Mayea Y, Mir C, Masson F, Paciucci R and ME LL: Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin Cancer Biol. 60:166–180. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Satar NA, Fakiruddin KS, Lim MN, Mok PL, Zakaria N, Fakharuzi NA, Abd Rahman AZ, Zakaria Z, Yahaya BH and Baharuddin P: Novel triple-positive markers identified in human non-small cell lung cancer cell line with chemotherapy-resistant and putative cancer stem cell characteristics. Oncol Rep. 40:669–681. 2018.PubMed/NCBI

50 

Duran GE, Wang YC, Francisco EB, Rose JC, Martinez FJ, Coller J, Brassard D, Vrignaud P and Sikic BI: Mechanisms of resistance to cabazitaxel. Mol Cancer Ther. 14:193–201. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Schreiber RD, Old LJ and Smyth MJ: Cancer immunoediting: Integrating immunity's roles in cancer suppression and promotion. Science. 331:1565–1570. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Zhang D, Sun B, Zhao X, Ma Y, Ji R, Gu Q, Dong X, Li J, Liu F, Jia X, et al: Twist1 expression induced by sunitinib accelerates tumor cell vasculogenic mimicry by increasing the population of CD133+ cells in triple-negative breast cancer. Mol Cancer. 13:2072014. View Article : Google Scholar : PubMed/NCBI

53 

Liu B, Li L, Yang G, Geng C, Luo Y, Wu W, Manyam GC, Korentzelos D, Park S, Tang Z, et al: PARP inhibition suppresses GR-MYCN-CDK5-RB1-E2F1 signaling and neuroendocrine differentiation in castration-resistant prostate cancer. Clin Cancer Res. 25:6839–6851. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Wang Z, Zhao Y, An Z and Li W: Molecular links between angiogenesis and neuroendocrine phenotypes in prostate cancer progression. Front Oncol. 9:14912020. View Article : Google Scholar : PubMed/NCBI

55 

Zhang Y, Zheng D, Zhou T, Song H, Hulsurkar M, Su N, Liu Y, Wang Z, Shao L, Ittmann M, et al: Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers. Nat Commun. 9:40802018. View Article : Google Scholar : PubMed/NCBI

56 

Ci X, Hao J, Dong X, Choi SY, Xue H, Wu R, Qu S, Gout PW, Zhang F, Haegert AM, et al: Heterochromatin protein 1α mediates development and aggressiveness of neuroendocrine prostate cancer. Cancer Res. 78:2691–2704. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Chang PC, Wang TY, Chang YT, Chu CY, Lee CL, Hsu HW, Zhou TA, Wu Z, Kim RH, Desai SJ, et al: Autophagy pathway is required for IL-6 induced neuroendocrine differen tiation and chemoresistance of prostate cancer LNCaP cells. PLoS One. 9:e885562014. View Article : Google Scholar : PubMed/NCBI

58 

Zhu Y, Liu C, Cui Y, Nadiminty N, Lou W and Gao AC: Interleukin-6 induces neuroendocrine differentiation (NED) through sup pression of RE-1 silencing transcription factor (REST). Prostate. 74:1086–1094. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Fang D and Kitamura H: Cancer stem cells and epithelial-mesenchymal transition in urothelial carcinoma: Possible pathways and potential therapeutic approaches. Int J Urol. 25:7–17. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Hiraga T, Ito S and Nakamura H: EpCAM expression in breast cancer cells is associated with enhanced bone metastasis formation. Int J Cancer. 138:1698–1708. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Rycaj K, Li H, Zhou J, Chen X and Tang DG: Cellular determinants and microenvironmental regulation of prostate cancer metastasis. Semin Cancer Biol. 44:83–97. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Jeon HM and Lee J: MET: Roles in epithelial-mesenchymal transition and cancer stemness. Ann Transl Med. 5:52017. View Article : Google Scholar : PubMed/NCBI

63 

Ruppender NS, Morrissey C, Lange PH and Vessella RL: Dormancy in solid tumors: Implications for prostate cancer. Cancer Metastasis Rev. 32:501–509. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Li H and Tang DG: Prostate cancer stem cells and their potential roles in metastasis. J Surg Oncol. 103:558–562. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Celià-Terrassa T and Kang Y: Metastatic niche functions and therapeutic opportunities. Nat Cell Biol. 20:868–877. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, Ziegler AM, et al: Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest. 121:1298–1312. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Kim JK, Jung Y, Wang J, Joseph J, Mishra A, Hill EE, Krebsbach PH, Pienta KJ, Shiozawa Y and Taichman RS: TBK1 regulates prostate cancer dormancy through mTOR inhibition. Neoplasia. 15:1064–1074. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Correnti M and Raggi C: Stem-like plasticity and heterogeneity of circulating tumor cells: Current status and prospect challenges in liver cancer. Oncotarget. 8:7094–7115. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Han H, Wang Y, Curto J, Gurrapu S, Laudato S, Rumandla A, Chakraborty G, Wang X, Chen H, Jiang Y, et al: Mesenchymal and stem-like prostate cancer linked to therapy-induced lineage plasticity and metastasis. Cell Rep. 39:1105952022. View Article : Google Scholar : PubMed/NCBI

70 

Wang H and Unternaehrer JJ: Epithelial-mesenchymal transition and cancer stem cells: At the crossroads of differentiation and dedifferentiation. Dev Dyn. 248:10–20. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Li P, Wang J, Chu M, Zhang K, Yang R and Gao WQ: Zeb1 promotes androgen independence of prostate cancer via induction of stem cell-like properties. Exp Biol Med (Maywood). 239:813–822. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Tsai YC, Chen WY, Abou-Kheir W, Zeng T, Yin JJ, Bahmad H, Lee YC and Liu YN: Androgen deprivation therapy-induced epithelial-mesenchymal transition of prostate cancer through downregulating SPDEF and activating CCL2. Biochim Biophys Acta Mol Basis Dis. 1864:1717–1727. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Sun Y, Wang BE, Leong KG, Yue P, Li L, Jhunjhunwala S, Chen D, Seo K, Modrusan Z, Gao WQ, et al: Androgen deprivation causes epithelial-mesenchymal transition in the prostate: Implications for androgen-deprivation therapy. Cancer Res. 72:527–536. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Tang Y, Hamburger AW, Wang L, Khan MA and Hussain A: Androgen deprivation and stem cell markers in prostate cancers. Int J Clin Exp Pathol. 3:128–138. 2009.PubMed/NCBI

75 

Qin J, Liu X, Laffin B, Chen X, Choy G, Jeter CR, Calhoun-Davis T, Li H, Palapattu GS, Pang S, et al: The PSA(−/lo) prostate cancer cell population harbors self-renewing long-term tumor-propagating cells that resist castration. Cell Stem Cell. 10:556–569. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Rybak AP, He L, Kapoor A, Cutz JC and Tang D: Characterization of sphere-propagating cells with stem-like properties from DU145 prostate cancer cells. Biochim Biophys Acta. 1813:683–694. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Han M, Li F, Zhang Y, Dai P, He J, Li Y, Zhu Y, Zheng J, Huang H, Bai F, et al: FOXA2 drives lineage plasticity and KIT pathway activation in neuroend ocrine prostate cancer. Cancer Cell. 40:1306–1323.e8. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Nouruzi S, Ganguli D, Tabrizian N, Kobelev M, Sivak O, Namekawa T, Thaper D, Baca SC, Freedman ML, Aguda A, et al: ASCL1 activates neuronal stem cell-like lineage programming through re modeling of the chromatin landscape in prostate cancer. Natu Commun. 13:22822022. View Article : Google Scholar : PubMed/NCBI

79 

Wang Z, Wang T, Hong D, Dong B, Wang Y, Huang H, Zhang W, Lian B, Ji B, Shi H, et al: Single-cell transcriptional regulation and genetic evolution of neuroe ndocrine prostate cancer. iScience. 25:1045762022. View Article : Google Scholar : PubMed/NCBI

80 

Singh N, Ramnarine VR, Song JH, Pandey R, Padi SKR, Nouri M, Olive V, Kobelev M, Okumura K, McCarthy D, et al: The long noncoding RNA H19 regulates tumor plasticity in neuroendocrin e prostate cancer. Nat Commun. 12:73492021. View Article : Google Scholar : PubMed/NCBI

81 

Long Z, Deng L, Li C, He Q, He Y, Hu X, Cai Y and Gan Y: Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer. Cell Death Dis. 12:462021. View Article : Google Scholar : PubMed/NCBI

82 

Luo J, Wang K, Yeh S, Sun Y, Liang L, Xiao Y, Xu W, Niu Y, Cheng L, Maity SN, et al: LncRNA-p21 alters the antiandrogen enzalutamide-induced prostate cancer neuroendocrine differentiation via modulating the EZH2/STAT3 signaling. Nat Commun. 10:25712019. View Article : Google Scholar : PubMed/NCBI

83 

Kaarijärvi R, Kaljunen H, Nappi L, Fazli L, Kung SHY, Hartikainen JM, Paakinaho V, Capra J, Rilla K, Malinen M, et al: DPYSL5 is highly expressed in treatment-induced neuroendocrine prostate cancer and promotes lineage plasticity via EZH2/PRC2. Commun Biol. 7:1082024. View Article : Google Scholar : PubMed/NCBI

84 

Shui X, Ren X, Xu R, Xie Q, Hu Y, Qin J, Meng H, Zhang C, Zhao J and Shi C: Monoamine oxidase A drives neuroendocrine differentiation in prostate cancer. Biochem Biophys Res Commun. 606:135–141. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Chen R, Li Y, Buttyan R and Dong X: Implications of PI3K/AKT inhibition on REST protein stability and neur oendocrine phenotype acquisition in prostate cancer cells. Oncotarget. 8:84863–84876. 2022. View Article : Google Scholar : PubMed/NCBI

86 

Bishop JL, Thaper D, Vahid S, Davies A, Ketola K, Kuruma H, Jama R, Nip KM, Angeles A, Johnson F, et al: The master neural transcription factor BRN2 is an androgen receptor-suppressed driver of neuroendocrine differentiation in prostate cancer. Cancer Discov. 7:54–71. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Nie J, Zhang P, Liang C, Yu Y and Wang X: ASCL1-mediated ferroptosis resistance enhances the progress of castrat ion-resistant prostate cancer to neurosecretory prostate cancer. Free Radic Biol Med. 205:318–331. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Akamatsu S, Wyatt AW, Lin D, Lysakowski S, Zhang F, Kim S, Tse C, Wang K, Mo F, Haegert A, et al: The placental gene PEG10 promotes progression of neuroendocrine prosta te cancer. Cell reports. 12:922–936. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Deep G, Jain AK, Ramteke A, Ting H, Vijendra KC, Gangar SC, Agarwal C and Agarwal R: SNAI1 is critical for the aggressiveness of prostate cancer cells with low E-cadherin. Mol Cancer. 13:372014. View Article : Google Scholar : PubMed/NCBI

90 

Bae KM, Su Z, Frye C, McClellan S, Allan RW, Andrejewski JT, Kelley V, Jorgensen M, Steindler DA, Vieweg J, et al: Expression of pluripotent stem cell reprogramming factors by prostate tumor initiating cells. J Urol. 183:2045–2053. 2010. View Article : Google Scholar : PubMed/NCBI

91 

Bae KM, Parker NN, Dai Y, Vieweg J and Siemann DW: E-cadherin plasticity in prostate cancer stem cell invasion. Am J Cancer Res. 1:71–84. 2011.PubMed/NCBI

92 

Orellana-Serradell O, Herrera D, Castellon EA and Contreras HR: The transcription factor ZEB1 promotes an aggressive phenotype in prostate cancer cell lines. Asian J Androl. 20:294–299. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, D'Alessio AC, Young RA and Weinberg RA: Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 154:61–74. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Kong D, Banerjee S, Ahmad A, Li Y, Wang Z, Sethi S and Sarkar FH: Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PLoS One. 5:e124452010. View Article : Google Scholar : PubMed/NCBI

95 

Onder TT, Gupta PB, Mani SA, Yang J, Lander ES and Weinberg RA: Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68:3645–3654. 2008. View Article : Google Scholar : PubMed/NCBI

96 

Akunuru S, James Zhai Q and Zheng Y: Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell Death Dis. 3:e3522012. View Article : Google Scholar : PubMed/NCBI

97 

Soundararajan R, Paranjape AN, Maity S, Aparicio A and Mani SA: EMT, stemness and tumor plasticity in aggressive variant neuroendocrine prostate cancers. Biochim Biophys Acta Rev Cancer. 1870:229–238. 2018. View Article : Google Scholar : PubMed/NCBI

98 

McKeithen D, Graham T, Chung LW and Odero-Marah V: Snail transcription factor regulates neuroendocrine differentiation in LNCaP prostate cancer cells. Prostate. 70:982–992. 2010. View Article : Google Scholar : PubMed/NCBI

99 

Dicken H, Hensley PJ and Kyprianou N: Prostate tumor neuroendocrine differentiation via EMT: The road less traveled. Asian J Urol. 6:82–90. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, et al: Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 122:947–956. 2005. View Article : Google Scholar : PubMed/NCBI

101 

Sarkar A and Hochedlinger K: The sox family of transcription factors: Versatile regulators of stem and progenitor cell fate. Cell Stem Cell. 12:15–30. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Zhang S and Cui W: Sox2, a key factor in the regulation of pluripotency and neural differ entiation. World J Stem Cells. 6:305–311. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Metz EP, Wilder PJ, Dong J, Datta K and Rizzino A: Elevating SOX2 in prostate tumor cells upregulates expression of neuro endocrine genes, but does not reduce the inhibitory effects of enzalut amide. J Cell Physiol. 235:3731–3740. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Kwon OJ, Zhang L, Jia D and Xin L: Sox2 is necessary for androgen ablation-induced neuroendocrine differe ntiation from Pten null Sca-1+ prostate luminal cells. Oncogene. 40:203–214. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Tiwari R, Manzar N, Bhatia V, Yadav A, Nengroo MA, Datta D, Carskadon S, Gupta N, Sigouros M, Khani F, et al: Androgen deprivation upregulates SPINK1 expression and potentiates cel lular plasticity in prostate cancer. Nature Commun. 11:3842020. View Article : Google Scholar : PubMed/NCBI

106 

Li H, Wang L, Li Z, Geng X, Li M, Tang Q, Wu C and Lu Z: SOX2 has dual functions as a regulator in the progression of neuroendo crine prostate cancer. Lab Invest. 100:570–582. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Kregel S, Kiriluk KJ, Rosen AM, Cai Y, Reyes EE, Otto KB, Tom W, Paner GP, Szmulewitz RZ and Vander Griend DJ: Sox2 is an androgen receptor-repressed gene that promotes castration-r esistant prostate cancer. PLoS One. 8:e537012013. View Article : Google Scholar : PubMed/NCBI

108 

Kareta MS, Gorges LL, Hafeez S, Benayoun BA, Marro S, Zmoos AF, Cecchini MJ, Spacek D, Batista LF, O'Brien M, et al: Inhibition of pluripotency networks by the Rb tumor suppressor restricts reprogramming and tumorigenesis. Cell Stem Cell. 16:39–50. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Lovnicki J, Gan Y, Feng T, Li Y, Xie N, Ho CH, Lee AR, Chen X, Nappi L, Han B, et al: LIN28B promotes the development of neuroendocrine prostate cancer. J Clin Invest. 130:5338–5348. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Yasumizu Y, Rajabi H, Jin C, Hata T, Pitroda S, Long MD, Hagiwara M, Li W, Hu Q, Liu S, et al: MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat Commun. 11:3382020. View Article : Google Scholar : PubMed/NCBI

111 

Patel GK, Dutta S, Syed MM, Ramachandran S, Sharma M, Rajamanickam V, Ganapathy V, DeGraff DJ, Pruitt K, Tripathi M, et al: TBX2 Drives Neuroendocrine Prostate Cancer through Exosome-Mediated Re pression of miR-200c-3p. Cancers (Basel). 13:50202021. View Article : Google Scholar : PubMed/NCBI

112 

Sotomayor P, Godoy A, Smith GJ and Huss WJ: Oct4A is expressed by a subpopulation of prostate neuroendocrine cells. Prostate. 69:401–410. 2009. View Article : Google Scholar : PubMed/NCBI

113 

Quintanal-Villalonga A, Kawasaki K, Redin E, Uddin F, Rakhade S, Durani V, Sabet A, Shafer M, Karthaus WR, Zaidi S, et al: CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation. Signal Transduct Target Ther. 9:1892024. View Article : Google Scholar : PubMed/NCBI

114 

Dardenne E, Beltran H, Benelli M, Gayvert K, Berger A, Puca L, Cyrta J, Sboner A, Noorzad Z, MacDonald T, et al: N-Myc Induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell. 30:563–577. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Berger A, Brady NJ, Bareja R, Robinson B, Conteduca V, Augello MA, Puca L, Ahmed A, Dardenne E, Lu X, et al: N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest. 129:3924–3940. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, Wang Y, Sheikh KL, Terry S, Tagawa ST, et al: Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 1:487–495. 2011. View Article : Google Scholar : PubMed/NCBI

117 

Otto T, Horn S, Brockmann M, Eilers U, Schüttrumpf L, Popov N, Kenney AM, Schulte JH, Beijersbergen R, Christiansen H, et al: Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell. 15:67–78. 2009. View Article : Google Scholar : PubMed/NCBI

118 

Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al: Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell. 24:75–89. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Quintanal-Villalonga Á, Chan JM, Yu HA, Pe'er D, Sawyers CL, Sen T and Rudin CM: Lineage plasticity in cancer: A shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 17:360–371. 2020. View Article : Google Scholar : PubMed/NCBI

120 

McQuillen CN and Brady NJ: ASCL1 drives the development of neuroendocrine prostate cancer. Cancer Res. 84:3499–3501. 2024. View Article : Google Scholar : PubMed/NCBI

121 

Davies A, Zoubeidi A and Selth LA: The epigenetic and transcriptional landscape of neuroendocrine prostate cancer. Endocr Relat Cancer. 27:R35–R50. 2024. View Article : Google Scholar : PubMed/NCBI

122 

Clermont PL, Lin D, Crea F, Wu R, Xue H, Wang Y, Thu KL, Lam WL, Collins CC, Wang Y, et al: Polycomb-mediated silencing in neuroendocrine prostate cancer. Clin Epigenetics. 7:402015. View Article : Google Scholar : PubMed/NCBI

123 

Donaldson-Collier MC, Sungalee S, Zufferey M, Tavernari D, Katanayeva N, Battistello E, Mina M, Douglass KM, Rey T, Raynaud F, et al: EZH2 oncogenic mutations drive epigenetic, transcriptional, and struct ural changes within chromatin domains. Nat Genet. 51:517–528. 2019. View Article : Google Scholar : PubMed/NCBI

124 

Sreekumar A and Saini S: Role of transcription factors and chromatin modifiers in driving linea ge reprogramming in treatment-induced neuroendocrine prostate cancer. Front Cell Dev Biol. 11:10757072023. View Article : Google Scholar : PubMed/NCBI

125 

Paranjape AN, Soundararajan R, Werden SJ, Joseph R, Taube JH, Liu H, Rodriguez-Canales J, Sphyris N, Wistuba I, Miura N, et al: Inhibition of FOXC2 restores epithelial phenotype and drug sensitivity in prostate cancer cells with stem-cell properties. Oncogene. 35:5963–5976. 2016. View Article : Google Scholar : PubMed/NCBI

126 

Zhou Q, Yang M, Fu J, Sun X, Wang J, Zhang H, Hu J and Han B: KIF1A promotes neuroendocrine differentiation in prostate cancer by regulating the OGT-mediated O-GlcNAcylation. Cell Death Dis. 15:7962024. View Article : Google Scholar : PubMed/NCBI

127 

Ward C, Volpe G, Cauchy P, Ptasinska A, Almaghrabi R, Blakemore D, Nafria M, Kestner D, Frampton J, Murphy G, et al: Fine-tuning mybl2 is required for proper mesenchymal-to-epithelial transition during somatic reprogramming. Cell Rep. 24:1496–1511.e8. 2018. View Article : Google Scholar : PubMed/NCBI

128 

German B, Alaiwi SA, Ho KL, Nanda JS, Fonseca MA, Burkhart DL, Sheahan AV, Bergom HE, Morel KL, Beltran H, et al: MYBL2 drives prostate cancer plasticity: Inhibiting its transcriptional target CDK2 for RB1-deficient neuroendocrine prostate cancer. Cancer Res Commun. 4:2295–2307. 2024. View Article : Google Scholar : PubMed/NCBI

129 

Hsieh CL, Do AD, Hsueh CY, Raboshakga MO, Thanh TN, Tai TT, Kung HJ and Sung SY: L1CAM mediates neuroendocrine phenotype acquisition in prostate cancer cells. Prostate. 84:1434–1447. 2024. View Article : Google Scholar : PubMed/NCBI

130 

Ku SY, Rosario S, Wang Y, Mu P, Seshadri M, Goodrich ZW, Goodrich MM, Labbé DP, Gomez EC, Wang J, et al: Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science. 355:78–83. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Mu P, Zhang Z, Benelli M, Karthaus WR, Hoover E, Chen CC, Wongvipat J, Ku SY, Gao D, Cao Z, et al: SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science. 355:84–88. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Beltran H, Oromendia C, Danila DC, Montgomery B, Hoimes C, Szmulewitz RZ, Vaishampayan U, Armstrong AJ, Stein M, Pinski J, et al: A Phase II Trial of the aurora kinase a inhibitor alisertib for patients with castration-resistant and neuroendocrine prostate cancer: Efficacy and biomarkers. Clin Cancer Res. 25:43–51. 2019. View Article : Google Scholar : PubMed/NCBI

133 

Blackhall F, Jao K, Greillier L, Cho BC, Penkov K, Reguart N, Majem M, Nackaerts K, Syrigos K, Hansen K, et al: Efficacy and safety of rovalpituzumab tesirine compared with topotecan as second-line therapy in DLL3-High SCLC: Results from the phase 3 TAHOE study. J Thorac Oncol. 16:1547–1558. 2021. View Article : Google Scholar : PubMed/NCBI

134 

Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, Marotz C, Giannopoulou E, Chakravarthi BV, Varambally S, et al: Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 22:298–305. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Takahashi K and Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. 2006. View Article : Google Scholar : PubMed/NCBI

136 

Lin T, Chao C, Saito S, Mazur SJ, Murphy ME, Appella E and Xu Y: p53 induces differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nat Cell Biol. 7:165–171. 2005. View Article : Google Scholar : PubMed/NCBI

137 

Luongo F, Colonna F, Calapà F, Vitale S, Fiori ME and De Maria R: PTEN Tumor-suppressor: The dam of stemness in cancer. Cancers (Basel). 11:10762019. View Article : Google Scholar : PubMed/NCBI

138 

Zou M, Toivanen R, Mitrofanova A, Floch N, Hayati S, Sun Y, Le Magnen C, Chester D, Mostaghel EA, Califano A, et al: Transdifferentiation as a mechanism of treatment resistance in a mouse model of Castration-resistant prostate cancer. Cancer Discov. 7:736–749. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Tan HL, Sood A, Rahimi HA, Wang W, Gupta N, Hicks J, Mosier S, Gocke CD, Epstein JI, Netto GJ, et al: Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin Cancer Res. 20:890–903. 2014. View Article : Google Scholar : PubMed/NCBI

140 

Bracken AP, Pasini D, Capra M, Prosperini E, Colli E and Helin K: EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer. EMBO J. 22:5323–5335. 2003. View Article : Google Scholar : PubMed/NCBI

141 

Song Z, Cao Q, Guo B, Zhao Y, Li X, Lou N, Zhu C, Luo G, Peng S, Li G, et al: Overexpression of RACGAP1 by E2F1 promotes neuroendocrine differentiat ion of prostate cancer by stabilizing EZH2 expression. Aging Dis. 14:1757–1774. 2023. View Article : Google Scholar : PubMed/NCBI

142 

Zhou Z, Flesken-Nikitin A, Corney DC, Wang W, Goodrich DW, Roy-Burman P and Nikitin AY: Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer Res. 66:7889–7898. 2006. View Article : Google Scholar : PubMed/NCBI

143 

Rajabi H and Kufe D: MUC1-C Oncoprotein integrates a program of EMT, epigenetic reprogramming and immune evasion in human carcinomas. Biochim Biophys Acta Rev Cancer. 1868:117–122. 2017. View Article : Google Scholar : PubMed/NCBI

144 

Tong D: Unravelling the molecular mechanisms of prostate cancer evolution from genotype to phenotype. Crit Rev Oncol Hematol. 163:1033702021. View Article : Google Scholar : PubMed/NCBI

145 

Rajabi H, Hiraki M and Kufe D: MUC1-C activates polycomb repressive complexes and downregulates tumor suppressor genes in human cancer cells. Oncogene. 37:2079–2088. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Rajabi H, Ahmad R, Jin C, Joshi MD, Guha M, Alam M, Kharbanda S and Kufe D: MUC1-C oncoprotein confers androgen-independent growth of human prostate cancer cells. Prostate. 72:1659–1668. 2012. View Article : Google Scholar : PubMed/NCBI

147 

Hagiwara M, Yasumizu Y, Yamashita N, Rajabi H, Fushimi A, Long MD, Li W, Bhattacharya A, Ahmad R, Oya M, et al: MUC1-C activates the BAF (mSWI/SNF) complex in prostate cancer stem cells. Cancer Res. 81:1111–1122. 2021. View Article : Google Scholar : PubMed/NCBI

148 

Chang YT, Lin TP, Campbell M, Pan CC, Lee SH, Lee HC, Yang MH, Kung HJ and Chang PC: REST is a crucial regulator for acquiring EMT-like and stemness phenotypes in hormone-refractory prostate cancer. Sci Rep. 7:427952017. View Article : Google Scholar : PubMed/NCBI

149 

Chang YT, Lin TP, Tang JT, Campbell M, Luo YL, Lu SY, Yang CP, Cheng TY, Chang CH, Liu TT, et al: HOTAIR is a REST-regulated lncRNA that promotes neuroendocrine differe ntiation in castration resistant prostate cancer. Cancer Lett. 433:43–52. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Lin YC, Chang YT, Campbell M, Lin TP, Pan CC, Lee HC, Shih JC and Chang PC: MAOA-a novel decision maker of apoptosis and autophagy in hormone refr actory neuroendocrine prostate cancer cells. Sci Rep. 7:463382017. View Article : Google Scholar : PubMed/NCBI

151 

Lee CF, Chen YA, Hernandez E, Pong RC, Ma S, Hofstad M, Kapur P, Zhau H, Chung LW, Lai CH, et al: The central role of Sphingosine kinase 1 in the development of neuroen docrine prostate cancer (NEPC): A new targeted therapy of NEPC. Clin Transl Med. 12:e6952022. View Article : Google Scholar : PubMed/NCBI

152 

O'Reilly D, Johnson P and Buchanan PJ: Hypoxia induced cancer stem cell enrichment promotes resistance to androgen deprivation therapy in prostate cancer. Steroids. 152:1084972019. View Article : Google Scholar : PubMed/NCBI

153 

Alumkal JJ, Sun D, Lu E, Beer TM, Thomas GV, Latour E, Aggarwal R, Cetnar J, Ryan CJ, Tabatabaei S, et al: Transcriptional profiling identifies an androgen receptor activity-low, stemness program associated with enzalutamide resistance. Proc Natl Acad SciUSA. 117:12315–12323. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Bishop JL, Thaper D and Zoubeidi A: The multifaceted roles of STAT3 signaling in the progression of prostate cancer. Cancers. 6:829–859. 2014. View Article : Google Scholar : PubMed/NCBI

155 

Albino D, Civenni G, Rossi S, Mitra A, Catapano CV and Carbone GM: The ETS factor ESE3/EHF represses IL-6 preventing STAT3 activation and expansion of the prostate cancer stem-like compartment. Oncotarget. 7:76756–76768. 2016. View Article : Google Scholar : PubMed/NCBI

156 

Song N, Cui K, Zeng L, Li M, Fan Y, Shi P, Wang Z, Su W and Wang H: Advance in the role of chemokines/chemokine receptors in carcinogenesis: Focus on pancreatic cancer. Eur J Pharmacol. 967:1763572024. View Article : Google Scholar : PubMed/NCBI

157 

Liu RY, Zeng Y, Lei Z, Wang L, Yang H, Liu Z, Zhao J and Zhang HT: JAK/STAT3 signaling is required for TGF-β-induced epithelial-mesenchymal transition in lung cancer cells. Int J Oncol. 44:1643–1651. 2014. View Article : Google Scholar : PubMed/NCBI

158 

Deng S, Wang C, Wang Y, Xu Y, Li X, Johnson NA, Mukherji A, Lo UG, Xu L, Gonzalez J, et al: Ectopic JAK-STAT activation enables the transition to a stem-like and multilineage state conferring AR-targeted therapy resistance. Nat Cancer. 3:1071–1087. 2022. View Article : Google Scholar : PubMed/NCBI

159 

Ware KE, Thomas BC, Olawuni PD, Sheth MU, Hawkey N, Yeshwanth M, Miller BC, Vietor KJ, Jolly MK, Kim SY, et al: A synthetic lethal screen for Snail-induced enzalutamide resistance identifies JAK/STAT signaling as a therapeutic vulnerability in prostate cancer. Front Mol Biosci. 10:11045052023. View Article : Google Scholar : PubMed/NCBI

160 

Chen L, Wang Y and Zhang B: Hypermethylation in the promoter region inhibits AJAP1 expression and activates the JAK/STAT pathway to promote prostate cancer cell migration and stem cell sphere formation. Pathol Res Pract. 241:1542242023. View Article : Google Scholar : PubMed/NCBI

161 

Spiotto MT and Chung TD: STAT3 mediates IL-6-induced neuroendocrine differentiation in prostate cancer cells. Prostate. 42:186–195. 2000. View Article : Google Scholar : PubMed/NCBI

162 

Smith ND, Schulze-Hoepfner FT, Veliceasa D, Filleur S, Shareef S, Huang L, Huang XM and Volpert OV: Pigment Epithelium-derived factor and interleukin-6 control prostate neuroendocrine differentiation via feed-forward mechanism. J Urol. 179:2427–2434. 2008. View Article : Google Scholar : PubMed/NCBI

163 

Dai J, Keller J, Zhang J, Lu Y, Yao Z and Keller ET: Bone morphogenetic protein-6 promotes osteoblastic prostate cancer bone metastases through a dual mechanism. Cancer Res. 65:8274–8285. 2005. View Article : Google Scholar : PubMed/NCBI

164 

Dubrovska A, Kim S, Salamone RJ, Walker JR, Maira SM, García-Echeverría C, Schultz PG and Reddy VA: The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA. 106:268–273. 2009. View Article : Google Scholar : PubMed/NCBI

165 

Zhang A, Lau NA, Wong A, Brown LG, Coleman IM, De Sarkar N, Li D, DeLucia DC, Labrecque MP, Nguyen HM, et al: Concurrent targeting of HDAC and PI3K to overcome phenotypic heterogeneity of castration-resistant and neuroendocrine prostate cancers. Cancer Res Commun. 3:2358–2374. 2023. View Article : Google Scholar : PubMed/NCBI

166 

Wu J, Cang S, Liu C, Ochiai W and Chiao JW: Development of human prostate cancer stem cells involves epigenomic alteration and PI3K/AKT pathway activation. Exp Hematol Oncol. 9:122020. View Article : Google Scholar : PubMed/NCBI

167 

Morell C, Bort A, Vara D, Ramos-Torres A, Rodríguez-Henche N and Díaz-Laviada I: The cannabinoid WIN 55,212-2 prevents neuroendocrine differentiation of LNCaP prostate cancer cells. Prostate Cancer Prostatic Dis. 19:248–257. 2016. View Article : Google Scholar : PubMed/NCBI

168 

Gonzalez DM and Medici D: Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 7:re82014. View Article : Google Scholar : PubMed/NCBI

169 

Yoo YA, Kang MH, Kim JS and Oh SC: Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Carcinogenesis. 29:480–490. 2008. View Article : Google Scholar : PubMed/NCBI

170 

Petrova R and Joyner AL: Roles for Hedgehog signaling in adult organ homeostasis and repair. Development. 141:3445–3457. 2014. View Article : Google Scholar : PubMed/NCBI

171 

Merchant AA and Matsui W: Targeting Hedgehog-a cancer stem cell pathway. Clin Cancer Res. 16:3130–3140. 2010. View Article : Google Scholar : PubMed/NCBI

172 

Chang HH, Chen BY, Wu CY, Tsao ZJ, Chen YY, Chang CP, Yang CR and Lin DP: Hedgehog overexpression leads to the formation of prostate cancer stem cells with metastatic property irrespective of androgen receptor expression in the mouse model. J Biomed Sci. 18:62011. View Article : Google Scholar : PubMed/NCBI

173 

Wang L, Li H, Li Z, Li M, Tang Q, Wu C and Lu Z: Smoothened loss is a characteristic of neuroendocrine prostate cancer. Prostate. 81:508–520. 2021. View Article : Google Scholar : PubMed/NCBI

174 

Kishore C and Zi X: Wnt Signaling and therapeutic resistance in castration-resistant prostate cancer. Curr Pharmacol Rep. 9:261–274. 2023. View Article : Google Scholar : PubMed/NCBI

175 

Sha J, Han Q, Chi C, Zhu Y, Pan J, Dong B, Huang Y, Xia W and Xue W: PRKAR2B promotes prostate cancer metastasis by activating Wnt/β-catenin and inducing epithelial-mesenchymal transition. J Cell Biochem. 119:7319–7327. 2018. View Article : Google Scholar : PubMed/NCBI

176 

Duchartre Y, Kim YM and Kahn M: The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol. 99:141–149. 2016. View Article : Google Scholar : PubMed/NCBI

177 

Bisson I and Prowse DM: WNT signaling regulates self-renewal and differentiation of prostate cancer cells with stem cell characteristics. Cell Res. 19:683–697. 2009. View Article : Google Scholar : PubMed/NCBI

178 

Pan KF, Lee WJ, Chou CC, Yang YC, Chang YC, Chien MH, Hsiao M and Hua KT: Direct interaction of β-catenin with nuclear ESM1 supports stemness of metastatic prostate cancer. EMBO J. 40:e1054502021. View Article : Google Scholar : PubMed/NCBI

179 

Manzar N, Khan UK, Goel A, Carskadon S, Gupta N, Palanisamy N and Ateeq B: An integrative proteomics approach identifies tyrosine kinase KIT as a therapeutic target for SPINK1-positive prostate cancer. iScience. 27:1087942024. View Article : Google Scholar : PubMed/NCBI

180 

Bland T, Wang J, Yin L, Pu T, Li J, Gao J, Lin TP, Gao AC and Wu BJ: WLS-Wnt signaling promotes neuroendocrine prostate cancer. iScience. 24:1019702021. View Article : Google Scholar : PubMed/NCBI

181 

Uysal-Onganer P, Kawano Y, Caro M, Walker MM, Diez S, Darrington RS, Waxman J and Kypta RM: Wnt-11 promotes neuroendocrine-like differentiation, survival and migration of prostate cancer cells. Mol Cancer. 9:552010. View Article : Google Scholar : PubMed/NCBI

182 

Liu RJ, Xu ZP, Huang X, Xu B and Chen M: Yin Yang 1 promotes the neuroendocrine differentiation of prostate cancer cells via the non-canonical WNT pathway (FYN/STAT3). Clin Transl Med. 13:e14222023. View Article : Google Scholar : PubMed/NCBI

183 

Wen YC, Liu YN, Yeh HL, Chen WH, Jiang KC, Lin SR, Huang J, Hsiao M and Chen WY: TCF7L1 regulates cytokine response and neuroendocrine differentiation of prostate cancer. Oncogenesis. 10:812021. View Article : Google Scholar : PubMed/NCBI

184 

Terry S, Maillé P, Baaddi H, Kheuang L, Soyeux P, Nicolaiew N, Ceraline J, Firlej V, Beltran H, Allory Y, et al: Cross modulation between the androgen receptor axis and protocadherin-PC in mediating neuroendocrine transdifferentiation and therapeutic resistance of prostate cancer. Neoplasia. 15:761–772. 2013. View Article : Google Scholar : PubMed/NCBI

185 

Shan J, Al-Muftah MA, Al-Kowari MK, Abuaqel SWJ, Al-Rumaihi K, Al-Bozom I, Li P and Chouchane L: Targeting Wnt/EZH2/microRNA-708 signaling pathway inhibits neuroendocrine differentiation in prostate cancer. Cell Death Discov. 5:1392019. View Article : Google Scholar : PubMed/NCBI

186 

Unno K, Chalmers ZR, Pamarthy S, Vatapalli R, Rodriguez Y, Lysy B, Mok H, Sagar V, Han H, Yoo YA, et al: Activated ALK cooperates with N-Myc via Wnt/β-Catenin signaling to induce neuroendocrine prostate cancer. Cancer Res. 81:2157–2170. 2021. View Article : Google Scholar : PubMed/NCBI

187 

Kar R, Jha NK, Jha SK, Sharma A, Dholpuria S, Asthana N, Chaurasiya K, Singh VK, Burgee S and Nand P: A ‘NOTCH’ Deeper into the Epithelial-To-Mesenchymal Transition (EMT) program in breast cancer. Genes (Basel). 10:9612019. View Article : Google Scholar : PubMed/NCBI

188 

Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, et al: Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 18:99–115. 2004. View Article : Google Scholar : PubMed/NCBI

189 

Uribe-Etxebarria V, Pineda JR, García-Gallastegi P, Agliano A, Unda F and Ibarretxe G: Notch and wnt signaling modulation to enhance DPSC stemness and therapeutic potential. Int J Mol Sci. 24:73892023. View Article : Google Scholar : PubMed/NCBI

190 

Puca L, Gavyert K, Sailer V, Conteduca V, Dardenne E, Sigouros M, Isse K, Kearney M, Vosoughi A, Fernandez L, et al: Delta-like protein 3 expression and therapeutic targeting in neuroendo crine prostate cancer. Sci Transl Med. 11:eaav08912019. View Article : Google Scholar : PubMed/NCBI

191 

Danza G, Di Serio C, Rosati F, Lonetto G, Sturli N, Kacer D, Pennella A, Ventimiglia G, Barucci R, Piscazzi A, et al: Notch signaling modulates hypoxia-induced neuroendocrine differentiati on of human prostate cancer cells. Mol Cancer Res. 10:230–238. 2012. View Article : Google Scholar : PubMed/NCBI

192 

Menssouri N, Poiraudeau L, Helissey C, Bigot L, Sabio J, Ibrahim T, Pobel C, Nicotra C, Ngo-Camus M, Lacroix L, et al: Genomic profiling of metastatic castration-resistant prostate cancer s amples resistant to androgen-receptor pathway inhibitors. Clin Cancer Res. 29:4504–4517. 2023. View Article : Google Scholar : PubMed/NCBI

193 

Docherty NG, O'Sullivan OE, Healy DA, Murphy M, O'Neill AJ, Fitzpatrick JM and Watson RW: TGF-beta1-induced EMT can occur independently of its proapoptotic effects and is aided by EGF receptor activation. Am J Physiol Renal Physiol. 290:F1202–F1212. 2006. View Article : Google Scholar : PubMed/NCBI

194 

Yu B, Su J, Shi Q, Liu Q, Ma J, Ru G, Zhang L, Zhang J, Hu X and Tang J: KMT5A-methylated SNIP1 promotes triple-negative breast cancer metastasis by activating YAP signaling. Nat Commun. 13:21922022. View Article : Google Scholar : PubMed/NCBI

195 

Jiao C, Meng T, Zhou C, Wang X, Wang P, Lu M, Tan X, Wei Q, Ge X and Jin J: TGF-β signaling regulates SPOP expression and promotes prostate cancer cell stemness. Aging (Albany NY). 12:7747–7760. 2020. View Article : Google Scholar : PubMed/NCBI

196 

Natani S, Sruthi KK, Asha SM, Khilar P, Lakshmi PSV and Ummanni R: Activation of TGF-β-SMAD2 signaling by IL-6 drives neuroendocrine differentiation of prostate cancer through p38MAPK. Cell Signal. 91:1102402022. View Article : Google Scholar : PubMed/NCBI

197 

Wen S, Hou Y, Fu L, Xi L, Yang D, Zhao M, Qin Y, Sun K, Teng Y and Liu M: Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling. Cancer Lett. 442:320–332. 2019. View Article : Google Scholar : PubMed/NCBI

198 

Tang T, Wang LX, Yang ML and Zhang RM: lncRNA TPTEP1 inhibits stemness and radioresistance of glioma through miR-106a-5p-mediated P38 MAPK signaling. Mol Med Rep. 22:4857–4867. 2020. View Article : Google Scholar : PubMed/NCBI

199 

Du Y, Long Q, Zhang L, Shi Y, Liu X, Li X, Guan B, Tian Y, Wang X, Li L and He D: Curcumin inhibits cancer-associated fibroblast-driven prostate cancer invasion through MAOA/mTOR/HIF-1α signaling. Int J Oncol. 47:2064–2072. 2015. View Article : Google Scholar : PubMed/NCBI

200 

Han H, Li H, Ma Y, Zhao Z, An Q, Zhao J and Shi C: Monoamine oxidase A (MAOA): A promising target for prostate cancer therapy. Cancer Lett. 563:2161882023. View Article : Google Scholar : PubMed/NCBI

201 

Weng CC, Ding PY, Liu YH, Hawse JR, Subramaniam M, Wu CC, Lin YC, Chen CY, Hung WC and Cheng KH: Mutant Kras-induced upregulation of CD24 enhances prostate cancer stemness and bone metastasis. Oncogene. 38:2005–2019. 2019. View Article : Google Scholar : PubMed/NCBI

202 

De Luca P, Moiola CP, Zalazar F, Gardner K, Vazquez ES and De Siervi A: BRCA1 and p53 regulate critical prostate cancer pathways. Prostate Cancer Prostatic Dis. 16:233–238. 2013. View Article : Google Scholar : PubMed/NCBI

203 

Wang Y, Zhang YX, Kong CZ, Zhang Z and Zhu YY: Loss of P53 facilitates invasion and metastasis of prostate cancer cells. Mol Cell Biochem. 384:121–127. 2013. View Article : Google Scholar : PubMed/NCBI

204 

Ren D, Wang M, Guo W, Zhao X, Tu X, Huang S, Zou X and Peng X: Wild-type p53 suppresses the epithelial-mesenchymal transition and stemness in PC-3 prostate cancer cells by modulating miR-145. Int J Oncol. 42:1473–1481. 2013. View Article : Google Scholar : PubMed/NCBI

205 

Kogan-Sakin I, Tabach Y, Buganim Y, Molchadsky A, Solomon H, Madar S, Kamer I, Stambolsky P, Shelly A, Goldfinger N, et al: Mutant p53(R175H) upregulates Twist1 expression and promotes epithelial-mesenchymal transition in immortalized prostate cells. Cell Death Differ. 18:271–281. 2011. View Article : Google Scholar : PubMed/NCBI

206 

Kumaraswamy A, Duan Z, Flores D, Zhang C, Sehrawat A, Hu YM, Swaim OA, Rodansky E, Storck WK, Kuleape JA, et al: LSD1 promotes prostate cancer reprogramming by repressing TP53 signaling independently of its demethylase function. JCI Insight. 8:e1674402023. View Article : Google Scholar : PubMed/NCBI

207 

Chaves LP, Melo CM, Saggioro FP, Reis RBD and Squire JA: Epithelial-mesenchymal transition signaling and prostate cancer stem cells: Emerging biomarkers and opportunities for precision therapeutics. Genes (Basel). 12:19002021. View Article : Google Scholar : PubMed/NCBI

208 

Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Bahonar A, Nakhaee Z, Entezari M, Beig Goharrizi MAS, Salimimoghadam S, Ren J, et al: New emerging targets in osteosarcoma therapy: PTEN and PI3K/Akt crosstalk in carcinogenesis. Pathol Res Pract. 251:1549022023. View Article : Google Scholar : PubMed/NCBI

209 

Mei W, Lin X, Kapoor A, Gu Y, Zhao K and Tang D: The contributions of prostate cancer stem cells in prostate cancer initiation and metastasis. Cancers (Basel). 11:4342019. View Article : Google Scholar : PubMed/NCBI

210 

Kaushik G, Seshacharyulu P, Rauth S, Nallasamy P, Rachagani S, Nimmakayala RK, Vengoji R, Mallya K, Chirravuri-Venkata R, Singh AB, et al: Selective inhibition of stemness through EGFR/FOXA2/SOX9 axis reduces pancreatic cancer metastasis. Oncogene. 40:848–862. 2021. View Article : Google Scholar : PubMed/NCBI

211 

Liu Q, Pang J, Wang LA, Huang Z, Xu J, Yang X, Xie Q, Huang Y, Tang T, Tong D, et al: Histone demethylase PHF8 drives neuroendocrine prostate cancer progres sion by epigenetically upregulating FOXA2. J Pathol. 253:106–118. 253 View Article : Google Scholar : PubMed/NCBI

212 

Qi J, Pellecchia M and Ronai ZeA: The Siah2-HIF-FoxA2 axis in prostate cancer-new markers and therapeu tic opportunities. Oncotarget. 1:379–385. 2010. View Article : Google Scholar : PubMed/NCBI

213 

Qi J, Nakayama K, Cardiff RD, Borowsky AD, Kaul K, Williams R, Krajewski S, Mercola D, Carpenter PM, Bowtell D, et al: Siah2-dependent concerted activity of HIF and FoxA2 regulates formatio n of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell. 18:23–38. 2010. View Article : Google Scholar : PubMed/NCBI

214 

Masone MC: FOXA2-KIT-driven lineage plasticity in NEPC. Nat Rev Urol. 20:82023. View Article : Google Scholar

215 

Singh N, Padi SKR, Bearss JJ, Pandey R, Okumura K, Beltran H, Song JH, Kraft AS and Olive V: PIM protein kinases regulate the level of the long noncoding RNA H19 t o control stem cell gene transcription and modulate tumor growth. Mol Oncol. 14:974–990. 2020. View Article : Google Scholar : PubMed/NCBI

216 

Lee AR, Gan Y, Tang Y and Dong X: A novel mechanism of SRRM4 in promoting neuroendocrine prostate cancer development via a pluripotency gene network. EBioMedicine. 35:167–177. 2018. View Article : Google Scholar : PubMed/NCBI

217 

Fan L, Gong Y, He Y, Gao WQ, Dong X, Dong B, Zhu HH and Xue W: TRIM59 is suppressed by androgen receptor and acts to promote lineage plasticity and treatment-induced neuroendocrine differentiation in pro state cancer. Oncogene. 42:559–571. 2023. View Article : Google Scholar : PubMed/NCBI

218 

Monga J, Adrianto I, Rogers C, Gadgeel S, Chitale D, Alumkal JJ, Beltran H, Zoubeidi A and Ghosh J: Tribbles 2 pseudokinase confers enzalutamide resistance in prostate ca ncer by promoting lineage plasticity. J Biol Chem. 298:1015562022. View Article : Google Scholar : PubMed/NCBI

219 

Kim S, Thaper D, Bidnur S, Toren P, Akamatsu S, Bishop JL, Colins C, Vahid S and Zoubeidi A: PEG10 is associated with treatment-induced neuroendocrine prostate can cer. J Mol Endocrinol. 63:39–49. 2019. View Article : Google Scholar : PubMed/NCBI

220 

Yoshie H, Sedukhina AS, Minagawa K, Oda K, Ohnuma S, Yanagisawa N, Maeda I, Takagi M, Kudo H, Nakazawa R, et al: A bioinformatics-to-clinic sequential approach to analysis of prostate cancer biomarkers using TCGA datasets and clinical samples: A new met hod for precision oncology? Oncotarget. 8:99601–99611. 2017. View Article : Google Scholar : PubMed/NCBI

221 

Choi WW, Boland JL and Lin J: ONECUT2 as a key mediator of androgen receptor-independent cell growth and neuroendocrine differentiation in castration-resistant prostate c ancer. Cancer Drug Resist. 5:165–170. 2022.PubMed/NCBI

222 

Tenjin Y, Kudoh S, Kubota S, Yamada T, Matsuo A, Sato Y, Ichimura T, Kohrogi H, Sashida G, Sakagami T and Ito T: Ascl1-induced Wnt11 regulates neuroendocrine differentiation, cell proliferation, and E-cadherin expression in small-cell lung cancer and Wnt11 regulates small-cell lung cancer biology. Lab Invest. 99:1622–1635. 2019. View Article : Google Scholar : PubMed/NCBI

223 

DeLucia DC, Cardillo TM, Ang L, Labrecque MP, Zhang A, Hopkins JE, De Sarkar N, Coleman I, da Costa RMG, Corey E, et al: Regulation of CEACAM5 and Therapeutic Efficacy of an Anti-CEACAM5-SN38 Antibody-drug conjugate in neuroendocrine prostate cancer. Clin Cancer Res. 27:759–774. 2021. View Article : Google Scholar : PubMed/NCBI

224 

Meder L, König K, Ozretić L, Schultheis AM, Ueckeroth F, Ade CP, Albus K, Boehm D, Rommerscheidt-Fuss U, Florin A, et al: NOTCH, ASCL1, p53 and RB alterations define an alternative pathway dri ving neuroendocrine and small cell lung carcinomas. Int J Cancer. 138:927–938. 2016. View Article : Google Scholar : PubMed/NCBI

225 

Tang Q, Chen J, Di Z, Yuan W, Zhou Z, Liu Z, Han S, Liu Y, Ying G, Shu X, et al: TM4SF1 promotes EMT and cancer stemness via the Wnt/β-catenin/SOX2 pathway in colorectal cancer. J Exp Clin Cancer Res. 39:2322020. View Article : Google Scholar : PubMed/NCBI

226 

Zhang Z, Wang X, Kim M, He D, Wang C, Fong KW and Liu X: Downregulation of EZH2 inhibits epithelial-mesenchymal transition in enzalutamide-resistant prostate cancer. Prostate. 83:1458–1469. 2023. View Article : Google Scholar : PubMed/NCBI

227 

Mirzaei S, Gholami MH, Hushmandi K, Hashemi F, Zabolian A, Canadas I, Zarrabi A, Nabavi N, Aref AR, Crea F, et al: The long and short non-coding RNAs modulating EZH2 signaling in cancer. J Hematol Oncol. 15:182022. View Article : Google Scholar : PubMed/NCBI

228 

Zhang W, Bado IL, Hu J, Wan YW, Wu L, Wang H, Gao Y, Jeong HH, Xu Z, Hao X, et al: The bone microenvironment invigorates metastatic seeds for further dissemination. Cell. 184:2471–2486.e20. 2021. View Article : Google Scholar : PubMed/NCBI

229 

Kufe D: Dependence on MUC1-C in progression of neuroendocrine prostate cancer. Int J Mol Sci. 24:37192023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2025
Volume 54 Issue 3

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
Li Y, Su S, Luo Y, Wei C, He J, Song L, Han K, Wang J, Gan X, Wang D, Wang D, et al: Widespread activation and critical role of EMT and stemness in the neuroendocrine differentiation of prostate cancer (Review). Oncol Rep 54: 109, 2025.
APA
Li, Y., Su, S., Luo, Y., Wei, C., He, J., Song, L. ... Wang, D. (2025). Widespread activation and critical role of EMT and stemness in the neuroendocrine differentiation of prostate cancer (Review). Oncology Reports, 54, 109. https://doi.org/10.3892/or.2025.8942
MLA
Li, Y., Su, S., Luo, Y., Wei, C., He, J., Song, L., Han, K., Wang, J., Gan, X., Wang, D."Widespread activation and critical role of EMT and stemness in the neuroendocrine differentiation of prostate cancer (Review)". Oncology Reports 54.3 (2025): 109.
Chicago
Li, Y., Su, S., Luo, Y., Wei, C., He, J., Song, L., Han, K., Wang, J., Gan, X., Wang, D."Widespread activation and critical role of EMT and stemness in the neuroendocrine differentiation of prostate cancer (Review)". Oncology Reports 54, no. 3 (2025): 109. https://doi.org/10.3892/or.2025.8942