3.1. Targeting genomic alterations
NEPC cell lines show enhanced sensitivity to AURKA inhibitors [
27]. The ATP competitive pan-Aurora kinase inhibitor Danusertib (PHA-739358) has demonstrated significant effects both in vitro and in vivo, inhibiting NEPC cell lines proliferation and reducing tumor volume and NE activity in xenograft models [
27,
107]. However, a randomized phase II study showed minimal efficacy of Danusertib monotherapy in non-selected patients with mCRPC after docetaxel failure [
108], possibly as ATP competitive inhibitors can leave the AURKA-MYCN complex unflawed [
93,
109]. On the contrary, the AURKA inhibitors Alisertib (MLN8237) and CD532 can effectively disrupt this complex, resulting in MYCN destabilization and cytotoxic activity in vitro [
28,
34,
110]. Alisertib has been evaluated in a phase II clinical trial for NEPC treatment. While the trial did not achieve its primary endpoint of progression-free survival (PFS) in a biomarker-unselected population, two patients showed an exceptional response with complete eradication of liver metastases [
111]. Ongoing pre-clinical studies are investigating the efficacy of other small molecules targeting aurora kinases, such as VX680, that has demonstrated potent anti-tumor activity in PC cell lines [
112]. Additionally, Ton et al. have developed a molecule called 7082 that effectively targets both MYCN and AURKA, suppressing proliferation of PC and NEPC cell lines [
113]. The small molecule VPC-70619 specifically targets MYCN and has shown strong anti-proliferative activity against cell lines expressing MYCN, including NEPC cell lines. Pharmacokinetic studies have revealed that VPC-70619 exhibits high bioavailability through intraperitoneal and oral administration, making it a potentially valuable compound for treatment of lethal NEPC [
114].
Pharmacological targeting of factors upstream of MYCN and AURKA has been proposed as complementary therapeutic strategies for NEPC. NK1R (Tachykinin Receptor 1) activates the AURKA/MYCN signaling pathway through PKCα and its knockdown results in the reduction of tumor burden and suppression of NE features in vivo. Aprepitant, an FDA-approved selective NK1R antagonist, exerts anti-proliferative effects in NE-like and NEPC cells, and the PKC inhibitor GF109203X induces cell cycle G2/M arrest [
110].
3.2. Targeting epigenetic factors
MYCN redirects EZH2 activity and NEPC cells are sensitive to EZH2 inhibition [
34]. A variety of EZH2 inhibitors are currently being tested in a wide range of cancers [
115]. EZH2 inhibitors tested in pre-clinical studies include GSK343, GSK503, GSE126, DZNEP and EPZ6438 (Tazemetostat) [
24,
26,
34,
116]. Treatment with EZH2 inhibitors reduces MYCN-EZH2 interaction, results in downregulation of NE genes and can curb cell viability of NEPC cells [
24,
34,
116]. Moreover, some of these inhibitors have demonstrated the ability to enhance AR expression and increase sensitivity to ADT in vitro and in vivo [
26]. Based on these promising results, several clinical trials are currently investigating EZH2 inhibitor therapy alone or in combination with potent AR inhibitors for mCRPC (NCT03480646 and NCT04179864). Additional pre-clinical results demonstrate that GSK126 exhibits increased toxicity in NEPC cells when combined with the chemotherapy agent docetaxel [
76], suggesting a potential novel treatment regimen to be evaluated.
In NEPC arising after ARSI therapy, EZH2 activity has been shown to be enhanced by the induction of the PKA/CREB (cAMP-response element binding protein) pathway. Of note, treatment with the β-adrenergic antagonist and PKA/CREB inhibitor Propranolol has been found to significantly reduce tumor growth, NE differentiation and angiogenesis in vivo [
116].
PRC2 trimethylates H3K27, a histone mark recognized by the CBX subunit of the canonical PRC1 complex. Among the different CBX paralogs, CBX2 has been identified as a key player in NEPC progression [
33,
80]. A CBX2-specific chromodomain inhibitor named SW2_152F has been developed to target this pathway with promising results as it effectively blocks NE fate and promotes PC cell death [
80].
The link between EZH2 and DNMTs has sparked interest in DNMTs as potential drug targets in NEPC. The hypomethylation agent decitabine reverts basal and NE markers and inhibits NEPC tumor growth in mice [
89]. Its analog, azacytidine, has also been shown to partially re-sensitize ARSI-resistant NE-like PC cell lines [
117]. Decitabine and azacytidine are already FDA-approved for the treatment of myelodysplastic syndromes and could be repurposed for NEPC treatment, although the latter showed weak anti-tumor activity in a phase II clinical trial in patients with mCRPC [
118]. Currently, there are two trials in progress evaluating the combination of decitabine and guadecitabine (SGI-110) with enzalutamide and the immunotherapeutic drug pembrolizumab, respectively, in mCRPC patients (NCT05037500, NCT02998567).
Other drugs directed against epigenetic modulators have been investigated for targeting plasticity and the NEPC phenotype. NSD2 depletion using short hairpin RNAs (shRNAs) inhibits PC tumorigenicity both in vitro and in vivo [
36]. Similar effects were obtained after treatment of DU145 xenografts with the small molecule inhibitor of NSD2 (MCTP-39) [
36]. The epigenetic regulator LSD1, an emerging target for small cell lung cancer (SCLC) [
119], has also been explored in NEPC. The allosteric inhibitors of LSD1, SP-2509 and SP-2577, are molecules that potentially could be used for NEPC treatment, as they effectively suppress NE cells growth and show good tolerability in in vivo models [
39,
120]. The reversible LSD1 inhibitor CC-90011 has been evaluated in a phase I clinical trial of advanced malignancies, including NEPC, and has shown an acceptable tolerability profile and promising overall clinical activity [
121]. Currently, a clinical trial conducted exclusively in patients with mCRPC is trying to assess whether CC-90011 can induce AR expression and, consequently, re-sensitize tumors to anti-hormonal therapy (NCT04628988).
Depletion of the chromatin modulator DEK with siRNAs (small interfering RNA) suppresses cell growth, migration and invasion of PC3 cells, an AR-negative adenocarcinoma cell line sometimes used as an NEPC model [
44]. DEK-targeted aptamers (DTAs) have been studied in the context of inflammatory arthritis [
122] and could potentially be explored as a novel therapeutic approach for patients with NEPC.
Pre-clinical studies have revealed that BET inhibitors such as JQ1, ZEN-3694 and OTX-15 block the NE program and suppress NEPC growth by inhibiting the BRD4-E2F1 program [
47] and MYCN-driven NE differentiation [
123]. In a phase Ib/IIa clinical trial ZEN-3694 in combination with enzalutamide has demonstrated acceptable tolerability and potential efficacy in patients with mCRPC, providing clinical evidence that BET inhibition may be able to abrogate resistance mechanisms and re-sensitize patients to AR-signaling inhibitors [
124]. Other phase II clinical trials evaluating ZEN-3694 are currently active for mCRPC (NCT04471974, NCT04986423).
3.3. Targeting transcription factors
Although TP53 and RB1 mutations are almost universal in NEPC, they are not readily targetable. As an alternative strategy, there is growing interest in targeting common downstream effectors of both tumor suppressor genes. One such effector is PEG10, which has gained significant attention in recent studies. Targeting PEG10 using siRNAs or shRNAs effectively reduce the proliferation rate and expression of NE markers both in vitro and in vivo [
55]. PEG10 possesses a unique ribosomal frameshift sequence and a protease domain similar to the HIV (human immunodeficiency virus), which makes it a suitable candidate for drug targeting [
92]. Another targetable transcription factor is ONECUT2, which has been shown to be a survival factor in mCRPC. Inhibition of ONECUT2 can be achieved using a small molecule named CSRM617 that reduces tumor size/weight and metastasis in xenograft models [
62]. Additionally, the synergistic interaction between ONECUT2 and hypoxia has led to the investigation of an alternative therapeutic strategy involving the use of the hypoxia-activated prodrug TH-302, which reduces NEPC tumor growth in both xenograft and PDX (patient-derived xenografts) models [
61].
Recent efforts have focused on the identification of inhibitors against the driver of lineage plasticity, BRN2. Deletion or stable knockdown of BRN2 prevents NE differentiation, thus reducing invasiveness and tumor proliferation in both enzalutamide- and castration-resistant PC [
63]. While the first-in-field BRN2 inhibitor is developed, one approach to inhibit BRN2 signaling is by targeting its upstream regulator MUC1-C. Silencing MUC1-C leads to the downregulation of BRN2, decreasing self-renewal capacity and tumorigenicity of PC cells [
64]. Cell-penetrating peptides (CPPs), such as GO-203, block MUC1-C homodimerization and nuclear localization. GO-203 has already undergone evaluation in early-phase clinical trials for solid tumors (NCT01279603). However, its short half-life presents a challenge in the clinical setting, and new strategies, such as polymeric nanoparticle encapsulation (GO-203/NPs) are being explored [
125]. In addition to CPPs, other approaches targeting the extracellular domain of MUC1-C have been investigated. Antibody-based approaches, including antibody-drug conjugates (ADCs) [
126] and chimeric antigen receptor (CAR) T cells, have demonstrated potential in drugging the extracellular domain of MUC1-C. CAR-T cells targeting MUC1-C-expressing cancers are already undergoing phase I evaluation (NCT05239143), although no PC patients are included in this cohort.
Other strategies have been directed towards targeting FOX transcription factors. Paranjape et al. identified a critical nexus between p38MAPK signaling and FOXC2 for NEPC development [
57]. This study has demonstrated that targeting FOXC2 using the p38 MAPK inhibitor, SB203580, can restore the epithelial phenotype and increase sensitivity to AR inhibition. Consequently, its combination with enzalutamide results in a substantial reduction of tumor growth in vivo [
57]. FOXA2 knockdown induces the reversal of adeno-to-NE lineage transition [
55]. As FOXA2 has been an elusive drug target, alternative strategies are being considered, such as inhibition of the SIAH2/HIF/FOXA2 axis. The FDA-approved drug menadione (Vitamin K3) is an inhibitor of SIAH2 that, in combination with ADT, delays occurrence of CRPC [
127]. Another novel SIAH2 inhibitor, RLS-24, has demonstrated ability to reduce PC cell viability [
128]. These inhibitors are particularly relevant in the context of NEPC, as SIAH2 depletion with shRNAs results in a marked suppression of NEPC tumors [
60].
3.4. Targeting pathways and biological processes
FOXA2 promotes NEPC by direct activation of KIT expression. Targeting the KIT pathway with tyrosine kinase inhibitors (TKIs) such as imatinib, sorafenib, sunitinib and cabozantinib suppresses mouse and human NEPC tumor growth [
58,
129]. However, it is important to note that the TKI dovitinib has demonstrated unexpected effects, inducing NE differentiation instead of repressing it [
130]. Various TKIs have been evaluated in clinical trials yielding different efficacies. Notably, cabozantinib showed promising results in phase II trials [
131,
132] but did not significantly improve overall survival in phase III trials [
133]. Cabozantinib is currently being assessed as monotherapy in a biomarker-selected subgroup of CRPC (NCT04631744) as well as in combination with the checkpoint inhibitor nivolumab (NCT05502315) and atezolizumab (NCT04446117), among others clinical trials.
As a TKI, cabozantinib has the ability to block the RET kinase, an essential factor in NEPC development, as evidenced by the strong growth suppression of NEPC cell lines upon RET knockdown. Importantly, the molecules AD80, LOXO-292, and BLU-667 exhibit a higher degree of selectivity in inhibiting the RET pathway compared to cabozantinib, and effectively induce cell death in NEPC 3D cultures and xenograft models, opening new possibilities for targeted therapies in NEPC treatment [
88].
As above mentioned, SRC family kinases are also potential therapeutic targets in NEPC. Dasatinib (BMS-354825) is a Src/ABL TKI that has shown pre-clinical activity in PC cells [
134]. Dasatinib demonstrated biological effects only in chemotherapy-naïve mCRPC patients [
135,
136], and the subsequent phase III study that combined dasatinib with docetaxel in mCRPC patients did not result in an improvement in overall survival compared to chemotherapy alone [
137]. c-SRC also activates the MEK/ERK cascade to drive NE transdifferentiation from prostate adenocarcinoma [
138]. The MEK1/2 inhibitor trametinib (TMT212) and the ERK inhibitor SCH772984 have shown anti-proliferative effects in human NE cell lines [
139]. Trametinib is currently being evaluated in a phase 2 trial for patients with mCRPC (NCT02881242). In addition, SPHK1 (Sphingosine Kinase 1) plays an autocrine role to promote NEPC transdifferentiation by activating ERK, eventually leading to REST proteasomal degradation. FDA-approved SPHK1-specific inhibitors, such as FTY720 or SKI-II, have demonstrated ability to inhibit NEPC tumors growth and block REST protein degradation, resulting in reduced expression of NE markers in PDX models [
140].
PI3K/AKT inhibitors have also been explored to treat NE tumors. Pan-PI3K inhibitors such as buparlisib (BKM-120) and dactolisib (BEZ235), which also inhibits mTOR, can effectively reduce cell viability in PC cells, particularly in those overexpressing MYCN. The pan-AKT inhibitors ipatasertib and MK2206, and the mTOR inhibitor RAD001 have also shown favorable pre-clinical results in mCRPC [
34]. Several of these compounds blocking the PI3K/AKT pathway have been assessed in clinical trials. While some PI3K inhibitors like buparlisib, dactolisib, or PX-866 did not show significant activity in patients with mCRPC [
141,
142,
143], AKT inhibitors appear to be more promising: MK2206 has demonstrated partial responses in two NEPC patients during a phase I trial [
144] and ipatasertib combined with abiraterone have shown trends toward improved radiographical progression-free survival (rPFS) in mCRPC patients, particularly in cases with PTEN-loss [
145,
146]. Other trial combining ipatasertib with chemotherapy and immunotherapy is ongoing for mCRPC (NCT03072238). Of note, two studies have provided insights into the effects of PI3K/AKT-targeted therapies, demonstrating that the PI3K inhibitor LY294002 can induce differentiation towards NEPC [
147,
148]. This finding highlights a potential risk associated with the AR and PI3K/AKT co-targeting strategy.
The Wnt pathway can be blocked with the small molecule inhibitor LGK974, which is undergoing a phase I clinical trial in a wide range of solid tumors (NCT01351103). Bland et al. demonstrated that LGK974 treatment effectively inhibits NEPC tumor growth and reduces the expression of the NE marker CD56 both in vitro and in vivo [
85]. Other molecules tested in NEPC cells include the Wnt inhibitor ICG-001 and the β-catenin inhibitor XAV-939 [
86]. Notably, ICG-001 showed an additive effect in combination with the ALK inhibitor alectinib, leading to the suppression of NEPC proliferation in vitro and the inhibition of tumor growth and metastasis in vivo [
86].
Given the high similarities between NEPC and SCLC, DLL3-targeted therapies employed in SCLC are also being studied in the context of NEPC [
149]. DLL3+ NEPC xenografts have been shown to be sensitive to rovalpituzumab tesirine (SC16LD6.5), a DLL3-targeted ADC [
83]. In a phase I basket trial, a patient with DLL3-expressing NEPC experienced a significant reduction of nodal metastases upon treatment with this drug (NCT02709889). Ongoing studies are investigating other antibodies targeting DLL3 in NEPC, including the bispecific antibodies tarlatamab (NCT04702737) and PT217 (NCT05652686).
Among the EMT/NEPC-associated factors, ZBTB46 lacks specific inhibitors directly targeting its activity. Thus, efforts are being made to target downstream effectors of ZBTB46 such as LIF (Leukemia Inhibitory Factor), PTGS1 (Prostaglandin G/H synthase 1) and NGF (Nerve Growth Factor)/CHRM4 (Cholinergic Receptor Muscarinic 4) [
150,
151,
152]. The LIF inhibitor EC330 [
150], the PTGS1 inhibitor NS-398 [
151], the NGF inhibitor RO08-2750 [
152] and the CHRM4 inhibitor ceritinib [
153] have shown efficacy suppressing tumor growth and NE differentiation in PC. Knockdown of SNAI1 can block NE differentiation [
94] and its inhibition can be achieved by the novel proteasome inhibitor NPI-0052 (salinosporamide A) [
154,
155]. MLN4924 (pevonedistat), a small molecule currently undergoing phase II clinical trials for cancer, inhibits SNAI2 [
156] and when combined with ADT or ARSIs significantly enhances growth suppression of PC [
157]. Importantly, it has been shown that MLN4924 suppresses SOX2 expression [
158], a particularly relevant target in the context of NEPC.
Another novel therapeutic strategy for NEPC consists of targeting the MYCN-PARP-DDR pathway [
107] The PARP1 inhibitors talazoparib and olaparib can reverse the NE phenotype induced by TROP2 in PC cells and decrease tumor growth in TROP2-expressing NEPC xenografts [
92]. The possibility of co-targeting AURKA and PARP has also been studied. Inhibition of AURKA with PHA739358 and olaparib successfully suppressed growth in pre-clinical studies [
107]. Additionally, PARP1 inhibitors have been tested together with CDK4/6 inhibitors, which suppress E2F1 signaling frequently found activated in NEPC. The combination of olaparib and the FDA approved CDK4/6 inhibitors palbociblib or abemaciclib results in suppression of NE markers and tumor growth [
159]. Similar results have been observed combining olaparib and dinaciclib, a CDK2/5 inhibitor [
160].
Regarding the NRP1/PKC pathway, inhibition of NRP1 protein expression or suppression of PKC activation leads to the inhibition of NE differentiation and prevents tumor progression towards castration resistance. Enzastaurin, a potent pan-PKC inhibitor, can reduce the expression of NE markers in LNCaP-NE cells and enhances the cytotoxic effects of docetaxel in NEPC cells in in vitro and in vivo models [
90].