1,521
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PROTAC technology for prostate cancer treatment

      Published
      review-article

            Abstract

            Prostate cancer (PrCa) is the most prevalent urogenital cancer affecting men. PrCa is marked by uncontrolled cellular growth that leads to abnormal enlargement of the prostate gland. The metastatic spread of PrCa is the primary cause of mortality, causing cancer cell dissemination to distant sites, such as bones, the pelvis, and various visceral organs. Key contributors to PrCa progression include genetic mutations, elevated androgen receptor expression, gene amplification, and the rise of androgen receptor splice variants. Although androgen deprivation therapy remains the mainstay for early-stage PrCa treatment, efficacy is temporary because many cases advance to castration-resistant PrCa (CRPC), presenting a significant therapeutic hurdle. This review explores key biomarkers for PrCa and the latest therapeutic strategies for CRPC with a particular focus on the innovative proteolysis-targeting chimera (PROTAC) technology. This approach offers a novel means of degrading target proteins and we discuss how PROTAC holds potential as effective strategies to combat resistance mechanisms in CRPC.

            Main article text

            1. INTRODUCTION

            Prostate cancer (PrCa) is characterized by uncontrolled cellular proliferation that results in abnormal growth of the prostate gland [1]. PrCa is the most common cancer affecting the male urinary and reproductive systems [1]. Metastatic PrCa cell spread to distant sites, including bones, pelvis, lumbar vertebrae, bladder, rectum, and even the brain, are the primary driver of PrCa-related mortality [2]. Notably, nearly 48% of cancer cases in men are attributed to malignancies (prostate, lung, bronchus, and colorectal cancers) with PrCa presenting approximately 27% of all diagnoses. According to the NIH, 299,010 new PrCa cases (14.9% of all new cancer cases) and 35,250 deaths occurred in the United States in 2024 [3].

            Several factors contribute to PrCa progression, including genetic alterations, elevated androgen receptor (AR) expression, AR gene amplification, upregulation of cytochrome P450 17A1 (CYP17A1), and the development of AR variants [4]. Among these factors, AR splice variants (AR-Vs), particularly AR-V7 and AR-V9, lack the ligand-binding domain (LBD), rendering AR-Vs resistant to therapies targeting this domain, such as enzalutamide and abiraterone. These AR-Vs remain active in promoting AR signaling and contribute to tumor growth and survival, even in the presence of androgen deprivation or AR-targeted therapies [5, 6]. Notably, the standard treatment for localized PrCa involves androgen deprivation therapy (ADT), which is achieved through surgical and medical castration. Although ADT can induce remission for approximately 2–3 years, PrCa often progresses to castration-resistant PrCa (CRPC), a stage that presents challenges due to limited prognosis and significant therapeutic hurdles [7].

            This review provides an overview of the key PrCa biomarkers and the latest therapeutic strategies for CRPC. We place particular emphasis on the emerging proteolysis-targeting chimera (PROTAC) approach, which has garnered considerable attention for the potential to target previously “undruggable” proteins, thereby offering a promising avenue for advancing CRPC treatment [8]. PROTACs function as bispecific small molecules that facilitate proximity between target proteins of interest (POIs) and E3 ligases, thereby promoting the catalytic ubiquitination and subsequent target protein degradation. This review will delve into the various PROTACs that have been developed for PrCa, highlighting the mechanisms of actions and potential clinical applications. Ultimately, we hope to shed light on how these innovative strategies offer promising therapeutic avenues for CRPC by targeting diverse resistance mechanism.

            2. PRCA BIOMARKERS

            2.1 AR

            Without its ligand, AR is mainly located in the cytosol and forms complexes with heat shock proteins (HSPs) [911], cytoskeletal elements [12], and other chaperone proteins [10, 13]. This interaction with HSP not only stabilizes the receptor, but also induces conformational changes that prepare the AR for optimal binding when a ligand becomes available [14, 15] ( Figure 1 ).

            Next follows the figure caption
            Figure 1 |

            The key biomarkers and the related signaling pathways in prostate cancer (PrCa).

            Androgen stimulates androgen receptor (AR) signalling, resulting in the induction and secretion of prostate-specific antigen (PSMA), while illustrating the enzymatic function of prostate-specific membrane antigen PSMA and its role in folate metabolism and mGluR signaling pathways. AR, androgen receptor; DHT, dihydrotestosterone; HSP, heat shock protein; ARE, androgen response elements; BRD4, bromodomain-containing protein 4; PSMA, prostate-specific membrane antigen; PSA, prostate-specific antigen; TMPRSS2-ERG, transmembrane protease serine 2:v-ets erythroblastosis virus E26 oncogene homolog; mGluR, metabotropic glutamate receptor; PI3K, phosphoinositide 3-kinases; AKT, protein kinase B; mTOR, mammalian target of rapamycin; PCFT, proton-coupled folate transporter; RFC1, reduced folate carrier

            When AR binds to androgens like testosterone or DHT, the AR molecule undergoes a significant structural change [16]. This change prompts the AR molecule to dissociate from HSPs, allowing AR to interact with ARA70, Filamin-A, and importin-α co-regulators. These interactions facilitate entry of AR into the nucleus, where AR predominantly functions as a homodimer. However, AR is also capable of forming heterodimers with estrogen receptor (ER) isoforms or orphan nuclear receptor testicular receptor 4 (TR4), leading to variations in transcriptional activity [10, 13, 1722].

            AR signalling is further regulated by upstream receptor tyrosine kinases (RTKs). For example, HER-2/neu and G-protein coupled receptor (GPCR) pathways can stimulate AR, even in the absence of androgen binding [2326]. AR will undergo phosphorylation with or without ligand binding at different serine sites, subsequently affecting protein stability and transcriptional activation activity [27]. It has been reported that androgen binding specifically stimulates phosphorylation of AR at Ser64/80/93 to protect AR from degradation [28]. The phosphorylation of other residues, such as Ser213/506/650, via mitogen-activated protein kinases (MAPKs) is pivotal in controlling AR transcriptional activation activity. This process heightens AR sensitivity to minimal levels of androgens, estrogens, and anti-androgens by recruiting essential nuclear co-activators [29].

            Upon AR nuclear translocation, the AR recognizes androgen response elements (AREs) in DNA [30]. AR recruitment of histone acetyltransferase (HAT), along with various co-regulators and core transcriptional elements, initiates target gene transcription, like PSA [18, 31, 32]. When the ligand binds to AR, the ligand will affect protein stability and transcriptional output of AR-DNA complexes. For example, when bound to antagonists, AR continues to move to the nucleus but exhibits reduced retention at AREs, which leads to diminished transcriptional activity [33, 34]. Following dissociation of the ligand, the nuclear export signal promotes the return of AR to the cytosol for preparation of another cycle of ligand binding or proteasomal degradation, thereby maintaining overall AR activity [18, 3538].

            2.2 Prostate-specific membrane antigen (PSMA)

            The FOLH1 gene is known to encode PSMA [39, 40]. PSMA is referred to as N-acetyl-l-aspartyl-l-glutamate peptidase I (NAALADase I) in the fields of neurology and metabolism [41, 42]. Despite these variations in nomenclature, PSMA acts as a glycoprotein on the cell membrane, catalyzing the hydrolysis of folate and carboxypeptides [43, 44] ( Figure 1 ). PSMA has a critical role in processing dietary folates and is primarily distributed in the prostate, kidneys, and duodenum. Interestingly, PSMA is also present in low levels within brain tissue, where PSMA stimulates activation of metabotropic glutamate receptors (mGluRs) [42].

            PSMA is comprised of an intracellular domain, a transmembrane segment, and a large extracellular domain [43, 45]. By cleaving glutamated folate, PSMA is responsible for releasing glutamate, the predominant dietary form of folate [46] ( Figure 1 ). Following the enzymatic action of PSMA, the generated mono-glutamated folate is then absorbed by transporters, such as the folate receptor (FR), reduced folate carrier (RFC)1, and proton-coupled folate transporter (PCFT) [47]. Folate uptake is especially advantageous for rapidly dividing cells because folate supports many cellular processes, including one-carbon metabolism, nucleotide synthesis, and epigenetic methylation, all of which intersect with the methionine cycle [48, 49] ( Figure 1 ). Additionally, under conditions of limited folate availability, hydrolysis of folate by PSMA might confer a growth advantage [50].

            AR and neuroendocrine PrCa (NEPC) modulate the one-carbon metabolic pathway, which is closely intertwined with folate-mediated transfer [5153]. Within the mammalian nervous system, N-acetyl-aspartyl-glutamate (NAAG) is among the most abundant neuropeptides and is particularly present at neuronal synapses [54, 55]. NAAG triggers activation of mGluR3 and is subsequently broken down by PSMA. By releasing glutamate, PSMA promotes phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-AKT-mTOR) signaling [56]. First, glutamate release initiates activation of mGluR I, which leads to phosphorylation of p110β and activation of PI3K. Activated PI3K, in turn, stimulates the AKT-mTOR pathway, driving protein synthesis and cellular proliferation to facilitate tumorigenesis [57] ( Figure 1 ). Notably, preclinical studies have shown that the PSMA inhibitor, 2-PMPA, effectively suppresses PI3K signaling, resulting in tumor regression [56]. Moreover, elevated levels of mGluRs are recognized as a possible adaptive strategy to maintain glutamatergic activity in NEPC [58].

            PSMA-regulated folate metabolism impacts a variety of cellular processes, including tumor cell proliferation, DNA repair, drug resistance, and resource prioritization in tumor microenvironments [5961]. As a result, PSMA contributes to tumor growth and proliferation by increasing folate availability, which drives anabolic metabolic pathways essential for cellular development and division [59]. Additionally, PSMA-regulated folate metabolism is closely linked to DNA repair mechanisms in cancer [60]. Metabolic intermediates, such as methylene tetrahydrofolate produced through one-carbon metabolism, are essential for thymidine synthesis and DNA repair [60]. Disruption of this pathway may result in genomic instability and address drug resistance. Moreover, PSMA regulates resource prioritization in the tumor microenvironment. By enhancing folate utilization, PSMA provides cancer cells with a competitive advantage to sustain high proliferation rates within nutrient-deprived tumor microenvironments [61]. This metabolic adaptation is believed to contribute to the aggressive phenotype of PSMA-positive tumors. Notably, PSMA upregulation corresponds to the increased metabolic demands of tumor cells, which allows tumor cells to outcompete surrounding normal cells for access to folate and related resources [61].

            2.3 Bromodomain-containing protein 4 (BRD4)

            Epigenetic readers, including BRD4, have been shown to be key molecules associated with transcription factors (TFs), like AR, and contribute to the development of aggressive cancers, including PrCa [6266]. The detailed mechanisms by which bromodomain inhibitors exert an effect remain largely undefined [64]. Recent research has shown that changes in DNA accessibility effectively distinguish PrCa from early-stage malignancies and non-cancerous tissues. The increased genomic accessibility observed in advanced tumors appears to be mediated, at least in part, by indirect pathways involving AR interaction with BRDs (BRD4, BRD2, and ATAD2), which have been identified as key facilitators contributing to enhanced accessibility. Notably, these proteins are overexpressed in CRPC and serve as prognostic tissue markers, underscoring their potential as targets in aggressive PrCa [67].

            2.4 Prostate-specific antigen (PSA)

            PSA, a serine protease, is secreted by prostate epithelial cells [68]. Although naturally occurring in normal prostatic secretions, PSA is frequently increased in PrCa patients [69, 70]. For over 2 decades, the serum PSA level has been served as a crucial biomarker in the detection of PrCa [71]. There has been a significant improvement in PrCa management since the 1980s with the introduction of PSA screening, which has contributed to better survival rates. Despite these advances, PSA has limitations as a biomarker, which has prompted the exploration of more accurate diagnostic tools and accelerated the development of PrCa biomarkers [71, 72].

            2.5 Transmembrane protease serine 2: v-ets erythroblastosis virus E26 oncogene homolog (TMPRSS2-ERG)

            A promising biomarker in PrCa diagnostics is the TMPRSS2-ERG fusion. ETS family transcription factors, such as ERG, when fused with the androgen-regulated TMPRSS2 gene represent the most frequently observed gene fusions, accounting for approximately 50% of PrCa cases [73, 74]. A study by Laxman et al. [75] first reported the presence of gene fusion events in the patients with PrCa. This discovery has since been supported by additional studies that indicate TMPRSS2-ERG fusions could function as a urinary biomarker. Notably, these gene fusions exhibit high specificity and a strong positive predictive value. However, the sensitivity of TMPRSS2-ERG fusions is relatively low [76, 77].

            Because most tumors consist of multiple distinct foci, the tumor heterogeneity characteristic of PrCa is a significant limitation to the use of this gene fusion as a biomarker [76]. Moreover, the prognostic value of TMPRSS2-ERG fusion has not been established. Although some studies have shown that this gene fusion correlates with increased cancer aggressiveness, metastasis, and mortality [78, 79], other studies have not confirmed a consistent relationship between this fusion and clinical outcomes, which warrants further in-depth investigations [80].

            3. TARGETED THERAPIES FOR PrCa BIOMARKERS

            The current treatment landscape for PrCa is progressive and adaptable and the strategies evolve according to the stage and status of the disease. PrCa is often detected through early screening methods in the initial diagnosis, with treatments typically including surgical removal of the prostate or radiotherapy. These initial treatments eliminate the tumor confined to the prostate. However, recurrence remains a concern because approximately 30% of PrCa patients experience a return of cancer following local therapy [81] ( Figure 2A ).

            Next follows the figure caption
            Figure 2 |

            Current therapy landscape for PrCa.

            (A). Available treatment options for different stages of PrCa. (B). Various therapies targeting BRD4, AR, PSMA, and immune check points in PrCa. ADT, androgen deprivation therapy; nmCRPC, non-metastatic castration-resistant PrCa; mCRPC, metastatic castration-resistant PrCa; Lu, lutetium; AR, androgen receptor; BRD4, bromodomain-containing protein 4; PD-L1, programmed cell death-ligand 1; PD-L2, programmed cell death-ligand 2; CTLA-4, cytotoxic T-lymphocyte antigen 4.

            Disease recurrence is diagnosed based on elevated PSA levels or imaging techniques that reveal the presence of residual or metastatic disease. When facing biochemical or localized recurrence, physicians may choose to closely monitor PrCa patients without resorting to ADT immediately to avoid the associated side effects. ADT is withheld until disease progression reaches a pre-established threshold. Once this threshold is reached, ADT is typically initiated using luteinizing hormone-releasing hormone (LHRH) antagonists or agonists to decrease androgen levels and inhibit cancer growth. This treatment regimen may be administered intermittently with treatment holidays to help manage side effects until evidence of disease progression occurs [82].

            While ADT can initially achieve disease control, many PrCa patients ultimately become resistant to this therapy and progress to a more advanced form of PrCa (CRPC). This progression marks a turning point in the treatment regimen because CRPC no longer responds to traditional hormone therapies and requires alternative strategies. In cases in which PrCa has already metastasized beyond the prostate gland before the initiation of local treatments, a more aggressive approach is necessary. To this end, physicians and patients must then decide on the early use of ADT, either in conjunction with radiotherapy [83] or docetaxel [84].

            As patients progress through these treatments the tumor acquires resistance to therapies targeting AR but a range of additional treatment options exist. These options include poly(ADP-ribose) polymerase (PARP) inhibitors, which take advantage of DNA repair deficiencies in cancer, and chemotherapy regimens designed to target rapidly dividing tumor cells [85]. Additionally, PSMA-targeted treatments represent a novel and promising option by selectively targeting PSMA-expressing PrCa cells [82]. Moreover, immunotherapy represents an expanding avenue for advanced PrCa by harnessing the immune system to attack cancer cells [82]. This extensive array of treatment strategies, as illustrated in Figure 2 , highlights the complexity and adaptability required in managing PrCa.

            3.1 BRD4 targeted inhibitors

            The molecular landscape in PrCa is highly intricate, with various signaling pathways and genetic mutations contributing to disease progression and drug resistance. One promising therapeutic approach in PrCa targets BRD4, which is implicated in cancer cell survival and proliferation [86, 87]. NEO2734, a novel dual inhibitor, has been developed to target both bromodomain and extra-terminal (BET) proteins (including BRD4) and CBP/p300, a family of transcriptional co-activators with histone acetyltransferase activity. NEO2734 represents an innovative treatment strategy, especially for the most frequent mutated gene in PrCa, Speckle-type POZ protein (SPOP) [88]. Under normal circumstances, wild-type SPOP is pivotal in regulating cellular protein levels by binding to and promoting BET protein degradation. However, this regulatory function is lost when SPOP is mutated, leading to the excessive accumulation of BET proteins. The subsequent dysregulation of gene expression pathways drives cancer growth. This loss of function often results in drug resistance against BET inhibitors (BETis) in SPOP-mutant PrCa patients [88].

            Preclinical studies have reported that NEO2734 significantly inhibits growth of SPOP-mutant PrCa [88]. Furthermore, NEO2734 has shown efficacy against enzalutamide-resistant PrCa cells and patient-derived organoids, indicating the potential use as a treatment strategy for cases that have developed resistance to anti-androgen therapies [89]. Recent research has also explored the function of AR splice variants (AR-Vs) in conferring increased castration resistance in PrCa. It has been observed that NEO2734 counteracts the mechanisms associated with anti-androgen-induced ferroptosis [90]. By inhibiting this process, NEO2734 offers a promising strategy for overcoming resistance that restricts the effectiveness of existing therapies [90]. Currently, NEO2734 is being investigated in clinical trials for CRPC patients (NCT05488548). This trial aims to evaluate the safety, tolerability, and preliminary efficacy of NEO2734 in CRPC patients, which offers hope for a novel therapeutic option that targets multiple components of the complex molecular machinery driving PrCa progression.

            3.2 PSMA targeted therapy: Lu 177-PSMA-617

            Pluvicto (177Lu-PSMA-617) is another promising therapeutic option for targeting androgen receptor variant-containing cells, especially those cells that also display PSMA. This innovative treatment consists of a molecule targeting PSMA conjugated to 177Lu, a radioactive isotope. The mechanism of action enables 177Lu-PSMA-617 to selectively bind PSMA-expressing PrCa cells, facilitating the precise delivery of radiation to the tumor without damaging normal tissues. As such, 177Lu emits radiation that directly harms the DNA of cancer cells, ultimately leading to cell death [91]. This precision-targeted approach has yielded highly promising outcomes, especially in advanced-stage PrCa, as demonstrated in the NCT03511664 clinical trial [92].

            Recent studies have explored molecular biomarkers in patients treated with Pluvicto. One study analysed circulating tumor cells and investigated clinical factors, such as protein levels of PSA and PSMA expression, tumor size, and the levels of AR and AR-V7 expression [5]. The findings revealed that AR and AR-V7 expression could be used as prognostic markers in metastatic CRPC patients before starting Pluvicto therapy. Interestingly, while these biomarkers correlated with tumor load, the biomarkers did not predict the patient’s response to PSMA-targeted therapies. Notably, AR-V7 was associated with higher AR expression, increased tumor load, and an elevated level of PSMA protein [5].

            Alternatively, Pathmanandavel et al. [93] reported that among AR-V7-positive metastatic (m)CRPC patients treated with 177Lu-PSMA-617, AR-V7 did not adversely impact overall survival. This observation suggested that 177Lu-PSMA-617 could be a viable therapy for mCRPC patients expressing AR-V7 along with established treatments, such as taxanes. However, exploring the function of protein expression and the potential value of these markers in determining treatment outcomes is important in further research.

            The therapeutic potential of 177Lu-PSMA-617 continues to be investigated across multiple ongoing clinical trials. The PSMA fore-trial (NCT04689828) aims to compare the efficacy of 177Lu-PSMA-617 therapies in mCRPC patients targeting the AR. Another active trial is investigating pairing Pluvicto with pembrolizumab, an immune checkpoint inhibitor [ICI] (NCT03805594) [94, 95].

            3.3 Immunotherapy: nivolumab and ipilimumab

            ICIs, including nivolumab and ipilimumab, have been shown to be a potential treatment strategy for advanced PrCa, especially in AR-V7+ PrCa patients. These inhibitors target immune response regulators, effectively enhancing the ability of immune cells to eliminate cancer cells [96]. Nivolumab targets immune cells by blocking the programmed cell death protein (PD)-1 receptor. Under normal conditions, PD-1 binds to physiologic ligands (PD-L1 and PD-L2) expressed on the cancer cell surface, resulting in suppression of the immune reaction and allowing the cancer cells to escape immune surveillance. Blocking this interaction, nivolumab reactivates the immune response, allowing the immune response to recognize and attack tumor cells more effectively. In contrast, ipilimumab targets another checkpoint molecule, cytotoxic T-lymphocyte antigen 4 (CTLA-4), a membrane receptor on immune cells that suppresses immune activity. By inhibiting CTLA-4, ipilimumab prevents the inhibitory signaling pathway, which keeps the immune cells active and enables tumor cells to target cancer cells [96].

            In clinical practice nivolumab and ipilimumab are frequently used together to create a synergistic immune reaction towards cancer cells. NCT02601014, a clinical trial evaluating this combination in AR-V7+ PrCa patients, demonstrated promising efficacy. However, the response was not consistent across AR-V7+ patients [97].

            The efficacy of combining nivolumab and ipilimumab, either along with or independent of enzalutamide, in AR-V7+ mCRPC patients was investigated in the same phase II trial. While combination therapy had an acceptable profile regarding safety, combination therapy had limited efficacy in this patient population, even when combined with enzalutamide [98]. Further insights were gained from another phase II trial (NCT02985957). Preliminary findings suggested that some CRPC patients experience complete responses to the combination of nivolumab and ipilimumab, with four participants achieving complete remission [99]. However, the study did not establish a consistent correlation between treatment efficacy and tumor mutation burden in blood or tissue samples [99].

            Numerous clinical trials are actively exploring the combination of ICIs with other therapeutic agents. These trials involved the combination of 177Lu-PSMA (NCT05150236) and stereotactic body radiotherapy [SBRT] (NCT05655715). Another promising immunotherapy for metastatic PrCa is the DNA vaccine, MVI-118, which encodes the ligand-binding domain (LBD) of the AR and induces the immune response against AR-overexpressing cancer cells mediated by CD8+ T. MVI-118 was shown to be safe and effective in NCT02411786 with respect to activating the immune system in PrCa [100]. Two other clinical trials are currently investigating the MVI-118. One trial is exploring the use of the pTVG-HP DNA vaccine with or without MVI-118 and pembrolizumab in mCRPC patients, while the other trial is examining ADT with or without MVI-118 and/or nivolumab in PrCa patients, according to the NCT04090528 and NCT04989946 clinical trials, respectively.

            3.4 Limitations of current approaches

            Despite the development of numerous targeted therapies aimed at PrCa biomarkers, these strategies often encounter substantial challenges, which highlight the pressing need for novel therapeutic innovations. PROTAC technology has emerged as a transformative approach, offering unique advantages that address key limitations associated with conventional methods [8]. Compared to the aforementioned therapeutic modalities, PROTACs overcome several notable constraints, underscoring the potential of this technology to redefine treatment paradigms for PrCa.

            3.4.1 Resistance to AR inhibitors

            Enzalutamide and abiraterone, the current standard AR inhibitors, are effective in prolonging survival [101104]. However, several problems, including AR gene amplification, overexpression of splice variants with constitutive activity (e.g., AR-V7), mutations in the AR LBD, and activation of compensatory signaling pathways, contribute to resistance mechanisms and limit clinical efficacy [5, 6]. Compared to the inhibitors, PROTACs offer a unique approach by degrading AR proteins, including splice variants and mutant forms, and may potentially overcome these resistance mechanisms [8].

            3.4.2 Toxicity and side effects of chemotherapy

            Chemotherapy remains a standard treatment option for advanced CRPC, including docetaxel and cabazitaxel for PrCa therapy [105]. However, these agents have been reported to exhibit significant toxicities, such as neutropenia, neuropathy, and fatigue, which limit long-term use [106]. PROTACS may offer a safer therapeutic alternative with fewer off-target effects, in part due to the specificity and targeted mechanism of action.

            3.4.3 Mechanistic limitations of conventional therapies

            The mechanisms of action of traditional therapies primarily focus on inhibiting AR signaling [106] rather than eliminating the functional AR, which results in drug resistance. In contrast, PROTACs aim to entirely degrade the AR protein, the key driver of CRPC progression. This degradation likely disrupts downstream transcriptional activity and reduces the likelihood of reactivation.

            3.4.4 Addressing unmet needs

            PROTACs are designed to target proteins beyond AR that are involved in CRPC progression, thereby expanding the therapeutic scope and enabling personalized treatment strategies based on specific molecular profiles. However, achieving this goal may be challenging with traditional inhibitors.

            In conclusion, the unique therapeutic value of PROTAC technology lies in an ability to address the key challenges of resistance, toxicity, and limited mechanistic action, which underscores the potential in treating CRPC.

            3.5 PROTACs towards AR

            Among the innovative therapeutic strategies under investigation for PrCa, PROTACs have emerged as promising strategies to selective degrade target proteins (POI) [8]. One of the leading PROTACs in clinical development is ARV-110, which became the first PROTAC used in clinical trials by targeting AR for destruction. By leveraging the ubiquitin-proteasome system (UPS), ARV-110 specifically induces AR degradation [107].

            The mechanism by which ARV-110 functions involves forming a ternary complex that induces the AR proximity between POI with an E3 ligase (specifically, cereblon [CRBN]) [8]. CRBN consists of two distinct components linked by a flexible linker. One side features a non-covalently binder that binds to the AR and the other side contains a thalidomide-derived “warhead” that covalently attaches to the CRBN. This binding induces the formation of the E3 ligase-ARV-110-AR complex, consisting of the E3 ligase, ARV-110, and the AR protein. By bringing AR and CRBN into proximity, ARV-110 facilitates tagging of ubiquitin molecules to AR, ultimately leading to degradation ( Figure 2B ). This process results in complete degradation of the AR, rather than merely inhibiting the AR, thereby significantly reducing the AR level within cancer cells.

            What makes ARV-110 particularly innovative is that, unlike traditional AR inhibitors that simply block AR activity, ARV-110 actively triggers the AR degradation. This mechanism is advantageous, especially in cases in which PrCa cells have developed resistance to conventional AR therapies. By degrading, rather than merely inhibiting the AR protein, ARV-110 offers a potential solution to overcoming resistance mechanisms arising from mutations in the AR LBD, which often reduce the treatment efficacy of standard therapies. Another essential advantage of ARV-110 is the catalytic mechanism. The free PROTAC and E3 ligase complex can be recycled and participate in the next round of degradation. This catalytic mechanism renders ARV-110 a highly efficient therapeutic agent, offering superiority over traditional inhibitors in a stoichiometric manner. By exploiting the UPS to degrade AR, ARV-110 represents a new frontier in targeted cancer therapy, potentially offering a more effective treatment method for PrCa patients [8].

            4. PROXIMITY STRATEGIES

            4.1 Membrane protein degradation strategy

            Endocytosis has an important role in the macromolecule uptake pathway ( Figure 3A ). The internalized macromolecules bind to the specific receptors on cell surface and accumulate in the clathrin-coated pits. After detaching from the membrane, these pits facilitate the enclosure of receptors and corresponding ligands through forming vesicles. The vesicles will subsequently merge with early endosomes and are then directed to lysosomes or back to the cell surface for recycling [108].

            Next follows the figure caption
            Figure 3 |

            Targeted protein degradation therapies utilize different pathways, such as the endocytosis, proteosome, and autophagy pathways.

            (A). The endocytosis pathway is exploited to degrade membrane proteins (POIs) using engineered bispecific antibodies. (B). The proteosome pathway is leveraged by PROTACs to ubiquitinate and degrade target POIs. (C). AUTACs and ATTECs are designed to degrade POI or organelles in cells by hijacking the autophagy pathway. Ub, ubiquitin; ASGPR, asialoglycoprotein receptor; CXCR7, C-X-C chemokine receptor type 7; RNF43, ring finger protein 43; POI, protein of interest; KineTAC, cytokine receptor targeting chimera; LYTAC, lysosome-targeting chimera; PROTAB, proteolysis-targeting antibody; E1, ubiquitin-activating enzyme; E2, ubiquitin-conjugating enzyme; E3, ubiquitin ligase; PROTAC, proteolysis targeting chimera; ATTEC, autophagosome-tethering compound; AUTAC, autophagy-targeting chimera; LC3, microtubule-associated protein light chain 3.

            Various strategies have hijacked this endocytosis pathway to degrade membrane proteins. The strategies are as follows lysosome-targeting chimeras (LYTACs), which utilize receptors for lysosomal degradation of target proteins, such as the mannose-6-phosphate receptor [109, 110]; proteolysis-targeting antibodies (PROTABs), bifunctional antibodies that hijack RNF43 to ubiquitinate membrane proteins [111]; cytokine receptor-targeting chimeras (KineTACs), bispecific antibodies that use cytokine receptors to guide target proteins to lysosomes [112]; and transferrin receptor-targeting chimeras (TransTAC), an efficient degrader that functions through leveraging the transferrin receptor to target and eliminate membrane proteins [113]. These emerging technologies present promising strategies for disease-associated membrane protein degradation, revolutionizing the treatment landscape by rendering previously “undruggable” targets amenable to therapeutic intervention.

            4.2 PROTACs strategy

            In addition to endocytosis, a key mechanism for regulating protein levels in eukaryotic cells is ubiquitination, followed by targeted protein degradation. Cellular ubiquitination-induced proteolysis is a pathway in which proteins undergo polyubiquitination, then degradation by the proteasome or lysosome [114117]. This mechanism is precisely controlled and consists of a cascade of enzymatic activities, including E1, E2, and E3, to facilitate attachment of ubiquitin to the substrate protein lysine residues [118]. Importantly, the ubiquitination process is reversible because deubiquitinases (DUBs) cleave polyubiquitin chains from modified proteins, thereby allowing for dynamic regulation of protein stability in cells [119].

            Various types of polyubiquitin chains are formed, each serving distinct biological functions, due to the presence of seven lysine residues in ubiquitin protein. For example, two polyubiquitin chains (K11 and K48) primarily tag proteins, then degrade the proteins, while K63-linked chains typically function as docking sites for protein-protein interactions, facilitating signal transduction events [116, 117]. The specificity of the ubiquitination process is controlled by E3 ligase [120]. However, a limited number of these ligases have clearly defined degron recognition capabilities among > 600 E3 ligases [121]. Degron recognition is a crucial process for removing misfolded or damaged proteins and maintaining intracellular protein balance. Targeted protein degradation (TPD) that selectively degrades pathogenic proteins is a promising therapeutic strategy.

            PROTACs, novel bifunctional molecules, are engineered to exploit the ubiquitin-proteasome system for the degradation of POIs [122]. This kind of design allows PROTACs to form a ternary structure between the POI and an E3 ligase, facilitating targeted protein ubiquitination and degradation ( Figure 3B ). Notably, an AR targeting the PROTAC compound, ARV-110, is currently undergoing clinical trials for treating PrCa [8].

            4.3 AUTACs strategy

            Recent advances in autophagy-based degraders have introduced two notable examples (autophagy-targeting chimeras [AUTACs] and autophagosome-tethering compounds [ATTECs]). AUTACs represent one of the pioneering approaches in this field, as reported by Takahashi et al. [123, 124]. ATTECs developed by Li et al. [125, 126] function as molecular glues that mimic autophagy adaptors. This model involves the targeted degradation of specific proteins via autophagy, in which ATTECs facilitate the process by acting similar to natural autophagy adaptors ( Figure 3C ). While ATTECs and AUTACs have not been used for AR degradation, these platforms hold significant promise for future applications in CRPC therapy.

            5. PROTAC STRATEGIES

            5.1 Current PROTACs with different component designs

            A PROTAC molecule consists of three essential parts (the E3 ligase ligand, a POI ligand, and a linker that connects these two ligands). Each of these elements is crucial in facilitating the targeted degradation. The detailed characteristics are discussed in the following sections.

            5.1.1 PROTAC with various E3 ligases

            Various E3 ligases, such as β-transducin repeat-containing protein (β-TrCP) [127], von Hippel-Lindau (VHL) [128], MDM2 [129], CRBN [130], and cellular inhibitor of apoptosis protein (cIAP)1 [131] have been used in PROTAC development. The catalytic nature of PROTACs allows PROTACs to act repeatedly, making PROTACs highly effective in POI degradation [132, 133].

            5.1.2 PROTAC with different linkers

            Figure 4A provides a classification of the most frequent used linkers in according to the structural data in the database [134]. The current linker categories include flexible linkers, rigid linkers, triazole-based linkers, bio-orthogonal clickable linkers, and photo-switchable linkers.

            Next follows the figure caption
            Figure 4 |

            PROTAC construct and the mechanism for controllable PROTAC.

            (A). The PROTAC construct, which includes E3 ligase ligands, target protein ligands, and the linkers connecting the E3 ligase and target protein ligands. The reported types of E3 ligases, linkers, and target proteins in PROTACs are summarized in the indicated boxes. B. The design and working mechanism of controllable PROTACs.

            A common strategy in modifying the linkers to enhance the solubility of PROTACs involves incorporating heterocycles (saturated) with a basic center. Examples, such as the piperidine or piperazine rings, belong to the category of rigid linkers frequently used in PROTAC design ( Figure 4A ) [135137]. The inclusion of piperidine or piperazine not only contributes to the increased rigidity of the PROTAC, which can potentially enhance PROTAC activity but also introduces a protonable amino group that may improve solubility. Piperazine-containing linkers offer the advantage of enhancing the overall solubility of the molecule. However, data from the PROTAC-DB, an open-access database compiling PROTAC information, indicates that the pKa of an acidic or basic center within a molecule can be significantly influenced by neighbouring groups [138, 139] and affecting PROTAC properties.

            5.1.3 PROTAC targets of different POIs

            The ideal targets for PROTAC therapy typically possess a set of common characteristics, especially alterations from their natural state, such as overexpression, mutation, protein aggregation, and changes in isoform expression. Additionally, an optimal target should feature an accessible surface that allows E3 ligase binding. Furthermore, the presence of a flexible domain that can facilitate efficient and effective translocation into the proteasome is considered advantageous for facilitating protein degradation [139, 140].

            5.1.3.1 Inflammation, immunity, and immuno-oncology

            Two promising candidates, a Bruton’s tyrosine kinase (BTK) degrader (NX-5948) [141], and an IRAK4 degrader (KT-474) [142], are currently being investigated to treat a range of immuno-inflammatory conditions in clinical trials. There is a highly validated therapeutic target-BTK in inflammatory diseases and oncology with several approved treatments already available [143, 144]. However, the rise in the C481S resistance mutation in BTK has diminished the efficacy of first-generation BTK inhibitors, driving significant interest in developing PROTAC-based therapies to address this challenge [145147]. PROTACs offer a key advantage through event-driven pharmacology, enabling PROTACs to catalytically degrade target proteins rather than merely inhibit target proteins. This mechanism holds promise for overcoming resistance mutations like C481S, which reduce the potency of conventional, occupancy-driven inhibitors [148, 149].

            5.1.3.2 Neurology and neurodegeneration

            One breakthrough in PROTAC technology is the ability to degrade undruggable proteins. These proteins typically cannot be targeted by inhibitors due to a lack of binding sites. The advantage of PROTAC make it possible to address these disease-causing proteins, especially in neurodegenerative diseases, such as tau [150157], α-synuclein [158, 159], and mutant huntingtin (mHTT) [125, 160162].

            Lu et al. [154] identified Peptide 1, a KEAP1-dependent PROTAC, which was shown to degrade tau protein, highlighting the potential in the future application. In 2020, Qu et al. [159] introduced another TAT-PBD-PTM based on peptides that specifically targets and degrades α-synuclein in Parkinson’s disease [159]. Additionally, Tomoshige [162] designed hybrid small molecules (compounds 1 and 2), which effectively degrade mHTT, demonstrating promise for the treatment of Huntington’s disease [162].

            5.1.3.3 Transcription factor

            Transcription factors and RNA-binding proteins (RBPs) have critical roles in DNA integrity, synthesis, gene regulation, and diverse RNA-mediated mechanisms [163165]. Despite the importance of transcription factors and RBPs, many of these POIs without ligand-binding sites are notoriously challenging to target with traditional drugs [166, 167]. To address these “undruggable” targets, various studies have been conducted to establish innovative approaches. For example, transcription factors have been targeted using strategies, such as transcription factor-targeting chimera (TRAFTAC) [168], transcription factor PROTAC (TF-PROTAC) [169], oligonucleotide-PROTAC (O’PROTAC) [170], and G-quadruplex PROTAC (G4-PROTAC) [171]. Beyond transcription factors, telomeres have been targeted using telomere-targeting chimera (TeloTAC) [172], showcasing the potential to expand to more sophisticated methodologies. Additional advanced strategies include developing Myc-targeting PROTACs using a TNA-DNA bivalent binder [173], MeCP2 degraders that utilize methylated DNA [174], and structurally specific Z-DNA PROTACs for targeting Z-DNA binding proteins [175]. These DNA- or RNA-based PROTACs use consensus RNA or DNA sequences, which are linked to an E3 ligase ligand. This innovative approach significantly broadens the possibilities for targeting previously undruggable proteins within cells, offering new avenues for therapeutic intervention.

            5.2 Current controllable PROTACs

            Several controllable strategies for PROTACs have been reported, as summarized in Table 1 , including stimuli-responsive approaches, such as photoactivation, folate targeting, hypoxia sensitivity, X-ray induction, and other external triggers.

            Table 1 |

            Current controllable PROTACs.

            Caging strategyPROTAC nameTarget proteinE3 ligandReference
            Photo-controlledpc-PROTAC1dBET1CRBN[176]
            Opto-pomalidomideIKZF1/3CRBN[177180]
            Opto-dBET1BRD4CRBN
            Opto-dALKALKCRBN
            Trans-photoPROTAC-1BRD2VHL[181]
            DEACMERRαVHL[179]
            NPOMBRD4CRBN[179]
            DMNBBRD4VHL[180]
            Folate-controlledfolate-PROTACBRD3/4VHL[182, 183]
            Hypoxia-controlledha-PROTACEGFRCRBN[184]
            NTR-PROTACEGFRVHL[185]
            X-ray-controlledRT-PROTACsBRD4VHL[186]
            Stimuli-responsivesr-PROTACBRD4CRBN[187]
            5.3 Recent developments in AR degradation technology

            AR degraders utilizing the PROTAC principle have facilitated significant progress with a growing number of unique AR degraders being synthesized and tested for efficacy against PrCa against PrCa ( Table 2 ).

            Table 2 |

            Summary of AR-PROTAC.

            E3PROTAC nameReference
            CRBNARV-110[8]
            PROTAC 35[188]
            TD-802[189]
            A16[190]
            Compound 13b/13c[191]
            ARD-2128[192]
            ARD-2585[193]
            BWA-522[200]
            ARD-2051[201]
            ARD-1676[202]
            VHLARD-266[194]
            A031[195]
            Niclo-Click PROTAC 5[196]
            MTX-23[197]
            ARCC-4[198]
            ARD-69[137]
            ARD-61[199]

            CRBN-based PROTACs, especially ARV-110, are among the most widely studied PROTACs for AR degradation [8]. One notable study modified the FDA-approved AR inhibitor, enzalutamide, using various scaffolds in combination with different linkers. The most promising compound from this subseries, PROTAC 35 (compound 1), exhibited potent degradation capabilities with an approximate half-maximal degradation concentration (DC50) of 80 nM in an LNCaP cell line. Specificity tests also indicated that compound 1 exhibited a strong preference for LNCaP PrCa, suggesting its potential for targeted therapy in AR-expressing PrCa [188]. In another study Takwale et al. [189] developed various CRBN-mediated AR PROTACs by combining the AR antagonist with the CRBN ligand. Compound TD-802 showed the highest degradation efficiency of AR in LNCaP cells [189]. Targeting AR using RU59063 derivatives, Liang and colleagues [190] developed novel AR PROTACs. Among these AR PROTACs, compound A16 (also known as compound 3) emerged as the leading candidate, matching the efficacy of enzalutamide [190]. Kim and colleagues [191] synthesized PROTACs using bicalutamide analogs with a PEG linker attached to thalidomide, which serves as a CRBN recruiter. Among these PROTACs, compound 13b (compound 4) effectively decreased AR-regulated gene expression. Compound 13c (compound 5) demonstrated even greater AR degradation, particularly in targeting AR-V7, making it more effective than the wild-type AR and suggesting its potential for treating AR-V-expressing PrCas [191]. In another significant development, Wang et al. [192] developed ARD-61 (compound 14), a highly potent AR degrader. To address the poor oral bioavailability, Wang et al. [192] optimized the molecule by incorporating the CRBN ligand, resulting in the development of ARD-2128 (compound 6). This compound exhibited excellent oral bioavailability and showed potent AR degradation, significantly reducing AR, PSA, TMPRSS2, and FKBP5 levels, demonstrating the potential as an orally administered AR-targeted therapy [192]. Finally, Xiang et al. [193] developed ARD-2585 (compound 7), which emerged as an efficient degrader of AR. Pharmacokinetic and tissue distribution studies confirmed ARD-2585 promise as an AR degrader, suggesting that ARD-2585 could become a highly effective therapy for PrCa, particularly for patients with resistance to conventional AR inhibitors [193].

            In addition to CRBN-recruiting PROTACs, VHL-recruiting PROTACs have also been developed to target AR for degradation with several novel compounds demonstrating promising potential as therapeutic agents in PrCa treatment. Han et al. [194] analysed the correlation between the VHL ligand binding affinity and degradation efficacy. The findings advocated using ligand with a relatively low binding affinity (2–3 μM) could still produce highly effective AR degraders. Optimized ARD-266 (compound 8) emerged as a standout candidate, achieving > 90% AR degradation in 10 nM after further optimization. ARD-266 degraded > 95% AR in PrCa within 6 h [194]. Chen and colleagues [195] reported another degrader, A031 (compound 9), which was optimized from two AR antagonists and several E3 ligands. By comparing the linker composition with different heterocyclic and phenyl rings, Chen et al. [195] reported that the AR ligand rather than the linker has a substantial effect on the overall activity of the PROTACs. Munoz et al. [196] developed niclosamide-based PROTACs that utilized VHL-032 as the VHL ligand. One compound, Niclo-Click PROTAC 5, achieved a half-maximal inhibitory concentration (IC50) of approximately 1 μM in the LNCaP cell line but the effectiveness was lower than clinical candidates, like ARV-110 [196].

            Lee et al. [197] introduced MTX-23 (compound 11), a unique PROTAC that has been demonstrated to degrade both AR-FL and AR-V7. Compound 11 The compound 11 DC50 for AR-V7 and AR-FL was 0.37 and 2 μM in immunoblotting studies, respectively. Compound 11 effectively inhibits cell division and induces programmed cell death in PrCa cells, even in models resistant to enzalutamide and abiraterone [197]. Salami et al. [198] synthesized VHL-based PROTACs targeting AR by utilizing enzalutamide. The most potent compound, ARCC-4 (compound 12), displayed a DC50 of 5 nM. ARCC-4 effectively degraded AR in VCaP cells and degraded several clinically relevant AR mutants, demonstrating enhanced efficacy in inducing apoptosis and inhibiting proliferation in CRPC cells [198].

            In further optimization efforts, Han et al. [137] developed ARD-69 (compound 13), which exhibits exceptional potency. ARD-69 was optimized by adjusting the linker length and incorporating a pyridine group, enhancing both activity and solubility [137]. Finally, Kregel et al. [199] reported ARD-61 (compound 14), an AR degrader effective against PrCa resistant to standard treatments. ARD-61 triggers PARP cleavage in multiple AR-driven cell lines and exhibits potent activity in the enzalutamide resistant xenograft model. Despite not directly degrading AR-V7, ARD-61 inhibits tumor growth in AR-V7 overexpressing models, suggesting that full-length AR remains crucial for survival in CRPC [199].

            Zhang et al. [200] developed a novel PROTAC (BWA-522), which is designed to target to the N-terminal transcriptional domain of AR (AR-NTD). By binding to this domain, BWA-522 induces degradation of AR-FL and the AR-V7 variant in PrCa cell lines. BWA-522 exhibits promising therapeutic value in in vitro and in vivo studies, with a sub-micromolar DC50 value in VCaP cells and a 76% inhibition of tumor growth in LNCaP xenograft mice studies, respectively [200].

            ARD-2051, another potent PROTAC degrader of AR, was developed by Han et. al in 2023 [201]. ARD-2051 exhibits a 0.6 nM DC50 and effectively inhibits AR-regulated genes, thereby suppressing cancer cell growth. Moreover, oral bioavailability studies indicate favorable pharmacokinetic properties of ARD-2051, making ARD-2051 a potential agent for PrCa [201].

            Moreover, ARD-1676, which was developed by Xiang et al. [202], exhibits highly efficient AR degradation, with DC50 values of 0.1 and 1.1 nM in VCaP and LNCaP cells, respectively.

            In conclusion, the advances in AR-targeted PROTACs provide novel strategies to overcome the limitations of inhibitors, such as enzalutamide, paving the way for enhanced treatment strategies for PrCa.

            5.4 Recent developments in non-AR targets

            While initially developed to degrade the AR in PrCa due to its critical role in disease progression, the application of PROTACs has been broadened to include non-AR proteins implicated in PrCa, such as BET [203, 204], BCL6 [205], FAK [206], and CDKs [207].

            BET proteins, which are key epigenetic regulators, have a central role in gene transcription [208]. The degradation of BET proteins has been shown to inhibit PrCa cell growth, motivating the development of PROTACs targeting BET proteins. For example, the degrader, MZ1, selectively degrades BRD4 by disrupting its interaction with acetylated chromatin [203]. This disruption suppresses MYC-driven gene transcription, ultimately inhibiting cancer cell proliferation. Furthermore, MZ1 has demonstrated potent anti-proliferative effects in prostate cancer models [203].

            PROTACs targeting B-cell lymphoma 6 (BCL6), a transcriptional repressor, have been primarily investigated in lymphomas [205]. For example, the PROTAC, BI-3802, induces the proteasomal degradation of BCL6, leading to reactivation of tumor-suppressor genes. While BI-3802 has primarily been studied in lymphomas, the mechanism of action offers valuable insight into the potential for targeting BCL6 pathways in PrCa.

            Focal adhesion kinase (FAK), a non-receptor tyrosine kinase involved in cell adhesion and survival signaling pathways, is specifically degraded by PROTACs (PROTAC-FAK-01) [206]. In prostate cancer models, PROTAC-FAK-01 disrupts focal adhesion complexes, effectively suppressing cell migration and proliferation.

            Cyclin-dependent kinases (CDKs), such as CDK4/6, serve as key regulators of the cell cycle [209]. PROTACs targeting specific CDKs have been investigated to induce cancer cell apoptosis [207]. Jiang et al. [207] developed dTAG-CDK4/6, a PROTAC that degrades these kinases, with significant tumor-suppressive activity observed in prostate cancer xenograft models.

            These developments highlight the versatility of PROTAC technology in targeting a broad range of proteins beyond AR in PrCa, potentially addressing various oncogenic pathways involved in the disease.

            5.5 Recent clinical progress of PROTAC technology

            Table 3 presents an overview of recent clinical progress in PROTAC technology, highlighting key details, such as disease indications, developmental phases, preliminary outcomes, and the corresponding challenges and implications [8, 107, 141, 142, 200202].

            Table 3 |

            Summary of clinical progress involving PROTACs and inhibitors.

            PROTACTargetIndicationPhase of DevelopmentPreliminary OutcomesChallenges and Implications
            Enzalutamide [102]ARmCRPCFDA approvedImproved overall survival and radiographic progression-free survival in phase 3 trials; PREVAIL trial showed a median survival of 32.4 months vs. 30.2 months (placebo).A standard treatment across prostate cancer stages; resistance development and side effects (fatigue, hypertension, and seizures) limit long-term efficacy.
            Bavdegalutamide (ARV-110) [8, 107]ARmCRPCPhase I/IIAntitumor activity observed, particularly in patients with AR ligand-binding domain mutations (T878 and H875). Demonstrated a manageable tolerability profile in clinical trials.Companion diagnostics remain essential for identifying patients harboring specific AR mutations. Addressing resistance mechanisms and ensuring therapeutic specificity are critical for success.
            NX-5948 [141]BTKB-cell malignancies and autoimmune diseasesPhase IResponses observed within 8 weeks; treatment was well-tolerated with common adverse events including purpura, thrombocytopenia, and neutropenia.Ensuring selective BTK degradation is vital to minimize off-target effects. Determining optimal dosing regimens and managing immune-related adverse events remain key challenges.
            KT-474 [142]IRAK4Autoimmune and inflammatory diseasesPhase IGreater than 95% reduction in IRAK4 levels and up to 97% reduction in pro-inflammatory cytokines observed in healthy volunteers. Maintained a favorable safety profile, with only mild-to-moderate adverse events reported.Translating findings from healthy volunteers to patients with autoimmune diseases poses challenges. Long-term studies are needed to confirm safety and efficacy for therapeutic potential.
            BWA-522 [200]AR-NTDProstate cancerPreclinicalDemonstrated efficacy in degrading AR-FL and AR-V7, inducing apoptosis in cell lines and achieving significant tumor growth inhibition in xenograft models.Offers a strategy for targeting resistant AR variants. Challenges include ensuring the translation of preclinical success into clinical efficacy and overcoming resistance mechanisms.
            ARD-2051 [201]ARAdvanced prostate cancerPreclinicalHigh degradation potency with Dmax > 90%, suppression of AR-regulated genes, and inhibition of cancer cell growth were observed in preclinical models. Demonstrated good oral bioavailabilityDeveloping companion diagnostics for AR mutations and refining oral dosing strategies is critical for therapeutic benefit in patients.
            ARD-1676 [202]AR and AR mutantsAR-positive prostate cancerPreclinicalDC50 values ranging from 0.1–1.1 nM in cell lines achieved >90% degradation. Oral administration effectively reduced AR protein levels in tumor tissues and inhibited tumor growth without toxicity.Exceptional pharmacokinetics and activity against AR mutants suggest significant therapeutic potential. Addressing off-target effects and achieving robust clinical trial outcomes remain key hurdles.

            6. PERSPECTIVE AND FUTURE DIRECTIONS

            This section explores the key mechanisms driving AR signaling in CRPC and discusses the future directions for PrCa treatment by harnessing the PROTAC strategy. We focused on the mechanisms underlying AR amplification and overexpression, AR splice variants, mutations leading to receptor promiscuity, post-translational modifications, interactions with co-activators and co-repressors, and intra-tumoral steroid hormone synthesis. Figure 5 highlights the diverse AR-dependent pathways, showcasing therapeutic targets that could potentially be leveraged to enhance CRPC treatment strategies.

            Next follows the figure caption
            Figure 5 |

            Potential molecular mechanisms underlying CRPC.

            The scheme illustrates seven potential mechanisms of CRPC, including aberrant activation of AR, AR amplification, intra-tumoral steroid synthesis, AR splicing variants, AR promiscuity, post-translational modifications of AR, and coregulator modification.

            6.1 AR amplification and overexpression

            AR amplification, characterized by an increased AR gene count surpassing the normal diploid level, is recognized as a prominent characteristic of CRPC and has been identified in 20–31% of cases [210212]. Fluorescence in situ hybridization analysis has shown that AR amplification is uncommon in the early-stage of PrCa but becomes more prevalent as the disease advances to castration-resistant stages, correlating with increased AR mRNA expression. For example, a two-fold increase in AR mRNA levels has been demonstrated in CRPC tumors, highlighting the essential function of AR amplification in enabling tumor adaptation to androgen-depleted conditions [212, 213]. Chen et al. [214] further validated that CRPC cells maintain elevated AR protein levels, emphasizing the function of AR amplification and overexpression in sustaining AR signalling, even in low androgen conditions. In response to these findings, degrader-based therapies have emerged as promising strategies for addressing CRPC driven by AR amplification and overexpression. A notable example is ARV-110, a PROTAC targeting AR, has shown potential in clinical trials by degrading AR while mitigating the effects of overactive AR signalling pathways in CRPC [8].

            6.2 Posttranslational modifications

            Moreover, post-translational modifications showcase a pivotal role in the stabilization of AR protein in CRPC, thereby contributing to receptor sustained activity. These modifications, such as phosphorylation, acetylation, ubiquitination, and SUMOylation, are crucial regulators that modulate AR stability, promote AR translocation into the nucleus, and fine-tuning AR gene expression and function [215]. Collectively, these post-translational changes ensure prolonged AR signaling, further complicating the management of CRPC and highlighting the urgent need for targeted therapeutic strategies [215]. The interaction with HSPs, particularly the HSP family, acts as a critical molecular chaperone, safeguarding AR from degradation while preserving the active conformation [216]. This protective mechanism allows PrCa cells to remain highly responsive to even minimal levels of circulating hormones, enabling sustained proliferative growth in an androgen-deficient environment. Consequently, the involvement of HSPs in maintaining AR stability further exacerbates the challenge of targeting AR signaling in CRPC, necessitating more advanced therapeutic approaches to disrupt this protective interaction.

            6.3 AR splice variants

            Another critical adaptation mechanism in CRPC is the emergence of AR splice variants, which generate truncated AR protein. Among these, AR-V1 and AR-V7 are particularly notable for their roles in therapy resistance. Lacking the LBD but retaining the N-terminal and DNA-binding domains, AR-V1 and V7 function as constitutively active transcription factors independent of androgen binding. This ligand-independent activity enables the splice variants to drive AR signaling continuously, contributing to therapeutic resistance and tumor progression in CRPC [217, 218]. This ligand-independent activity enables AR splice variants to drive downstream pathways even without circulating hormones. The expression has a particularly significant role in driving resistance to enzalutamide and abiraterone [219, 220]. These therapies aim to decrease the activity of AR by inhibiting the LBD domain. However, the AR variants circumvent this blockade because the AR variants lack the LBD. Consequently, AR splice variants contribute to primary resistance, which occurs in 20%–40% of patients, and secondary resistance, which almost invariably develops over time in nearly all treated patients. This finding highlights the pressing need for therapeutic strategies that effectively target these variants in CRPC [221223]. This resistance mechanism underscores the urgent need for therapeutic strategies capable of targeting AR splice variants or the downstream effectors to overcome treatment failures in CRPC. Recently, the development of degraders ( Figure 5 ) that target AR and AR-V7 [197, 223] has opened promising new avenues for CRPC treatment, offering a broader range of therapeutic options. As these AR-V7-targeting strategies continue to advance, the strategies warrant increased focus and exploration within the PrCa research field because the strategies hold considerable potential for addressing resistance mechanisms.

            In summary, AR amplification, stabilization, splice variants, and other factors have been shown to be critical in maintaining persistent AR signaling in CRPC, underscoring critical pathways that can be exploited for the development of future therapies. A deeper study of these regulatory mechanisms will pave the way for more precise and potent PROTAC strategies, tailored to target these distinct alterations in AR signaling ( Figure 5 ). These advances offer the potential for enhanced therapeutic efficacy and better treatment to patients with PrCa.

            CONFLICTS OF INTEREST

            The authors declared that they have no conflicts of interest.

            REFERENCES

            1. Denmeade SR, Isaacs JT. A History of Prostate Cancer Treatment. Nature Reviews. Cancer. 2002. Vol. 2:389–396

            2. Wang Z, Wu S, Zhu C, Shen J. The Role of Ferroptosis in Esophageal Cancer. Cancer Cell International. 2022. Vol. 22:266

            3. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2022. CA: A Cancer Journal for Clinicians. 2022. Vol. 72:7–33

            4. Gandhi J, Afridi A, Vatsia S, Joshi G, Joshi G, Kaplan SA, et al.. The Molecular Biology of Prostate Cancer: Current Understanding and Clinical Implications. Prostate Cancer and Prostatic Diseases. 2018. Vol. 21:22–36

            5. Kessel K, Seifert R, Weckesser M, Roll W, Humberg V, Schlack K, et al.. Molecular Analysis of Circulating Tumor Cells of Metastatic Castration-resistant Prostate Cancer Patients receiving (177)Lu-PSMA-617 Radioligand Therapy. Theranostics. 2020. Vol. 10:7645–7655

            6. Liang J, Wang L, Poluben L, Nouri M, Arai S, Xie L, et al.. Androgen Receptor Splice Variant 7 Functions Independently of the Full Length Receptor in Prostate Cancer Cells. Cancer Letters. 2021. Vol. 519:172–184

            7. Yap TA, Smith AD, Ferraldeschi R, Al-Lazikani B, Workman P, de Bono JS. Drug Discovery in Advanced Prostate Cancer: Translating Biology into Therapy. Nature Reviews. Drug Discovery. 2016. Vol. 15:699–718

            8. Bekes M, Langley DR, Crews CM. PROTAC Targeted Protein Degraders: the Past is Prologue. Nature Reviews Drug Discovery. 2022. Vol. 21:181–200

            9. He B, Kemppainen JA, Wilson EM. FXXLF and WXXLF Sequences Mediate the NH2-terminal Interaction with the Ligand Binding Domain of the Androgen Receptor. The Journal of Biological Chemistry. 2000. Vol. 275:22986–22994

            10. Loy CJ, Sim KS, Yong EL. Filamin-A Fragment Localizes to the Nucleus to Regulate Androgen Receptor and Coactivator Functions. Proceedings of the National Academy of Sciences of the United States of America. 2003. Vol. 100:4562–4567

            11. He B, Kemppainen JA, Voegel JJ, Gronemeyer H, Wilson EM. Activation Function 2 in the Human Androgen Receptor Ligand Binding Domain Mediates Interdomain Communication with the NH(2)-Terminal Domain. The Journal of Biological Chemistry. 1999. Vol. 274:37219–37225

            12. Veldscholte J, Berrevoets CA, Zegers ND, van der Kwast TH, Grootegoed JA, Mulder E. Hormone-induced Dissociation of the Androgen Receptor-heat-shock Protein Complex: Use of a New Monoclonal Antibody to Distinguish Transformed from Nontransformed Receptors. Biochemistry. 1992. Vol. 31:7422–7430

            13. Ozanne DM, Brady ME, Cook S, Gaughan L, Neal DE, Robson CN. Androgen Receptor Nuclear Translocation is Facilitated by the f-actin Cross-linking Protein Filamin. Molecular Endocrinology. 2000. Vol. 14:1618–1626

            14. Cardozo CP, Michaud C, Ost MC, Fliss AE, Yang E, Patterson C, et al.. C-Terminal Hsp-interacting Protein Slows Androgen Receptor Synthesis and Reduces Its Rate of Degradation. Archives of Biochemistry and Biophysics. 2003. Vol. 410:134–140

            15. Shatkina L, Mink S, Rogatsch H, Klocker H, Langer G, Nestl A, et al.. The Cochaperone Bag-1L Enhances Androgen Receptor Action via Interaction with the NH2-terminal Region of the Receptor. Molecular and Cellular Biology. 2003. Vol. 23:7189–7197

            16. Liao G, Chen LY, Zhang A, Godavarthy A, Xia F, Ghosh JC, et al.. Regulation of Androgen Receptor Activity by the Nuclear Receptor Corepressor SMRT. The Journal of Biological Chemistry. 2003. Vol. 278:5052–5061

            17. Cutress ML, Whitaker HC, Mills IG, Stewart M, Neal DE. Structural Basis for the Nuclear Import of the Human Androgen Receptor. Journal of Cell Science. 2008. Vol. 121:957–968

            18. He B, Lee LW, Minges JT, Wilson EM. Dependence of Selective Gene Activation on the Androgen Receptor NH2- and COOH-terminal Interaction. The Journal of Biological Chemistry. 2002. Vol. 277:25631–25639

            19. Lee YF, Shyr CR, Thin TH, Lin WJ, Chang C. Convergence of Two Repressors Through Heterodimer Formation of Androgen Receptor and Testicular Orphan Receptor-4: A Unique Signaling Pathway in the Steroid Receptor Superfamily. Proceedings of the National Academy of Sciences of the United States of America. 1999. Vol. 96:14724–14729

            20. Rahman M, Miyamoto H, Chang C. Androgen Receptor Coregulators in Prostate Cancer: Mechanisms and Clinical Implications. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2004. Vol. 10:2208–2219

            21. Schaufele F, Carbonell X, Guerbadot M, Borngraeber S, Chapman MS, Ma AA, et al.. The Structural Basis of Androgen Receptor Activation: Intramolecular and Intermolecular Amino-carboxy Interactions. Proceedings of the National Academy of Sciences of the United States of America. 2005. Vol. 102:9802–9807

            22. Zhou ZX, Sar M, Simental JA, Lane MV, Wilson EM. A Ligand-dependent Bipartite Nuclear Targeting Signal in the Human Androgen Receptor. Requirement for the DNA-binding Domain and Modulation by NH2-terminal and Carboxyl-terminal Sequences. The Journal of Biological Chemistry. 1994. Vol. 269:13115–13123

            23. Cao X, Qin J, Xie Y, Khan O, Dowd F, Scofield M, et al.. Regulator of G-protein Signaling 2 (RGS2) Inhibits Androgen-independent Activation of Androgen Receptor in Prostate Cancer Cells. Oncogene. 2006. Vol. 25:3719–3734

            24. Manin M, Baron S, Goossens K, Beaudoin C, Jean C, Veyssiere G, et al.. Androgen Receptor Expression is Regulated by the Phosphoinositide 3-kinase/Akt Pathway in Normal and Tumoral Epithelial Cells. The Biochemical Journal. 2002. Vol. 366:729–736

            25. Scheid MP, Woodgett JR. PKB/AKT: Functional Insights from Genetic Models. Nature Reviews. Molecular Cell Biology. 2001. Vol. 2:760–768

            26. Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C. From HER2/Neu Signal Cascade to Androgen Receptor and Its Coactivators: A Novel Pathway by Induction of Androgen Target Genes Through MAP Kinase in Prostate Cancer Cells. Proceedings of the National Academy of Sciences of the United States of America. 1999. Vol. 96:5458–5463

            27. Gioeli D, Ficarro SB, Kwiek JJ, Aaronson D, Hancock M, Catling AD, et al.. Androgen Receptor Phosphorylation. Regulation and Identification of the Phosphorylation Sites. The Journal of Biological Chemistry. 2002. Vol. 277:29304–29314

            28. Blok LJ, de Ruiter PE, Brinkmann AO. Forskolin-induced Dephosphorylation of the Androgen Receptor Impairs Ligand Binding. Biochemistry. 1998. Vol. 37:3850–3857

            29. Rochette-Egly C. Nuclear Receptors: Integration of Multiple Signalling Pathways Through Phosphorylation. Cellular Signalling. 2003. Vol. 15:355–366

            30. van Royen ME, Cunha SM, Brink MC, Mattern KA, Nigg AL, Dubbink HJ, et al.. Compartmentalization of Androgen Receptor Protein-protein Interactions in Living Cells. The Journal of Cell Biology. 2007. Vol. 177:63–72

            31. Heinlein CA, Chang C. Androgen Receptor (AR) Coregulators: An Overview. Endocrine Reviews. 2002. Vol. 23:175–200

            32. Powell SM, Christiaens V, Voulgaraki D, Waxman J, Claessens F, Bevan CL. Mechanisms of Androgen Receptor Signalling via Steroid Receptor Coactivator-1 in Prostate. Endocrine-Related Cancer. 2004. Vol. 11:117–130

            33. Farla P, Hersmus R, Trapman J, Houtsmuller AB. Antiandrogens Prevent Stable DNA-binding of the Androgen Receptor. Journal of Cell Science. 2005. Vol. 118:4187–4198

            34. Klokk TI, Kurys P, Elbi C, Nagaich AK, Hendarwanto A, Slagsvold T, et al.. Ligand-specific Dynamics of the Androgen Receptor at Its Response Element in Living Cells. Molecular and Cellular Biology. 2007. Vol. 27:1823–1843

            35. Gaughan L, Logan IR, Neal DE, Robson CN. Regulation of Androgen Receptor and Histone Deacetylase 1 by Mdm2-mediated Ubiquitylation. Nucleic Acids Research. 2005. Vol. 33:13–26

            36. Haelens A, Tanner T, Denayer S, Callewaert L, Claessens F. The Hinge Region Regulates DNA Binding, Nuclear Translocation, and Transactivation of the Androgen Receptor. Cancer Research. 2007. Vol. 67:4514–4523

            37. Lee DK, Chang C. Endocrine Mechanisms of Disease: Expression and Degradation of Androgen Receptor: Mechanism and Clinical Implication. The Journal of Clinical Endocrinology and Metabolism. 2003. Vol. 88:4043–4054

            38. Sheflin L, Keegan B, Zhang W, Spaulding SW. Inhibiting Proteasomes in Human HepG2 and LNCaP Cells Increases Endogenous Androgen Receptor Levels. Biochemical and Biophysical Research Communications. 2000. Vol. 276:144–150

            39. Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C. Prostate-specific Membrane Antigen Expression in Normal and Malignant Human Tissues. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 1997. Vol. 3:81–85

            40. Wright GL Jr, Haley C, Beckett ML, Schellhammer PF. Expression of Prostate-specific Membrane Antigen in Normal, Benign, and Malignant Prostate Tissues. Urologic Oncology. 1995. Vol. 1:18–28

            41. Rahn KA, Slusher BS, Kaplin AI. Glutamate in CNS Neurodegeneration and Cognition and Its Regulation by GCPII Inhibition. Current Medicinal Chemistry. 2012. Vol. 19:1335–1345

            42. Rahn KA, Watkins CC, Alt J, Rais R, Stathis M, Grishkan I, et al.. Inhibition of Glutamate Carboxypeptidase II (GCPII) Activity as a Treatment for Cognitive Impairment in Multiple Sclerosis. Proceedings of the National Academy of Sciences of the United States of America. 2012. Vol. 109:20101–20106

            43. Davis MI, Bennett MJ, Thomas LM, Bjorkman PJ. Crystal Structure of Prostate-specific Membrane Antigen, A Tumor Marker and Peptidase. Proceedings of the National Academy of Sciences of the United States of America. 2005. Vol. 102:5981–5986

            44. Schulke N, Varlamova OA, Donovan GP, Ma D, Gardner JP, Morrissey DM, et al.. The Homodimer of Prostate-specific Membrane Antigen is a Functional Target for Cancer Therapy. Proceedings of the National Academy of Sciences of the United States of America. 2003. Vol. 100:12590–12595

            45. Rawlings ND, Barrett AJ. Structure of Membrane Glutamate Carboxypeptidase. Biochimica et Biophysica Acta. 1997. Vol. 1339:247–252

            46. Bernstein LH, Gutstein S, Weiner S, Efron G. The Absorption and Malabsorption of Folic Acid and Its Polyglutamates. The American Journal of Medicine. 1970. Vol. 48:570–579

            47. Zhao R, Diop-Bove N, Visentin M, Goldman ID. Mechanisms of Membrane Transport of Folates into Cells and Across Epithelia. Annual Review of Nutrition. 2011. Vol. 31:177–201

            48. Figueiredo JC, Grau MV, Haile RW, Sandler RS, Summers RW, Bresalier RS, et al.. Folic Acid and Risk of Prostate Cancer: Results from a Randomized Clinical Trial. Journal of the National Cancer Institute. 2009. Vol. 101:432–435

            49. Sanderson SM, Gao X, Dai Z, Locasale JW. Methionine Metabolism in Health and Cancer: A Nexus of Diet and Precision Medicine. Nature Reviews. Cancer. 2019. Vol. 19:625–637

            50. Yao V, Bacich DJ. Prostate Specific Membrane Antigen (PSMA) Expression Gives Prostate Cancer Cells a Growth Advantage in a Physiologically Relevant Folate Environment In Vitro. The Prostate. 2006. Vol. 66:867–875

            51. Corbin JM, Ruiz-Echevarría MJ. One-Carbon Metabolism in Prostate Cancer: The Role of Androgen Signaling. International Journal of Molecular Sciences. 2016. Vol. 17:1208

            52. Zadra G, Ribeiro CF, Chetta P, Ho Y, Cacciatore S, Gao X, et al.. Inhibition of De Novo Lipogenesis Targets Androgen Receptor Signaling in Castration-resistant Prostate Cancer. Proceedings of the National Academy of Sciences of the United States of America. 2019. Vol. 116:631–640

            53. Bakht MK, Beltran H. Biological Determinants of PSMA Expression, Regulation and Heterogeneity in Prostate Cancer. Nature Reviews Urology. 2024. Vol. 22:26–45

            54. Neale JH, Bzdega T, Wroblewska B. N-Acetylaspartylglutamate: The Most Abundant Peptide Neurotransmitter in the Mammalian Central Nervous System. Journal of Neurochemistry. 2000. Vol. 75:443–452

            55. Morland C, Nordengen K. N-Acetyl-Aspartyl-Glutamate in Brain Health and Disease. International Journal of Molecular Sciences. 2022. Vol. 23:1268

            56. Kaittanis C, Andreou C, Hieronymus H, Mao N, Foss CA, Eiber M, et al.. Prostate-specific Membrane Antigen Cleavage of Vitamin B9 Stimulates Oncogenic Signaling Through Metabotropic Glutamate Receptors. The Journal of Experimental Medicine. 2018. Vol. 215:159–175

            57. Palamiuc L, Emerling BM. PSMA Brings New Flavors to PI3K Signaling: A Role for Glutamate in Prostate Cancer. The Journal of Experimental Medicine. 2018. Vol. 215:17–19

            58. Bakht MK, Hayward JJ, Shahbazi-Raz F, Skubal M, Tamura R, Stringer KF, et al.. Identification of Alternative Protein Targets of Glutamate-ureido-lysine Associated with PSMA Tracer Uptake in Prostate Cancer Cells. Proceedings of the National Academy of Sciences of the United States of America. 2022. Vol. 119:e2025710119. [Cross Ref]

            59. Pellegrino S, Fonti R. A Look into the Future: The Role of PSMA Beyond Prostate Cancer. European Journal of Nuclear Medicine and Molecular Imaging. 2023. Vol. 51:278–280

            60. Lee MS, Asomaning K, Su L, Wain JC, Mark EJ, Christiani DC. MTHFR Polymorphisms, Folate Intake and Carcinogen DNA Adducts in the Lung. International Journal of Cancer. 2012. Vol. 131:1203–1209

            61. Yao V, Berkman CE, Choi JK, O’Keefe DS, Bacich DJ. Expression of Prostate-specific Membrane Antigen (PSMA), Increases Cell Folate Uptake and Proliferation and Suggests a Novel Role for PSMA in the Uptake of the Non-polyglutamated Folate, Folic Acid. The Prostate. 2010. Vol. 70:305–316

            62. Asangani IA, Dommeti VL, Wang X, Malik R, Cieslik M, Yang R, et al.. Therapeutic Targeting of BET Bromodomain Proteins in Castration-resistant Prostate Cancer. Nature. 2014. Vol. 510:278–282

            63. Nagarajan S, Hossan T, Alawi M, Najafova Z, Indenbirken D, Bedi U, et al.. Bromodomain Protein BRD4 is Required for Estrogen Receptor-dependent Enhancer Activation and Gene Transcription. Cell Reports. 2014. Vol. 8:460–469

            64. Shi J, Vakoc CR. The Mechanisms Behind the Therapeutic Activity of BET Bromodomain Inhibition. Molecular Cell. 2014. Vol. 54:728–736

            65. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, et al.. BET Bromodomain Inhibition as a Therapeutic Strategy to Target c-Myc. Cell. 2011. Vol. 146:904–917

            66. Wyce A, Degenhardt Y, Bai Y, Le B, Korenchuk S, Crouthame MC, et al.. Inhibition of BET Bromodomain Proteins as a Therapeutic Approach in Prostate Cancer. Oncotarget. 2013. Vol. 4:2419–2429

            67. Urbanucci A, Barfeld SJ, Kytola V, Itkonen HM, Coleman IM, Vodak D, et al.. Androgen Receptor Deregulation Drives Bromodomain-Mediated Chromatin Alterations in Prostate Cancer. Cell Reports. 2017. Vol. 19:2045–2059

            68. Romero Otero J, Garcia Gomez B, Campos Juanatey F, Touijer KA. Prostate Cancer Biomarkers: An Update. Urologic Oncology. 2014. Vol. 32:252–260

            69. Lilja H. Testing New PSA Subforms to Enhance the Accuracy of Predicting Cancer Risk and Disease Outcome in Prostate Cancer. Clinical Chemistry. 2008. Vol. 54:1248–1249

            70. Lilja H, Ulmert D, Vickers AJ. Prostate-specific Antigen and Prostate Cancer: Prediction, Detection and Monitoring. Nature Reviews. Cancer. 2008. Vol. 8:268–278

            71. Cary KC, Cooperberg MR. Biomarkers in Prostate Cancer Surveillance and Screening: Past, Present, and Future. Therapeutic Advances in Urology. 2013. Vol. 5:318–329

            72. Farha MW, Salami SS. Biomarkers for Prostate Cancer Detection and Risk Stratification. Therapeutic Advances in Urology. 2022. Vol. 14:17562872221103988

            73. Sokoll LJ, Sanda MG, Feng Z, Kagan J, Mizrahi IA, Broyles DL, et al.. A Prospective Multicenter, National Cancer Institute Early Detection Research Network study of [-2]proPSA: Improving Prostate Cancer Detection and Correlating with Cancer Aggressiveness. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology. 2010. Vol. 19:1193–1200

            74. Bussemakers MJ, van Bokhoven A, Verhaegh GW, Smit FP, Karthaus HF, Schalken JA, et al.. DD3: A New Prostate-specific Gene, Highly Overexpressed in Prostate Cancer. Cancer Research. 1999. Vol. 59:5975–5979

            75. de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TW, et al.. DD3(PCA3), A Very Sensitive and Specific Marker to Detect Prostate Tumors. Cancer Research. 2002. Vol. 62:2695–2698

            76. Squire JA, Park PC, Yoshimoto M, Alami J, Williams JL, Evans A, et al.. Prostate Cancer as a Model System for Genetic Diversity in Tumors. Advances in Cancer Research. 2011. Vol. 112:183–216

            77. Auprich M, Bjartell A, Chun FK, de la Taille A, Freedland SJ, Haese A, et al.. Contemporary Role of Prostate Cancer Antigen 3 in the Management of Prostate Cancer. European Urology. 2011. Vol. 60:1045–1054

            78. Schoenborn JR, Nelson P, Fang M. Genomic Profiling Defines Subtypes of Prostate Cancer with the Potential for Therapeutic Stratification. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2013. Vol. 19:4058–4066

            79. Crawford ED, Rove KO, Trabulsi EJ, Qian J, Drewnowska KP, Kaminetsky JC, et al.. Diagnostic Performance of PCA3 to Detect Prostate Cancer in Men with Increased Prostate Specific Antigen: A Prospective Study of 1,962 Cases. The Journal of Urology. 2012. Vol. 188:1726–1731

            80. Vlaeminck-Guillem V, Ruffion A, Andre J, Devonec M, Paparel P. Urinary Prostate Cancer 3 Test: Toward the Age of Reason? Urology. 2010. Vol. 75:447–453

            81. Kupelian PA, Buchsbaum JC, Elshaikh M, Reddy CA, Zippe C, Klein EA. Factors Affecting Recurrence Rates after Prostatectomy or Radiotherapy in Localized Prostate Carcinoma Patients with Biopsy Gleason Score 8 or Above. Cancer. 2002. Vol. 95:2302–2307

            82. Sekhoacha M, Riet K, Motloung P, Gumenku L, Adegoke A, Mashele S. Prostate Cancer Review: Genetics, Diagnosis, Treatment Options, and Alternative Approaches. Molecules. 2022. Vol. 27:5730

            83. Parker CC, James ND, Brawley CD, Clarke NW, Hoyle AP, Ali A, et al.. Radiotherapy to the Primary Tumour for Newly Diagnosed, Metastatic Prostate Cancer (STAMPEDE): A Randomised Controlled Phase 3 Trial. Lancet. 2018. Vol. 392:2353–2366

            84. Sweeney CJ, Chen YH, Carducci M, Liu G, Jarrard DF, Eisenberger M, et al.. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. The New England Journal of Medicine. 2015. Vol. 373:737–746

            85. Grewal K, Grewal K, Tabbara IA. PARP Inhibitors in Prostate Cancer. Anticancer Research. 2021. Vol. 41:551–556

            86. Urbanucci A, Mills IG. Bromodomain-containing Proteins in Prostate Cancer. Molecular and Cellular Endocrinology. 2018. Vol. 462:31–40

            87. Jones K, Zhang Y, Kong Y, Farah E, Wang R, Li C, et al.. Epigenetics in Prostate Cancer Treatment. Journal of Translational Genetics and Genomics. 2021. Vol. 5:341–356

            88. Yan Y, Ma J, Wang D, Lin D, Pang X, Wang S, et al.. The Novel BET-CBP/p300 Dual Inhibitor NEO2734 is Active in SPOP Mutant and Wild-type Prostate Cancer. MBO Molecular Medicine. 2019. Vol. 11:e10659

            89. He Y, Wei T, Ye Z, Orme JJ, Lin D, Sheng H, et al.. A Noncanonical AR Addiction Drives Enzalutamide Resistance in Prostate Cancer. Nature Communications. 2021. Vol. 12:1521

            90. Sun R, Yan B, Li H, Ding D, Wang L, Pang J, et al.. Androgen Receptor Variants Confer Castration Resistance in Prostate Cancer by Counteracting Antiandrogen-Induced Ferroptosis. Cancer Research. 2023. Vol. 83:3192–3204

            91. Paller CJ, Piana D, Eshleman JR, Riel S, Denmeade SR, Isaacsson Velho P, et al.. A Pilot Study of Prostate-specific Membrane Antigen (PSMA) Dynamics in Men Undergoing Treatment for Advanced Prostate Cancer. The Prostate. 2019. Vol. 79:1597–1603

            92. Sartor O, de Bono J, Chi KN, Fizazi K, Herrmann K, Rahbar K, et al.. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. The New England Journal Medicine. 2021. Vol. 385:1091–1103

            93. Pathmanandavel S, Crumbaker M, Yam AO, Nguyen A, Rofe C, Hovey E, et al.. 177Lu-PSMA-617 and Idronoxil in Men with End-Stage Metastatic Castration-Resistant Prostate Cancer (LuPIN): Patient Outcomes and Predictors of Treatment Response in a Phase I/II Trial. Journal of Nuclear Medicine: Official Publication, Society of Nuclear Medicine. 2022. Vol. 63:560–566

            94. Aggarwal R, Starzinski S, de Kouchkovsky I, Koshkin V, Bose R, Chou J, et al.. Single-dose 177Lu-PSMA-617 Followed by Maintenance Pembrolizumab in Patients with Metastatic Castration-resistant Prostate Cancer: An Open-label, Dose-expansion, Phase 1 Trial. The Lancet. Oncology. 2023. Vol. 24:1266–1276

            95. Ramnaraign B, Sartor O. PSMA-Targeted Radiopharmaceuticals in Prostate Cancer: Current Data and New Trials. The Oncologist. 2023. Vol. 28:392–401

            96. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. American Journal of Clinical Oncology. 2016. Vol. 39:98–106

            97. Boudadi K, Suzman DL, Anagnostou V, Fu W, Luber B, Wang H, et al.. Ipilimumab Plus Nivolumab and DNA-repair Defects in AR-V7-expressing Metastatic Prostate Cancer. Oncotarget. 2018. Vol. 9:28561–28571

            98. Shenderov E, Boudadi K, Fu W, Wang H, Sullivan R, Jordan A, et al.. Nivolumab Plus Ipilimumab, With or Without Enzalutamide, in AR-V7-expressing Metastatic Castration-Resistant Prostate Cancer: A Phase-2 Nonrandomized Clinical Trial. The Prostate. 2021. Vol. 81:326–338

            99. Sharma P, Pachynski RK, Narayan V, Flechon A, Gravis G, Galsky MD, et al.. Nivolumab Plus Ipilimumab for Metastatic Castration-Resistant Prostate Cancer: Preliminary Analysis of Patients in the CheckMate 650 Trial. Cancer Cell. 2020. Vol. 38:489–499.e3

            100. Kyriakopoulos CE, Eickhoff JC, Ferrari AC, Schweizer MT, Wargowski E, Olson BM, et al.. Multicenter Phase I Trial of a DNA Vaccine Encoding the Androgen Receptor Ligand-binding Domain (pTVG-AR, MVI-118) in Patients with Metastatic Prostate Cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2020. Vol. 26:5162–5171

            101. Antonarakis ES. Current Understanding of Resistance to Abiraterone and Enzalutamide in Advanced Prostate Cancer. Clinical Advances in Hematology & Oncology. 2016. Vol. 14:316–319

            102. La J, Wang L, Corrigan JK, Lang D, Lee MH, Do NV, et al.. Abiraterone or Enzalutamide for Patients With Metastatic Castration-Resistant Prostate Cancer. JAMA Network Open. 2024. Vol. 7:e2428444

            103. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al.. Enzalutamide in Metastatic Prostate Cancer Before Chemotherapy. The New England Journal of Medicine. 2014. Vol. 371:424–433

            104. Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PF, Sternberg CN, et al.. Abiraterone Acetate Plus Prednisone Versus Placebo Plus Prednisone in Chemotherapy-naive Men with Metastatic Castration-resistant Prostate Cancer (COU-AA-302): Final Overall Survival Analysis of a Randomised, Double-blind, Placebo-controlled Phase 3 Study. The Lancet. Oncology. 2015. Vol. 16:152–160

            105. Quinn DI, Sandler HM, Horvath LG, Goldkorn A, Eastham JA. The Evolution of Chemotherapy for the Treatment of Prostate Cancer. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2017. Vol. 28:2658–2669

            106. Doyle-Lindrud S. Managing Side Effects of the Novel Taxane Cabazitaxel in Castrate-resistant Prostate Cancer. Clinical Journal of Oncology Nursing. 2012. Vol. 16:286–291

            107. Yedla P, Babalghith AO, Andra VV, Syed R. PROTACs in the Management of Prostate Cancer. Molecules. 2023. Vol. 28:3698

            108. Cooper G, Adams K. The Cell: A Molecular Approach. Oxford University Press. 2022

            109. Ahn G, Banik SM, Miller CL, Riley NM, Cochran JR, Bertozzi CR. LYTACs That Engage the Asialoglycoprotein Receptor for Targeted Protein Degradation. Nature Chemical Biology. 2021. Vol. 17:937–946

            110. Banik SM, Pedram K, Wisnovsky S, Ahn G, Riley NM, Bertozzi CR. Lysosome-targeting Chimaeras for Degradation of Extracellular Proteins. Nature. 2020. Vol. 584:291–297

            111. Marei H, Tsai WK, Kee YS, Ruiz K, He J, Cox C, et al.. Antibody Targeting of E3 Ubiquitin Ligases for Receptor Degradation. Nature. 2022. Vol. 610:182–189

            112. Pance K, Gramespacher JA, Byrnes JR, Salangsang F, Serrano JC, Cotton AD, et al.. Modular Cytokine Receptor-targeting Chimeras for Targeted Degradation of Cell Surface and Extracellular Proteins. Nature Biotechnology. 2023. Vol. 41:273–281

            113. Zhang D, Duque-Jimenez J, Facchinetti F, Brixi G, Rhee K, Feng WW, et al.. Transferrin Receptor Targeting Chimeras for Membrane Protein Degradation. Nature. 2024. [Cross Ref]

            114. Hershko A, Ciechanover A. The Ubiquitin System. Annual Review of Biochemistry. 1998. Vol. 67:425–479

            115. Komander D, Rape M. The Ubiquitin Code. Annual Review of Biochemistry. 2012. Vol. 81:203–229

            116. Vogel G. Nobel Prizes. Gold Medal from Cellular Trash. Science. 2004. Vol. 306:400–401

            117. Kanayama A, Seth RB, Sun L, Ea CK, Hong M, Shaito A, et al.. TAB2 and TAB3 Activate the NF-kappaB Pathway Through Binding to Polyubiquitin Chains. Molecular Cell. 2004. Vol. 15:535–548

            118. Dang F, Nie L, Wei W. Ubiquitin Signaling in Cell Cycle Control and Tumorigenesis. Cell Death & Differentiation. 2021. Vol. 28:427–438

            119. Mevissen TET, Komander D. Mechanisms of Deubiquitinase Specificity and Regulation. Annual Review of Biochemistry. 2017. Vol. 86:159–192

            120. Guharoy M, Bhowmick P, Sallam M, Tompa P. Tripartite Degrons Confer Diversity and Specificity on Regulated Protein Degradation in the Ubiquitin-proteasome System. Nature Communication. 2016. Vol. 7:10239

            121. Liu J, Tokheim C, Lee JD, Gan W, North BJ, Liu XS, et al.. Genetic Fusions Favor Tumorigenesis Through Degron Loss in Oncogenes. Nature Communication. 2021. Vol. 12:6704

            122. Liu J, Ma J, Liu Y, Xia J, Li Y, Wang ZP, et al.. PROTACs: A Novel Strategy for Cancer Therapy. Seminars in Cancer Biology. 2020. Vol. 67:171–179

            123. Takahashi D, Arimoto H. Targeting Selective Autophagy by AUTAC Degraders. Autophagy. 2020. Vol. 16:765–766

            124. Takahashi D, Moriyama J, Nakamura T, Miki E, Takahashi E, Sato A, et al.. AUTACs: Cargo-Specific Degraders Using Selective Autophagy. Molecular Cell. 2019. Vol. 76:797–810e10

            125. Li Z, Wang C, Wang Z, Zhu C, Li J, Sha T, et al.. Allele-selective lowering of mutant HTT protein by HTT-LC3 linker compounds. Nature. 2019. Vol. 575:203–209

            126. Li Z, Zhu C, Ding Y, Fei Y, Lu B. ATTEC: A Potential New Approach to Target Proteinopathies. Autophagy. 2020. Vol. 16:185–187

            127. Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ. Protacs: Chimeric Molecules that Target Proteins to the Skp1-Cullin-F Box Complex for Ubiquitination and Degradation. Proceedings of the National Academy of Sciences of the United States of America. 2001. Vol. 98:8554–8559

            128. Bondeson DP, Mares A, Smith IE, Ko E, Campos S, Miah AH, et al.. Catalytic In Vivo Protein Knockdown by Small-molecule PROTACs. Nature Chemical Biology. 2015. Vol. 11:611–617

            129. Schneekloth AR, Pucheault M, Tae HS, Crews CM. Targeted Intracellular Protein Degradation Induced by a Small Molecule: En Route to Chemical Proteomics. Bioorganic & Medicinal Chemistry Letters. 2008. Vol. 18:5904–5908

            130. Winter GE, Buckley DL, Paulk J, Roberts JM, Souza A, Dhe-Paganon S, et al.. DRUG DEVELOPMENT. Phthalimide Conjugation as a Strategy for In Vivo Target Protein Degradation. Science. 2015. Vol. 348:1376–1381

            131. Sekine K, Takubo K, Kikuchi R, Nishimoto M, Kitagawa M, Abe F, et al.. Small Molecules Destabilize cIAP1 by Activating Auto-ubiquitylation. The Journal of Biological Chemistry. 2008. Vol. 283:8961–8968

            132. Lee J, Lee Y, Jung YM, Park JH, Yoo HS, Park J. Discovery of E3 Ligase Ligands for Target Protein Degradation. Molecules. 2022. Vol. 27:6515

            133. Bricelj A, Steinebach C, Kuchta R, Gütschow M, Sosič I. E3 Ligase Ligands in Successful PROTACs: An Overview of Syntheses and Linker Attachment Points. Frontiers in Chemistry. 2021. Vol. 9:707317

            134. Weng G, Shen C, Cao D, Gao J, Dong X, He Q, et al.. PROTAC-DB: An Online Database of PROTACs. Nucleic Acids Research. 2021. Vol. 49:D1381–D1387

            135. He S, Dong G, Cheng J, Wu Y, Sheng C. Strategies for Designing Proteolysis Targeting Chimaeras (PROTACs). Medicinal Research Reviews. 2022. Vol. 42:1280–1342

            136. Troup RI, Fallan C, Baud MGJ. Current Strategies for the Design of PROTAC Linkers: A Critical Review. Exploration of Targeted Anti-tumor Therapy. 2020. Vol. 1:273–312

            137. Han X, Wang C, Qin C, Xiang W, Fernandez-Salas E, Yang CY, et al.. Discovery of ARD-69 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Androgen Receptor (AR) for the Treatment of Prostate Cancer. Journal of Medicinal Chemistry. 2019. Vol. 62:941–964

            138. Morgenthaler M, Schweizer E, Hoffmann-Roder A, Benini F, Martin RE, Jaeschke G, et al.. Predicting and Tuning Physicochemical Properties in Lead Optimization: Amine Basicities. ChemMedChem. 2007. Vol. 2:1100–1115

            139. Wiberg KB. Substituent Effects on the Acidity of Weak Acids. 1. Bicyclo[2.2.2]octane-1-carboxylic Acids and bicyclo[1.1.1]pentane-1-carboxylic Acids. Journal of Organic Chemistry. 2002. Vol. 67:1613–1617

            140. Davis C, Spaller BL, Matouschek A. Mechanisms of Substrate Recognition by the 26S Proteasome. Current Opinion in Structural Biology. 2021. Vol. 67:161–169

            141. Eyre TA, Riches JC. The Evolution of Therapies Targeting Bruton Tyrosine Kinase for the Treatment of Chronic Lymphocytic Leukaemia: Future Perspectives. Cancers (Basel). 2023. Vol. 15:2596

            142. Ackerman L, Acloque G, Bacchelli S, Schwartz H, Feinstein BJ, La Stella P, et al.. IRAK4 Degrader in Hidradenitis Suppurativa and Atopic Dermatitis: A Phase 1 Trial. Nature Medicine. 2023. Vol. 29:3127–3136

            143. Dobrovolsky D, Wang ES, Morrow S, Leahy C, Faust T, Nowak RP, et al.. Bruton Tyrosine Kinase Degradation as a Therapeutic Strategy for Cancer. Blood. 2019. Vol. 133:952–961

            144. Zarrin AA, Bao K, Lupardus P, Vucic D. Kinase Inhibition in Autoimmunity and Inflammation. Nature Reviews. Drug Discovery. 2021. Vol. 20:39–63

            145. Schiemer J, Horst R, Meng Y, Montgomery JI, Xu Y, Feng X, et al.. Snapshots and Ensembles of BTK and cIAP1 Protein Degrader Ternary Complexes. Natue Chemical Biology. 2021. Vol. 17:152–160

            146. Zorba A, Nguyen C, Xu Y, Starr J, Borzilleri K, Smith J, et al.. Delineating the Role of Cooperativity in the Design of Potent PROTACs for BTK. Proceedings of the National Academy of Sciences of the United States of America. 2018. Vol. 115:E7285–E7292

            147. Tinworth CP, Lithgow H, Dittus L, Bassi ZI, Hughes SE, Muelbaier M, et al.. PROTAC-Mediated Degradation of Bruton’s Tyrosine Kinase Is Inhibited by Covalent Binding. ACS Chemical Biology. 2019. Vol. 14:342–347

            148. Buhimschi AD, Armstrong HA, Toure M, Jaime-Figueroa S, Chen TL, Lehman AM, et al.. Targeting the C481S Ibrutinib-Resistance Mutation in Bruton’s Tyrosine Kinase Using PROTAC-Mediated Degradation. Biochemistry. 2018. Vol. 57:3564–3575

            149. Sun Y, Zhao X, Ding N, Gao H, Wu Y, Yang Y, et al.. PROTAC-induced BTK Degradation as a Novel Therapy for Mutated BTK C481S Induced Ibrutinib-resistant B-cell Malignancies. Cell Research. 2018. Vol. 28:779–781

            150. Goedert M. Tau Protein and Neurodegeneration. Seminars in Cell & Developmental Biology. 2004. Vol. 15:45–49

            151. Chang CW, Shao E, Mucke L. Tau: Enabler of Diverse Brain Disorders and Target of Rapidly Evolving Therapeutic Strategies. Science. 2021. Vol. 371:eabb8255

            152. DeVos SL, Miller RL, Schoch KM, Holmes BB, Kebodeaux CS, Wegener AJ, et al.. Tau Reduction Prevents Neuronal Loss and Reverses Pathological Tau Deposition and Seeding in Mice with Tauopathy. Science Translational Medicine. 2017. Vol. 9:eaag0481

            153. Wegmann S, DeVos SL, Zeitler B, Marlen K, Bennett RE, Perez-Rando M, et al.. Persistent Repression of Tau in the Brain Using Engineered Zinc Finger Protein Transcription Factors. Science Advances. 2021. Vol. 7:eabe1611

            154. Lu M, Liu T, Jiao Q, Ji J, Tao M, Liu Y, et al.. Discovery of a Keap1-dependent Peptide PROTAC to Knockdown Tau by Ubiquitination-proteasome Degradation Pathway. European Journal of Medicinal Chemistry. 2018. Vol. 146:251–259

            155. Wang W, Zhou Q, Jiang T, Li S, Ye J, Zheng J, et al.. A Novel Small-molecule PROTAC Selectively Promotes Tau Clearance to Improve Cognitive Functions in Alzheimer-like Models. Theranostics. 2021. Vol. 11:5279–5295

            156. Chien DT, Szardenings AK, Bahri S, Walsh JC, Mu F, Xia C, et al.. Early Clinical PET Imaging Results with the Novel PHF-tau Radioligand [F18]-T808. Journal of Alzheimer’s Disease. 2014. Vol. 38:171–184

            157. Silva MC, Ferguson FM, Cai Q, Donovan KA, Nandi G, Patnaik D, et al.. Targeted Degradation of Aberrant Tau in Frontotemporal Dementia Patient-derived Neuronal Cell Models. Elife. 2019. Vol. 8:e45457

            158. Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy Body Pathology in Alzheimer’s Disease. Journal of Molecular Neuroscience. 2001. Vol. 17:225–232

            159. Qu J, Ren X, Xue F, He Y, Zhang R, Zheng Y, et al.. Specific Knockdown of α-Synuclein by Peptide-Directed Proteasome Degradation Rescued Its Associated Neurotoxicity. Cell Chemical Biology. 2020. Vol. 27:763

            160. A Novel Gene Containing a Trinucleotide Repeat that is Expanded and Unstable on Huntington’s Disease Chromosomes. The Huntington’s Disease Collaborative Research Group. Cell. 1993. Vol. 72:971–983

            161. Sap KA, Reits EA. Strategies to Investigate Ubiquitination in Huntington’s Disease. Frontiers in Chemistry. 2020. Vol. 8:485

            162. Tomoshige S, Nomura S, Ohgane K, Hashimoto Y, Ishikawa M. Discovery of Small Molecules that Induce the Degradation of Huntingtin. Angewandte Chemie (International ed. in English). 2017. Vol. 56:11530–11533

            163. Li W, Deng X, Chen J. RNA-binding Proteins in Regulating mRNA Stability and Translation: Roles and Mechanisms in Cancer. Seminars in Cancer Biology. 2022. Vol. 86:664–677

            164. Kelaini S, Chan C, Cornelius VA, Margariti A. RNA-Binding Proteins Hold Key Roles in Function, Dysfunction, and Disease. Biology (Basel). 2021. Vol. 10:366

            165. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM. A Census of Human Transcription Factors: Function, Expression and Evolution. Nature Reviews. Genetics. 2009. Vol. 10:252–263

            166. Hagenbuchner J, Ausserlechner MJ. Targeting Transcription Factors by Small Compounds--Current Strategies and Future Implications. Biochemical Pharmacology. 2016. Vol. 107:1–13

            167. Henley MJ, Koehler AN. Advances in Targeting ‘Undruggable’ Transcription Factors with Small Molecules. Nature Reviews. Drug Discovery. 2021. Vol. 20:669–688

            168. Samarasinghe KTG, Jaime-Figueroa S, Burgess M, Nalawansha DA, Dai K, Hu Z, et al.. Targeted Degradation of Transcription Factors by TRAFTACs: TRAnscription Factor TArgeting Chimeras. Cell Chemical Biology. 2021. Vol. 28:648–661.e5

            169. Liu J, Chen H, Kaniskan HU, Xie L, Chen X, Jin J, et al.. TF-PROTACs Enable Targeted Degradation of Transcription Factors. Journal of the American Chemical Society. 2021. Vol. 143:8902–8910

            170. Shao J, Yan Y, Ding D, Wang D, He Y, Pan Y, et al.. Destruction of DNA-Binding Proteins by Programmable Oligonucleotide PROTAC (O’PROTAC): Effective Targeting of LEF1 and ERG. Advanced Science (Weinheim, Baden-Wurttemberg, Germany). 2021. Vol. 8:e2102555

            171. Patil KM, Chin D, Seah HL, Shi Q, Lim KW, Phan AT. G4-PROTAC: Targeted Degradation of a G-quadruplex Binding Protein. Chemical Communications. 2021. Vol. 57:12816–12819

            172. Wang Z, Liu J, Chen H, Qiu X, Xie L, Kaniskan HU, et al.. Telomere Targeting Chimera Enables Targeted Destruction of Telomeric Repeat-Binding Factor Proteins. Journal of the American Chemical Society. 2023. Vol. 145:10872–10879

            173. Li X, Zhang Z, Gao F, Ma Y, Wei D, Lu Z, et al.. c-Myc-Targeting PROTAC Based on a TNA-DNA Bivalent Binder for Combination Therapy of Triple-Negative Breast Cancer. Journal of the American Chemical Society. 2023. Vol. 145:9334–9342

            174. Wang Z, Liu J, Qiu X, Zhang D, Inuzuka H, Chen L, et al.. Methylated Nucleotide-Based Proteolysis-Targeting Chimera Enables Targeted Degradation of Methyl-CpG-Binding Protein 2. Journal of the American Chemical Society. 2023. Vol. 145:21871–21878

            175. Wang Z, Zhang D, Qiu X, Inuzuka H, Xiong Y, Liu J, et al.. Structurally Specific Z-DNA Proteolysis Targeting Chimera Enables Targeted Degradation of Adenosine Deaminase Acting on RNA 1. Journal of the American Chemical Society. 2024. Vol. 146:7584–7593

            176. Xue G, Wang K, Zhou D, Zhong H, Pan Z. Light-Induced Protein Degradation with Photocaged PROTACs. Journal of the American Chemical Society. 2019. Vol. 141:18370–18374

            177. Liu J, Chen H, Ma L, He Z, Wang D, Liu Y, et al.. Light-induced Control of Protein Destruction by Opto-PROTAC. Science Advances. 2020. Vol. 6:eaay5154

            178. Li Z, Ma S, Yang X, Zhang L, Liang D, Dong G, et al.. Development of Photocontrolled BRD4 PROTACs for Tongue Squamous Cell Carcinoma (TSCC). Europena Journal of Medicinal Chemistry. 2021. Vol. 222:113608

            179. Naro Y, Darrah K, Deiters A. Optical Control of Small Molecule-Induced Protein Degradation. Journal of the American Chemical Society. 2020. Vol. 142:2193–2197

            180. Kounde CS, Shchepinova MM, Saunders CN, Muelbaier M, Rackham MD, Harling JD, et al.. A Caged E3 Ligase Ligand for PROTAC-mediated Protein Degradation with Light. Chemical Communications (Cambridge, England). 2020. Vol. 56:5532–5535

            181. Pfaff P, Samarasinghe KTG, Crews CM, Carreira EM. Reversible Spatiotemporal Control of Induced Protein Degradation by Bistable PhotoPROTACs. ACS Central Science. 2019. Vol. 5:1682–1690

            182. Liu J, Chen H, Liu Y, Shen Y, Meng F, Kaniskan HU, et al.. Cancer Selective Target Degradation by Folate-Caged PROTACs. Journal of the American Chemical Society. 2021. Vol. 143:7380–7387

            183. Chen H, Liu J, Kaniskan HU, Wei W, Jin J. Folate-Guided Protein Degradation by Immunomodulatory Imide Drug-Based Molecular Glues and Proteolysis Targeting Chimeras. Journal of Medicinal Chemistry. 2021. Vol. 64:12273–12285

            184. Cheng W, Li S, Wen X, Han S, Wang S, Wei H, et al.. Development of Hypoxia-activated PROTAC Exerting a More Potent Effect in Tumor Hypoxia than in Normoxia. Chemical Communications. 2021. Vol. 57:12852–12855

            185. Shi S, Du Y, Zou Y, Niu J, Cai Z, Wang X, et al.. Rational Design for Nitroreductase (NTR)-Responsive Proteolysis Targeting Chimeras (PROTACs) Selectively Targeting Tumor Tissues. Journal of Medicinal Chemistry. 2022. Vol. 65:5057–5071

            186. Yang C, Yang Y, Li Y, Ni Q, Li J. Radiotherapy-Triggered Proteolysis Targeting Chimera Prodrug Activation in Tumors. Journal of the American Chemical Society. 2023. Vol. 145:385–391

            187. An K, Deng X, Chi H, Zhang Y, Li Y, Cheng M, et al.. Stimuli-Responsive PROTACs for Controlled Protein Degradation. Angewandte Chemie (International ed. in English). 2023. Vol. 62:e202306824

            188. Gockel LM, Pfeifer V, Baltes F, Bachmaier RD, Wagner KG, Bendas G, et al.. Design, Synthesis, and Characterization of PROTACs Targeting the Androgen Receptor in Prostate and Lung Cancer Models. Archiv der Pharmazie (Weinheim). 2022. Vol. 355:e2100467

            189. Takwale AD, Jo SH, Jeon YU, Kim HS, Shin CH, Lee HK, et al.. Design and Characterization of Cereblon-mediated Androgen Receptor Proteolysis-targeting Chimeras. European Journal of Medicinal Chemistry. 2020. Vol. 208:112769

            190. Liang JJ, Xie H, Yang RH, Wang N, Zheng ZJ, Zhou C, et al.. Designed, Synthesized and Biological Evaluation of Proteolysis Targeting Chimeras (PROTACs) as AR Degraders for Prostate Cancer Treatment. Bioorganic & Medicinal Chemistry. 2021. Vol. 45:116331

            191. Kim GY, Song CW, Yang YS, Lee NR, Yoo HS, Son SH, et al.. Chemical Degradation of Androgen Receptor (AR) Using Bicalutamide Analog-Thalidomide PROTACs. Molecules. 2021. Vol. 26:2525

            192. Han X, Zhao L, Xiang W, Qin C, Miao B, McEachern D, et al.. Strategies Toward Discovery of Potent and Orally Bioavailable Proteolysis Targeting Chimera Degraders of Androgen Receptor for the Treatment of Prostate Cancer. Journal of Medicinal Chemistry. 2021. Vol. 64:12831–12854

            193. Xiang W, Zhao L, Han X, Qin C, Miao B, McEachern D, et al.. Discovery of ARD-2585 as an Exceptionally Potent and Orally Active PROTAC Degrader of Androgen Receptor for the Treatment of Advanced Prostate Cancer. Journal of Medicinal Chemistry. 2021. Vol. 64:13487–13509

            194. Han X, Zhao L, Xiang W, Qin C, Miao B, Xu T, et al.. Discovery of Highly Potent and Efficient PROTAC Degraders of Androgen Receptor (AR) by Employing Weak Binding Affinity VHL E3 Ligase Ligands. Journal of Medicinal Chemistry. 2019. Vol. 62:11218–11231

            195. Chen L, Han L, Mao S, Xu P, Xu X, Zhao R, et al.. Discovery of A031 as Effective Proteolysis Targeting Chimera (PROTAC) Androgen Receptor (AR) Degrader for the Treatment of Prostate Cancer. European Journal of Medicinal Chemistry. 2021. Vol. 216:113307

            196. Munoz E, Chen G, Hossain A, Wu S, Oceguera Nava E, Hang J, et al.. Synthesis and Biological Evaluation of Niclosamide PROTACs. Bioorganic & Medicinal Chemistry Letters. 2022. Vol. 72:128870

            197. Lee GT, Nagaya N, Desantis J, Madura K, Sabaawy HE, Kim WJ, et al.. Effects of MTX-23, A Novel PROTAC of Androgen Receptor Splice Variant-7 and Androgen Receptor, on CRPC Resistant to Second-Line Antiandrogen Therapy. Molecular Cancer Therapeutics. 2021. Vol. 20:490–499

            198. Lotem J, Sachs L. Genetic Dissection of the Control of Normal Differentiation in Myeloid Leukemic Cells. Proceedings of the National Academy of Sciences of the United States of America. 1977. Vol. 74:5554–5558

            199. Kregel S, Wang C, Han X, Xiao L, Fernandez-Salas E, Bawa P, et al.. Androgen Receptor Degraders Overcome Common Resistance Mechanisms Developed During Prostate Cancer Treatment. Neoplasia. 2020. Vol. 22:111–119

            200. Zhang B, Liu C, Yang Z, Zhang S, Hu X, Li B, et al.. Discovery of BWA-522, a First-in-Class and Orally Bioavailable PROTAC Degrader of the Androgen Receptor Targeting N-Terminal Domain for the Treatment of Prostate Cancer. Journal of Medicinal Chemistry. 2023. Vol. 66:11158–11186

            201. Han X, Zhao L, Xiang W, Miao B, Qin C, Wang M, et al.. Discovery of ARD-2051 as a Potent and Orally Efficacious Proteolysis Targeting Chimera (PROTAC) Degrader of Androgen Receptor for the Treatment of Advanced Prostate Cancer. Journal of Medicinal Chemistry. 2023. Vol. 66:8822–8843

            202. Xiang W, Zhao L, Han X, Xu T, Kregel S, Wang M, et al.. Discovery of ARD-1676 as a Highly Potent and Orally Efficacious AR PROTAC Degrader with a Broad Activity against AR Mutants for the Treatment of AR + Human Prostate Cancer. Journal of Medicinal Chemistry. 2023. Vol. 66:13280–13303

            203. Ma L, Wang J, Zhang Y, Fang F, Ling J, Chu X, et al.. BRD4 PROTAC Degrader MZ1 Exerts Anticancer Effects in Acute Myeloid Leukemia by Targeting c-Myc and ANP32B Genes. Cancer Biology & Therapy. 2022. Vol. 23:1–15

            204. Raina K, Lu J, Qian Y, Altieri M, Gordon D, Rossi AM, et al.. PROTAC-induced BET Protein Degradation as a Therapy for Castration-resistant Prostate Cancer. Proceedings of the National Academy of Sciences of the United States of America. 2016. Vol. 113:7124–7129

            205. Slabicki M, Yoon H, Koeppel J, Nitsch L, Roy Burman SS, Di Genua C, et al.. Small-molecule-induced Polymerization Triggers Degradation of BCL6. Nature. 2020. Vol. 588:164–168

            206. Cromm PM, Samarasinghe KTG, Hines J, Crews CM. Addressing Kinase-Independent Functions of Fak via PROTAC-Mediated Degradation. Journal of the American Chemical Society. 2018. Vol. 140:17019–17026

            207. Jiang B, Wang ES, Donovan KA, Liang Y, Fischer ES, Zhang T, et al.. Development of Dual and Selective Degraders of Cyclin-Dependent Kinases 4 and 6. Angewandte Chemie (International ed. in English). 2019. Vol. 58:6321–6326

            208. Wang ZQ, Zhang ZC, Wu YY, Pi YN, Lou SH, Liu TB, et al.. Bromodomain and Extraterminal (BET) Proteins: Biological Functions, Diseases, and Targeted Therapy. Signal Transduction and Targeted Therapy. 2023. Vol. 8:420

            209. Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: From Discovery to Therapy. Cancer Discovery. 2016. Vol. 6:353–367

            210. Visakorpi T, Hyytinen E, Koivisto P, Tanner M, Keinanen R, Palmberg C, et al.. In Vivo Amplification of the Androgen Receptor Gene and Progression of Human Prostate Cancer. Nature Genetics. 1995. Vol. 9:401–406

            211. Bubendorf L, Kononen J, Koivisto P, Schraml P, Moch H, Gasser TC, et al.. Survey of Gene Amplifications During Prostate Cancer Progression by High-throughout Fluorescence In Situ Hybridization on Tissue Microarrays. Cancer Research. 1999. Vol. 59:803–806

            212. Linja MJ, Savinainen KJ, Saramaki OR, Tammela TL, Vessella RL, Visakorpi T. Amplification and Overexpression of Androgen Receptor Gene in Hormone-refractory Prostate Cancer. Cancer Research. 2001. Vol. 61:3550–3555

            213. Wadosky KM, Koochekpour S. Molecular Mechanisms Underlying Resistance to Androgen Deprivation Therapy in Prostate Cancer. Oncotarget. 2016. Vol. 7:64447–64470

            214. Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, et al.. Molecular Determinants of Resistance to Antiandrogen Therapy. Nature Medicine. 2004. Vol. 10:33–39

            215. van der Steen T, Tindall DJ, Huang H. Posttranslational modification of the androgen receptor in prostate cancer. International Journal of Molecular Sciences. 2013. Vol. 14:14833–14859

            216. Azad AA, Zoubeidi A, Gleave ME, Chi KN. Targeting Heat Shock Proteins in Metastatic Castration-resistant Prostate Cancer. Nature Reviews Urology. 2015. Vol. 12:26–36

            217. Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, et al.. Ligand-independent Androgen Receptor Variants Derived from Splicing of Cryptic Exons Signify Hormone-refractory Prostate Cancer. Cancer Research. 2009. Vol. 69:16–22

            218. Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ. Splicing of a Novel Androgen Receptor Exon Generates a Constitutively Active Androgen Receptor that Mediates Prostate Cancer Therapy Resistance. Cancer Research. 2008. Vol. 68:5469–5477

            219. Nakazawa M, Antonarakis ES, Luo J. Androgen Receptor Splice Variants in the Era of Enzalutamide and Abiraterone. Hormones & Cancer. 2014. Vol. 5:265–273

            220. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al.. AR-V7 and Resistance to Enzalutamide and Abiraterone in Prostate Cancer. The New England Journal of Medicine. 2014. Vol. 371:1028–1038

            221. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al.. Increased Survival with Enzalutamide in Prostate Cancer after Chemotherapy. The New England Journal of Medicine. 2012. Vol. 367:1187–1197

            222. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al.. Abiraterone and Increased Survival in Metastatic Prostate Cancer. The New England Journal of Medicine. 2011. Vol. 364:1995–2005

            223. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al.. Abiraterone in Metastatic Prostate Cancer Without Previous Chemotherapy. The New England Journal of Medicine. 2013. Vol. 368:138–148

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            30 January 2025
            : 4
            : 1
            : 99-121
            Affiliations
            [a ]Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
            Author notes
            *Correspondence: wwei2@ 123456bidmc.harvard.edu (W. Wei)

            1These authors contribute equally.

            Article
            10.15212/AMM-2024-0075
            a8dc4aae-b01c-4473-a495-241302e7fabc
            2025 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 06 November 2024
            : 18 December 2024
            : 11 January 2025
            Page count
            Figures: 5, Tables: 3, References: 223, Pages: 23
            Funding
            Funded by: US National Institutes of Health (NIH)
            Award ID: CA177910
            Due to space constraints, we regretfully could not cite all relevant studies and we apologize to our colleagues whose work was omitted in this paper. We are grateful to the members of the Wei laboratory for valuable feedback and discussions during the review of this manuscript. We thank the US National Institutes of Health (NIH) for supporting this work through grant CA177910 awarded to W.W.
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            PROTAC,Cancer biology,Prostate cancer,Chemical biology

            Comments

            Comment on this article