Cancer-cell-intrinsic mechanisms shaping the immunosuppressive landscape of prostate cancer

  • Zhu, Yini2
  • Duong, Loan2
  • Lu, Xuemin1,
  • Lu, Xin3,
Asian Journal of Andrology 25(2):p 171-178, Mar-Apr 2023. | DOI: 10.4103/aja202283

Although immunotherapy has revolutionized cancer treatment and achieved remarkable success across many different cancer types, only a subset of patients shows meaningful clinical responses. In particular, advanced prostate cancer exhibits overwhelming de novo resistance to immune checkpoint blockade therapy. This is primarily due to the immunosuppressive tumor microenvironment of prostate cancer. Therefore, it is paramount to understand how prostate cancer cell-intrinsic mechanisms promote immune evasion and foster an immunosuppressive microenvironment. Here, we review recent findings that reveal the roles of the genetic alterations, androgen receptor signaling, cancer cell plasticity, and oncogenic pathways in shaping the immunosuppressive microenvironment and thereby driving immunotherapy resistance. Based on preclinical and clinical observations, a variety of therapeutic strategies are being developed that may illuminate new paths to enhance immunotherapy efficacy in prostate cancer.

INTRODUCTION

Prostate cancer (PCa) has the second highest incidence of all malignancies in men worldwide. Patients with localized and treatment naïve PCa typically respond well to radiation therapy, radical prostatectomy, and androgen deprivation therapy (ADT), with a 5-year survival rate of over 98%. However, a subset of the tumors ultimately progresses to the stage of metastatic castration-resistant prostate cancer (mCRPC). Moderate benefits can be achieved with currently approved therapies for mCRPC, such as radium-223 and second-generation androgen receptor (AR) antagonists. Although Sipuleucel-T was approved by the Food and Drug Administration (FDA) in 2010 based on an overall survival benefit, it is not used often in the clinic. PCa appears to have been left out of the subsequent immunotherapeutic revolution, which has largely been fueled by immune checkpoint blockade (ICB). ICB using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death one ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types. Unfortunately, advanced PCa has shown overwhelming de novo resistance to ICB.-

Cancer-cell-intrinsic and extrinsic mechanisms collectively underlie the formation of an immunosuppressive tumor microenvironment (TME), which is a fundamental cause of immunotherapy resistance. Understanding these mechanisms is a crucial step in developing novel therapeutic strategies to increase the response rate of immunotherapy. In this review, we will summarize recent studies that demonstrate how the genetic background, AR functional ablation, the plasticity of PCa cells, and oncogenic pathways respectively contribute to the shaping of the TME of PCa and cause the low responsiveness of PCa to immunotherapy (Figure 1). Based on these findings, some potential therapeutic strategies emerge that may overcome immunotherapy resistance and ultimately allow immunotherapy to benefit more patients with mCRPC.

PROSTATE CANCER GENETICS AND THE RESPONSE TO IMMUNOTHERAPY

Moderate mutation rate and a subclass with DDR mutations

Neoantigen recognition by cytotoxic T-lymphocytes (CTLs) is believed to be required for CTL-induced tumor cell killing. For most human cancers without a viral etiology, tumor neoantigens form through the various forms of genetic mutations. Tumors with higher somatic mutation load are generally associated with higher neoantigen formation., Unlike lung cancer or melanoma, which have mutation rates of around ten somatic mutations per megabase (Mb) or higher, the mutation rate of primary PCa is at the level of one mutation per Mb. Thus, PCa cells are less likely to be recognized by autologous T-cells., Metastatic PCa can reach 4.4 mutations per Mb. A moderate neoantigen load in PCa may be a fundamental reason for the limited infiltration of CTLs into the tumor, which defines an immunologically cold microenvironment.,

A clinically meaningful deviation from the general impression of the low mutational burden of PCa is that a subset of the patients (around 20%) harbor mutations in genes involved in DNA damage repair (DDR)., DDR gene mutation is associated with a higher mutation burden across cancer types, including PCa.,- Therefore, it is conceivable that PCa patients carrying DDR gene mutations contain a higher number of neoantigens. Of note, patients with DDR mutations exhibit a significantly different TME and show an overall better response to immunotherapy.- Deficient mismatch repair (dMMR) status was analyzed in 381 advanced PCa samples and was found to be associated with worse overall survival, a higher T-cell infiltration, and more PD-L1 expression. Breast cancer gene 2 (BRCA2)-mutated prostate tumors have higher infiltration of immune cells. Nevertheless, the intratumoral CD8 to forkhead box P3 (Foxp3) ratio was lower in BRCA2-mutated tumors suggesting a more suppressed TME. However, the case number in this study was low. Thus, further studies with more cases should be performed to validate the findings. Loss of mutS homolog 2 (MSH2), one of the mismatch repair (MMR) genes, was examined in a large cohort (1133 primary prostatic adenocarcinomas and 43 prostatic small cell carcinomas). The results showed that MSH2 loss was associated with high-grade primary tumors, hypermutation, and higher tumor-infiltrating immune cells. Overall, the DDR-mutated PCa manifests high immune infiltration, defining a genotype with a higher chance of response to immunotherapy. FDA has approved pembrolizumab for unresectable or metastatic solid tumors that have microsatellite instability-high (MSI-H) or dMMR, which was the FDA’s first tissue/site-agnostic approval. Therefore, patients with PCa characterized by dMMR or MSI-H can now benefit from the anti-PD1 immunotherapy.

PARP inhibitors

In the mCRPC patients with DDR gene defects, especially with BRCA1/2 mutation, poly(ADP-ribose) polymerase (PARP) inhibitors olaparib and rucaparib showed impressive efficacy and were both approved by the FDA to treat this type of advanced PCa., During DDR, PARP1 (the PARP family member responsible for most mammalian poly-ADP-ribosylation [PARylation]) is activated to catalyze the PARylation of many proteins, including PARP1 itself. PARP inhibitors possess both PARP inhibition and PARP trapping activities. PARP trapping prevents PARP1 from dissociating from the DNA, leading to more DNA damage., PARP inhibitor-induced PARP trapping and DNA damage lead to double-stranded DNA (dsDNA) leakage into the cytoplasm. Cytosolic dsDNA activates the innate immune signaling through the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway and stimulates the release of type I interferons., The higher probability of DNA damage by DDR deficiency may lead to higher neoantigen load and render PCa cells more immunogenic. Besides, PARP inhibitors can upregulate PD-L1 expression at least partly through induction of glycogen synthase kinase 3 beta (GSK3β) phosphorylation., PD-L1 upregulation may lead to more pronounced T-cell exhaustion in the TME, but it also provides the immune checkpoint target exploitable for anti-PD1/PD-L1 therapy. These immune-related effects of PARP inhibitors have prompted tremendous interest, and preclinical studies are converging to show that PARP inhibitors can help establish the anti-tumor immunity and promote the efficacy of ICB therapies in both DDR-deficient and -proficient cancers.,,, In PCa, at least three clinical trials are testing the combination of olaparib and anti-PD1 or anti-PD-L1 in advanced PCa (NCT04336943, NCT03834519, and NCT02484404), and the results will soon reveal if this combination is sufficient to sensitize advanced PCa to ICB therapy. We list all the clinical trials mentioned in this review in Table 1.

Enhanced neoantigen expression for PCa immunotherapy

The majority of PCa patients carry a moderate mutational load and a limited pool of neoantigens. For these PCa patients, improving the neoantigen expression or enhancing the immune system’s reaction to existing neoantigens is a likely promising strategy. The advancement in next-generation sequencing and analysis algorithms now allows the efficient identification of putative neoantigens recognizable by CTLs based on the tumor genome. Once validated experimentally, these neoantigens form the basis for immunotherapies such as adoptive T transfer or vaccines.,, Another possible approach is to artificially express the foreign antigens in tumor cells, which may be facilitated by microbial-mediated transgene expression. For instance, highly immunogenic tetanus toxoid (TT) proteins were delivered to pancreatic tumor cells through attenuated Listeria, which induced a strong immunological reaction by reactivating preexisting TT-specific memory T-cells.

Even though microbial-based immunotherapies are promising, overexpressing neoantigens specifically in the tumor area is limited by the difficulty in directional trafficking of the microbes to the tumor. Interestingly, intratumoral administration of mRNA may provide a new solution. For example, systemic IL-12 administration is hardly effective, but the intratumor administration of IL-12 mRNA induced a dramatic CD8+ T-cell-dependent tumor regression and sensitized the tumors to ICB across multiple syngeneic models. A clinical trial combining IL-12 mRNA administrated intratumorally and anti-PD-L1 durvalumab to treat solid tumors is being investigated (NCT03946800). Therefore, the intratumoral mRNA delivery of foreign antigens and cytokines critical to CTLs and natural killer (NK) cells may be developed as a novel immunotherapy approach to treat advanced PCa.

ANTIANDROGEN AND THE TME

Effect of androgen ablation on the TME

The effect of androgen in the TME is relevant to both PCa and other cancer types because sex differences in immune functions are well documented. Both sex chromosome genes and sex hormones, including estrogens, progesterone, and androgens, contribute to the differential regulation of innate and adaptive immune responses between males and females. Women generally mount a stronger immune response than men. In terms of response to ICB therapy, male cancer patients tend to respond better than female patients, but ICB in combination with chemotherapy tends to be more effective in female patients than male patients., In patients with PCa, ADT shows a profound impact on immune infiltration, e.g., increased intratumoral T-cells and macrophages., Drake et al. reported that androgen ablation by castration mitigated the tolerance of adoptive T-cell transfer targeting an engineered prostate-restricted antigen in the transgenic adenocarcinoma of mouse prostate (TRAMP) transgenic mouse model of PCa, providing one of the early motivations for exploring the potential of combining ADT and immunotherapy for PCa. In a recent study that conducted single-cell RNA sequencing (scRNA-seq) for mCRPC resistant to enzalutamide and prior to pembrolizumab, AR was found to be expressed in CD8+ T-cells and directly bind to the enhancer region of interferon gamma (IFNγ) gene. In preclinical models, AR blockade in CD8 T-cells prevented T-cell exhaustion and improved the efficacy of anti-PD1 therapy through elevating IFNγ production. From another study of transcriptome profiling of six paired pre-ADT biopsies with post-ADT PCa lesions, ADT was found to significantly increase the infiltration and activity of CTLs in PCa TME. This immune activation caused by ADT was more pronounced at the early stage of ADT but became very limited when the tumor progressed to the CRPC stage. This result is consistent with our RNA profiling of the treatment-naïve and fully formed CRPC (tumors progressed to the CRPC stage after surgical castration and enzalutamide treatment) from the phosphatase and tensin homolog/SMAD family member 4 (Pten/Smad4) transgenic mouse model of PCa, where the two tumor stages shared a broadly conserved transcriptome with only modest alterations of a number of collagen-related and immune-related genes including C-C motif chemokine receptor 2 (CCR2).

While ADT is promising to condition prostate tumors for more effective immunotherapy, evidence shows that AR inhibition or deprivation can induce a more immunosuppressive TME. In a transplanted PCa model, B-cell infiltration in tumors following ADT promotes CRPC through lymphotoxin secretion and inhibitor of nuclear factor kappa-B kinase subunit alpha/signal transducer and activator of transcription 3 (IKK-α/STAT3) activation. Furthermore, castration in PCa-bearing mice increased the infiltration of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) to the tumors by elevating IL-8 production from PCa cells to recruit PMN-MDSCs that express the cognate receptor, C-X-C motif chemokine receptor 2 (CXCR2). Several studies established that PMN-MDSCs infiltrate the PCa microenvironment and promote PCa progression and resistance to ICB therapy through inhibiting the T-cell-receptor signaling, activation, proliferation, and infiltration of CTLs.- Antiandrogen therapy also induces infiltration of other malignancy-promoting immune cells, such as macrophages, and mast cells. Taken these studies together, it suffices to say that ADT and AR antagonists will remodel the TME by disrupting AR signaling inside both prostate carcinoma cells as well as specific immune cells. Clinical trials combining ant-PD1/PD-L1 with ADT and/or enzalutamide in metastatic hormone-sensitive PCa, non-metastatic hormone-sensitive PCa, or mCRPC are being actively explored (e.g., NCT02312557, NCT03543189, NCT04191096, and NCT01875250). The results from these trials will guide new designs of systemic treatment of PCa.

Considerations for combining ADT and immunotherapy

Due to the complex roles of androgen signaling in shaping the TME and modulating immune cell activities, careful considerations should be given to the design and implementation when combining ADT and immunotherapy in PCa treatment. First, choosing the appropriate ADT regimen is critical. Different ADT methods may have different or opposite outcomes when combined with immunotherapy. Pu et al. showed that ADT with nonsteroid AR antagonists such as flutamide and enzalutamide significantly inhibited IFNγ production of immune cells and suppressed TLR9 agonist (CpG) immunotherapy by an off-target effect on γ-aminobutyric acid receptor type A (GABA-A) receptor inhibition. Meanwhile, ADT with orchiectomy or androgen synthesis inhibitor abiraterone did not have this off-target effect and significantly enhanced the antitumor effects of immunotherapy. Second, it is essential to choose the sequential steps that perform better in combination therapy. In a recent clinical study treating nonmetastatic CRPC patients with either poxvirus-based vaccine or the anti-androgen nilutamide, patients who received the vaccine first followed by nilutamide showed a significantly higher survival rate compared with patients who received nilutamide treatment first. Third, foreseeing the possible immunosuppressive mechanisms that might be stimulated due to ADT, an additional targeted therapy may be added to the ADT/immunotherapy combination. For example, ADT can induce IL-8 secretion, thus recruiting PMN-MDSCs to blunt the immunotherapy efficacy. A phase 1/2 trial based on this finding was initiated to test the combination of nivolumab (anti-PD1), BMS-986253 (anti-IL-8), and ADT drug degarelix to treat men with castration-sensitive PCa (NCT03689699).

NEUROENDOCRINE TRANSDIFFERENTIATION AND THE TME

The lineage plasticity of PCa drives the resistance to ADT and AR antagonists. Luminal adenocarcinoma accounts for 90%–95% of PCa at the initial diagnosis and is sensitive to ADT. By contrast, de novo neuroendocrine PCa (NEPC) is very rare (<0.1%). However, after ADT and CRPC development, the NEPC rate in CRPC patients increases to 11%–17%., NEPC mostly presents as small cell neuroendocrine carcinoma (SCNC). Instead of acquiring the resistance to ADT through genetic changes of AR that sustain AR signaling, NEPC stands as CRPC based on its nearly complete independence on AR signaling. There are at least two hypotheses to explain the origin of treatment-induced NEPC. While the neuroendocrine cell origin hypothesis postulates that NEPC is derived from preexisting neuroendocrine cells in the prostate, the lineage plasticity hypothesis posits that NEPC is the result of the transdifferentiation from luminal adenocarcinoma lineage to the neuroendocrine lineage. The published evidence hitherto offers more support to the lineage transdifferentiation hypothesis.,,

What does the NEPC transdifferentiation mean to the TME and immunotherapy? Analysis based on genomics and transcriptome profiling of human PCa samples of various stages and histology subtypes indicated that the tumor mutational burden of NEPC was similar to CRPC but significantly lower than small cell lung carcinoma. Like other metastatic PCa subtypes, NEPC was characterized by an immunologically “cold” TME, with the immune infiltration inversely correlated with survival. Immunohistochemical staining on the clinical samples showed that NEPC tumors had higher PD-L1 expression but lower PD1 expression than primary adenocarcinoma or CRPC., Interestingly, in a multi-institutional prospective study that characterized the clinical and genomic features of NEPC emergent after AR-targeting therapy, the genomic alterations in the DNA repair pathway were nearly mutually exclusive with the NEPC cases, suggesting that NEPC may encode low numbers of neoantigens and thus display weak responses to immunotherapy. This speculation was bolstered by an early-terminated phase 2 clinical trial, which showed that NEPC patients had an overall low response to the anti-PD-L1 antibody avelumab, except for one complete response (CR) with MSH2-mutated, MSI-H NEPC.

Immunotherapy certainly goes beyond ICB therapy. The distinct gene expression pattern between adenocarcinoma and NEPC may offer opportunities to identify NEPC-specific vulnerabilities amenable to immunotherapeutic targeting. For example, immunotherapies based on the prostate adenocarcinoma marker prostate-specific membrane antigen (PSMA), such as PSMA-targeted antibody-drug conjugate or chimeric antigen receptor T (CAR-T) cells, exhibited promising anti-tumor effects in treating mCRPC., However, PSMA expression was significantly suppressed in NEPC. Thus, PSMA-targeted immunotherapy is unlikely to be effective in treating NEPC. A systemic surfaceome profiling study identified FXYD domain containing ion transport regulator 3 (FXYD3) and CEA cell adhesion molecule 5 (CEACAM5) as cell surface antigens enriched in prostate adenocarcinoma and NEPC, respectively. Engineered CAR-T-cells targeting CEACAM5 induced antigen-specific cytotoxicity in NEPC cell lines. This indicates that targeting NEPC-specific surface markers for cellular immunotherapies (e.g., CAR-T and CAR-NK) may create novel avenues for treating NEPC.

ONCOGENIC PATHWAYS AS THE PRIMARY FORCE TO SHAPE THE TME

Cancer-cell-intrinsic oncogenic pathways are defined here as pro-tumor pathways activated by either gain of function of oncogenes or loss of function of tumor suppressor genes. These mechanisms are not only critical to the initiation and progression of the neoplastic core of the solid tumors, but also play fundamentally essential roles in impairing the induction and execution of a local antitumor immune response and mediating resistance to ICB therapy.- For advanced PCa, the cancer genome is characterized by rampant chromosomal instability and copy number alterations, including deletions and amplifications. mCRPC accumulates recurrent missense mutations in critical tumor suppressor genes such as tumor protein p53 (TP53) and speckle-type POZ protein (SPOP; >10%) and many other genes at low frequency (<3%) following a long-tail distribution., Research is actively ongoing to decipher how genes altered by copy number alterations or point mutations constitute the primary force to shape the immunologically cold TME of PCa. Below, we highlight some recent findings in this research front (Table 2).

PTEN/phosphatidylinositol 3-kinase (PI3K) pathway

Nearly 20% of primary PCa and 50% of CRPC patients harbor PTEN loss-of-function alterations, leading to hyperactivation of the PI3K pathway and cell-autonomous mechanisms of tumor cell proliferation and metastasis. Accumulating evidence shows that PTEN/PI3K pathway is also involved in the immune evasion of PCa. Prostate epithelial-specific Pten knockout in mice generated prostate adenocarcinoma with abundant infiltration of Gr-1+ CD11b+ MDSCs through upregulation of inflammatory cytokines and chemokines. In the more aggressive Pten/Smad4 double knockout mouse model of metastatic PCa, MDSCs were recruited through the CXCR2 chemokine pathway to the tumor bed and constituted over 50% of all the live cells, forming a formidable barrier for the antitumor immunity by T-cells and NK cells.PTEN loss increased CXCL8 secretion and sustained tumor cell survival. Higher CXCL8 led to the infiltration of tumor-associated macrophages (TAMs) in PCa. A study with transcriptome-based immune deconvolution followed by validation with clinical sample staining showed that PTEN deficiency was correlated with increased indoleamine 2,3-dioxygenase 1 (IDO1) expression and FoxP3+ regulatory T-cells in PCa. Given the high prevalence of PTEN loss in advanced PCa and the evidence supporting its consequence in shaping the immunosuppressive TME, pharmacological targeting of the PI3K pathway is expected to elicit dual effects: inhibiting the survival and proliferation of prostate neoplastic cells, and sensitizing mCRPC to ICB therapy and perhaps other modalities of immunotherapy.

TP53 mutations

Like in other cancer types, TP53 is also one of the most frequently mutated genes in advanced PCa. In primary PCa, 8% of patients harbor TP53 gene alteration. In mCPRC patients, however, the mutation frequency of TP53 dramatically increases to 50%.TP53 mutation is a critical underlying mechanism for senescence bypass, metastasis, and resistance to second-generation AR antagonists. In other solid tumors, loss of function of TP53 has been shown to induce immunosuppression through upregulating PD-L1, reprogramming TAMs to be immunosuppressive through exosomal miR-1246, and activating the nuclear factor-kappa B (NF-κB) signaling to promote chronic inflammation. In PCa, on the genetic backdrop of Pten loss, co-deletion of Trp53 in the mouse prostate led to faster tumor formation and increased MDSC infiltration through CXCL17 secretion. A truncated TP53 isoform Δ133TP53β was found elevated in PCa, and patients with high Δ133TP53β expression had an immunosuppressive TME featured by a higher proportion of immune cells positive for PD1, PD-L1, and colony stimulating factor 1 receptor (CSF1R) as well as CD163+ macrophages. Although TP53 missense mutation was associated with increased tumor-infiltrating T-cells in prostate primary tumors, it is very likely that the infiltrated T-cells are rendered exhausted and dysfunctional by the severely immunosuppressive TME. Nevertheless, the T-cells in the prostate tumors harboring TP53 missense mutations may be a targetable population for combinatorial immunotherapy to waken the antitumor cytotoxicity.

Other oncogenic pathways impacting the TME

MYC is a major driver of PCa tumorigenesis and progression.,MYC is amplified at chromosome 8q24 in about 4% of primary PCa and 10% of mCRPC. Han et al. developed an MYC inhibitor MYCi361 which induced proteasome-mediated MYC degradation in cancer cells through MYC phosphorylation on threonine-58. Inhibition of MYC with MYCi361 in PCa models suppressed immune infiltration, upregulated PD-L1 on tumor cells, and sensitized the tumors to anti-PD1 immunotherapy.

In a prostate adenocarcinoma model driven by the loss of Pten and Smad4, MDSCs were the primary infiltrating immune cell type in the tumor. Mechanistically, the transcription factor YAP was translocated to the nuclei of cancer cells to drive CXCL5 upregulation. CXCL5 recruited CXCR2-expressing MDSCs to the tumor area, blockade of CXCL5-CXCR2 signaling with CXCR2 antagonist triggered a robust antitumor response and prolonged mice survival.

Oncogenic wingless-type MMTV integration site family (WNT)–β-catenin signaling was first revealed to reduce T-cell recruitment and cause resistance to ICB therapy in melanoma. In mCRPC, mutations of APC and catenin beta 1 (CTNNB1) occur at 8.7% and 4.0%, respectively, indicative of hyperactivated WNT–β-catenin signaling. Linch et al. explored the intratumoral heterogeneity of PCa by sequencing multiple regions in human PCa biopsies and identified that patients with dMMR and patients with activating mutations of β-catenin harbored high inflammatory infiltration. Consistent with the findings from melanoma, patients with activating mutations of β-catenin exhibited a low CD8+/FoxP3+ ratio, a potential surrogate marker for immune evasion.

Genomic profiling identified a subtype of primary PCa characterized by missense mutation of SPOP (approximately 11%) and homozygous deletion of chromodomain helicase DNA-binding protein 1 (CHD1, approximately 8%), both encoding proteins involved in genome integrity and DNA repair through homologous recombination.,,- Using genetic mouse models that carried the loss of function of the two genes individually or simultaneously, we found that SPOP and CHD1 synergistically promoted repair of naturally occurring or chemically induced DNA damages in prostate epithelial cells. It will be important to determine whether this PCa subtypes is particularly sensitive to PARP inhibition and ICB therapy.

Frequent nonsynonymous mutations of SPOP seem to be unique to PCa, with the underlying reasons still unresolved. Interestingly, SPOP mutations were enriched in primary PCa relative to mCRPC, whereas other gene mutations were more enriched in metastases.SPOP as the substrate adaptor for the cullin 3-RING E3 ubiquitin ligase was stabilized through CDK4-mediated phosphorylation. The CDK4/6 inhibition or SPOP loss-of-function mutations impaired PD-L1 degradation and increased PD-L1 protein levels. Therefore, in PCa patients with SPOP mutation, SPOP function was lost, leading to higher PD-L1 and lower tumor-infiltrating lymphocytes.

Interestingly, deletions of CHD1 and PTEN display a mutually exclusive pattern in the PCa genome because CHD1 is essential to PCa cell survival when PTEN is deficient. Using genetic mouse models of PCa, Zhao et al. further found that in PTEN-deficient PCa, CHD1 fostered the immunosuppressive TME through sustaining IL-6 production and promoting MDSC recruitment.

An integrative genomic analysis of mCRPC samples identified cyclin-dependent kinase 12 (CDK12) biallelic inactivating mutations defining a distinct PCa subtype.CDK12 loss was mutually exclusive with DNA repair deficiency, ETS fusion, and SPOP mutations. Further, CDK12 loss was associated with genomic instability, higher neoantigen burden and more T-cell infiltration. These suggest that patients with CDK12 loss might have higher response to ICB treatment.

Polycomb repressor complex 1 (PRC1) and PRC2, acting in tandem to silence target genes, were reported to promote dedifferentiation and stemness during development and cancer. Using AR/NEPC double-negative PCa models, Su et al. found that overexpression of PRC1 in PCa cells induced CCL2 secretion and promoted the recruitment of M2-like tumor-associated macrophages (TAMs) and regulatory T-cells (Tregs). PRC1 inhibitor cooperated with ICB to abrogate the metastasis of double-negative PCa. The histone methyltransferase, enhancer of zeste homolog 2 (EZH2), is a catalytic subunit of PRC2 for tri-methylation of histone H3 at Lys 27 (H3K27me3). EZH2 is overexpressed in mCRPC and promotes PCa progression. Inhibition of EZH2 was found to activate the dsRNA-STING-interferon stress axis and significantly increase the infiltration of CD8+ T-cells, and M1-polarized TAMs in the PCa models. The combination of EZH2 inhibitor and anti-PD1 demonstrated combinatorial benefit in preclinical models.

CONCLUSION

For PCa to be treated effectively with immunotherapy, it is imperative to gain a deep understanding of the TME so that strategies can emerge to sensitize PCa to immunotherapy. Because malignancies originate from genetic mutations, the mechanisms dysregulating the immune landscape in solid tumors (including PCa) can be traced back to cancer-cell-intrinsic mechanisms. In this review, we have summarized four aspects of the cancer-cell-intrinsic regulation of the immunosuppressive TME in PCa: the generally moderate mutation burden of PCa and the subclass with DDR mutations, AR signaling ablation and the opportunity of combining ADT and immunotherapy, the TME of NEPC presenting unique challenges and opportunities for immunotherapy, and the profound effect of various oncogenic pathways on the TME (Figure 1). We believe that the ongoing basic and clinical studies aimed at defining the key drivers of the cold TME of PCa (especially the different genetic subtypes) will pave the road to bring the excitement of ICB and other modalities of immunotherapy to the realm of PCa.

AUTHOR CONTRIBUTIONS

YZ drafted the manuscript. Xuemin L and Xin L revised the manuscript. LD proofread the manuscript. All authors read and approved the final manuscript.

COMPETING INTERESTS

All authors declare no competing interests.

Acknowledgments

This work was supported by the National Institutes of Health grant R01CA248033 (to Xin L), Department of Defense CDMRP PCRP grants W81XWH2010312 (to Xin L) and W81XWH2010332 (to Xin L), an Investigator-Initiated Research Grant from American Institute for Cancer Research (to Xin L), Indiana CTSI pilot grants (to Xin L) through the NIH NCATS CTSA grant UL1TR002529, an Exceptional Project Award Grant from Breast Cancer Alliance (to Xin L), and CCV and IITP grants from Walther Cancer Foundation (to YZ and LD).

REFERENCES

  • 1.

    Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, et al. Prostate cancer. Nat Rev Dis Primers 2021;7:9
    Cited Here
  • 2.

    Sandhu S, Moore CM, Chiong E, Beltran H, Bristow RG, et al. Prostate cancer. Lancet 2021;398:1075 90
    Cited Here
  • 3.

    Litwin MS, Tan HJ The diagnosis and treatment of prostate cancer: a review. JAMA 2017;317:2532 42
    Cited Here
  • 4.

    Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411 22
    Cited Here
  • 5.

    Sharma P, Allison JP Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015;161:205 14
    Cited Here
  • 6.

    Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700 12
    Cited Here
  • 7.

    Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol 2017;35:40 7
  • 8.

    Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443 54
    Cited Here
  • 9.

    Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707 23
    Cited Here
  • 10.

    Schumacher TN, Schreiber RD Neoantigens in cancer immunotherapy. Science 2015;348:69 74
    Cited Here
  • 11.

    Jiang T, Shi T, Zhang H, Hu J, Song Y, et al. Tumor neoantigens: from basic research to clinical applications. J Hematol Oncol 2019;12:93
    Cited Here
  • 12.

    Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239 43
    Cited Here
  • 13.

    Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015;161:1215 28
    Cited Here
  • 14.

    Strasner A, Karin M Immune infiltration and prostate cancer. Front Oncol 2015;5:128
    Cited Here
  • 15.

    Ness N, Andersen S, Valkov A, Nordby Y, Donnem T, et al. Infiltration of CD8+ lymphocytes is an independent prognostic factor of biochemical failure-free survival in prostate cancer. Prostate 2014;74:1452 61
    Cited Here
  • 16.

    Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 2015;163:1011 25
    Cited Here
  • 17.

    Mei P, Freitag CE, Wei L, Zhang Y, Parwani AV, et al. High tumor mutation burden is associated with DNA damage repair gene mutation in breast carcinomas. Diagn Pathol 2020;15:50
    Cited Here
  • 18.

    Dai J, Jiang M, He K, Wang H, Chen P, et al. DNA damage response and repair gene alterations increase tumor mutational burden and promote poor prognosis of advanced lung cancer. Front Oncol 2021;11:708294
  • 19.

    Qu M, Zhao X, Chen H, Bai Y 1964P Genomic characteristics of homologous recombination in prostate cancer patients. Ann Oncol 2020;31:S1105
    Cited Here
  • 20.

    Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409 13
    Cited Here
  • 21.

    Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol 2020;38:395 405
  • 22.

    Nava Rodrigues D, Rescigno P, Liu D, Yuan W, Carreira S, et al. Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer. J Clin Invest 2018;128:4441 53
    Cited Here
  • 23.

    Hansen AR, Massard C, Ott PA, Haas NB, Lopez JS, et al. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol 2018;29:1807 13
    Cited Here
  • 24.

    Jenzer M, Kess P, Nientiedt C, Endris V, Kippenberger M, et al. The BRCA2 mutation status shapes the immune phenotype of prostate cancer. Cancer Immunol Immunother 2019;68:1621 33
    Cited Here
  • 25.

    Guedes LB, Antonarakis ES, Schweizer MT, Mirkheshti N, Almutairi F, et al. MSH2 loss in primary prostate cancer. Clin Cancer Res 2017;23:6863 74
    Cited Here
  • 26.

    Maio M, Ascierto PA, Manzyuk L, Motola-Kuba D, Penel N, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase 2 KEYNOTE-158 study. Ann Oncol 2022;33:929 38
    Cited Here
  • 27.

    de Bono J, Mateo J, Fizazi K, Saad F, Shore N, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med 2020;382:2091 102
    Cited Here
  • 28.

    Abida W, Patnaik A, Campbell D, Shapiro J, Bryce AH, et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1or BRCA2 gene alteration. J Clin Oncol 2020;38:3763 72
    Cited Here
  • 29.

    Satoh MS, Lindahl T Role of poly(ADP-ribose) formation in DNA repair. Nature 1992;356:356 8
    Cited Here
  • 30.

    Hopkins TA, Ainsworth WB, Ellis PA, Donawho CK, DiGiammarino EL, et al. PARP1 trapping by PARP inhibitors drives cytotoxicity in both cancer cells and healthy bone marrow. Mol Cancer Res 2019;17:409 19
    Cited Here
  • 31.

    Murai J, Huang SY, Das BB, Renaud A, Zhang Y, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res 2012;72:5588 99
    Cited Here
  • 32.

    Ding L, Kim HJ, Wang Q, Kearns M, Jiang T, et al. PARP inhibition elicits STING-dependent antitumor immunity in Brca1-deficient ovarian cancer. Cell Rep 2018;25:2972 80.e5
    Cited Here
  • 33.

    Shen J, Zhao W, Ju Z, Wang L, Peng Y, et al. PARPI triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCANEss. Cancer Res 2019;79:311 9
    Cited Here
  • 34.

    Jiao S, Xia W, Yamaguchi H, Wei Y, Chen MK, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res 2017;23:3711 20
    Cited Here
  • 35.

    Sen T, Rodriguez BL, Chen L, Della Corte CM, Morikawa N, et al. Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung cancer. Cancer Discov 2019;9:646 61
    Cited Here
  • 36.

    Wang Z, Sun K, Xiao Y, Feng B, Mikule K, et al. Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Sci Rep 2019;9:1853
    Cited Here
  • 37.

    Wang RF, Wang HY Immune targets and neoantigens for cancer immunotherapy and precision medicine. Cell Res 2017;27:11 37
    Cited Here
  • 38.

    Blass E, Ott PA Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol 2021;18:215 29
    Cited Here
  • 39.

    Selvanesan BC, Chandra D, Quispe-Tintaya W, Jahangir A, Patel A, et al. Listeria delivers tetanus toxoid protein to pancreatic tumors and induces cancer cell death in mice. Sci Transl Med 2022;14:eabc1600
    Cited Here
  • 40.

    Huang X, Pan J, Xu F, Shao B, Wang Y, et al. Bacteria-based cancer immunotherapy. Adv Sci (Weinh) 2021;8:2003572
    Cited Here
  • 41.

    Hewitt SL, Bailey D, Zielinski J, Apte A, Musenge F, et al. Intratumoral IL12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin Cancer Res 2020;26:6284 98
    Cited Here
  • 42.

    Klein SL, Flanagan KL Sex differences in immune responses. Nat Rev Immunol 2016;16:626 38
    Cited Here
  • 43.

    Wang S, Cowley LA, Liu XS Sex differences in cancer immunotherapy efficacy, biomarkers, and therapeutic strategy. Molecules 2019;24:3214
    Cited Here
  • 44.

    Conforti F, Pala L, Bagnardi V, De Pas T, Martinetti M, et al. Cancer immunotherapy efficacy and patients'sex: a systematic review and meta-analysis. Lancet Oncol 2018;19:737 46
    Cited Here
  • 45.

    Mercader M, Bodner BK, Moser MT, Kwon PS, Park ES, et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proc Natl Acad Sci U S A 2001;98:14565 70
    Cited Here
  • 46.

    Sorrentino C, Musiani P, Pompa P, Cipollone G, Di Carlo E Androgen deprivation boosts prostatic infiltration of cytotoxic and regulatory T lymphocytes and has no effect on disease-free survival in prostate cancer patients. Clin Cancer Res 2011;17:1571 81
    Cited Here
  • 47.

    Drake CG, Doody AD, Mihalyo MA, Huang CT, Kelleher E, et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer Cell 2005;7:239 49
    Cited Here
  • 48.

    Guan X, Polesso F, Wang C, Sehrawat A, Hawkins RM, et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature 2022;606:791 6
    Cited Here
  • 49.

    Long X, Hou H, Wang X, Liu S, Diao T, et al. Immune signature driven by ADT-induced immune microenvironment remodeling in prostate cancer is correlated with recurrence-free survival and immune infiltration. Cell Death Dis 2020;11:779
    Cited Here
  • 50.

    Zhao Y, Peng X, Baldwin H, Zhang C, Liu Z, et al. Anti-androgen therapy induces transcriptomic reprogramming in metastatic castration-resistant prostate cancer in a murine model. Biochim Biophys Acta Mol Basis Dis 2021;1867:166151
    Cited Here
  • 51.

    Ammirante M, Luo JL, Grivennikov S, Nedospasov S, Karin M B-cell-derived lymphotoxin promotes castration-resistant prostate cancer. Nature 2010;464:302 5
    Cited Here
  • 52.

    Lopez-Bujanda ZA, Haffner MC, Chaimowitz MG, Chowdhury N, Venturini NJ, et al. Castration-mediated IL-8 promotes myeloid infiltration and prostate cancer progression. Nat Cancer 2021;2:803 18
    Cited Here
  • 53.

    Lu X, Horner JW, Paul E, Shang X, Troncoso P, et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 2017;543:728 32
  • 54.

    Wang G, Lu X, Dey P, Deng P, Wu CC, et al. Targeting YAP-dependent MDSC infiltration impairs tumor progression. Cancer Discov 2016;6:80 95
    Cited Here
  • 55.

    Lu X, Lu X Enhancing immune checkpoint blockade therapy of genitourinary malignancies by co-targeting PMN-MDSCs. Biochim Biophys Acta Rev Cancer 2022;1877:188702
  • 56.

    Feng S, Cheng X, Zhang L, Lu X, Chaudhary S, et al. Myeloid-derived suppressor cells inhibit T cell activation through nitrating LCK in mouse cancers. Proc Natl Acad Sci U S A 2018;115:10094 9
  • 57.

    Wen J, Huang G, Liu S, Wan J, Wang X, et al. Polymorphonuclear MDSCs are enriched in the stroma and expanded in metastases of prostate cancer. J Pathol Clin Res 2020;6:171 7
    Cited Here
  • 58.

    Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, et al. CSF1 receptor targeting in prostate cancer reverses macrophage-mediated resistance to androgen blockade therapy. Cancer Res 2015;75:950 62
    Cited Here
  • 59.

    Dang Q, Li L, Xie H, He D, Chen J, et al. Anti-androgen enzalutamide enhances prostate cancer neuroendocrine (NE) differentiation via altering the infiltrated mast cells ? androgen receptor (AR) ? miRNA32 signals. Mol Oncol 2015;9:1241 51
    Cited Here
  • 60.

    Pu Y, Xu M, Liang Y, Yang K, Guo Y, et al. Androgen receptor antagonists compromise T cell response against prostate cancer leading to early tumor relapse. Sci Transl Med 2016;8:333ra47
    Cited Here
  • 61.

    Madan RA, Gulley JL, Schlom J, Steinberg SM, Liewehr DJ, et al. Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy. Clin Cancer Res 2008;14:4526 31
    Cited Here
  • 62.

    Beltran H, Hruszkewycz A, Scher HI, Hildesheim J, Isaacs J, et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin Cancer Res 2019;25:6916 24
    Cited Here
  • 63.

    Wang Y, Wang Y, Ci X, Choi SY, Crea F, et al. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol 2021;18:581 96
    Cited Here
  • 64.

    Zaffuto E, Pompe R, Zanaty M, Bondarenko HD, Leyh-Bannurah SR, et al. Contemporary incidence and cancer control outcomes of primary neuroendocrine prostate cancer: a SEER database analysis. Clin Genitourin Cancer 2017;15:e793 800
    Cited Here
  • 65.

    Aggarwal R, Huang J, Alumkal JJ, Zhang L, Feng FY, et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J Clin Oncol 2018;36:2492 503
    Cited Here
  • 66.

    Abida W, Cyrta J, Heller G, Prandi D, Armenia J, et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc Natl Acad Sci U S A 2019;116:11428 36
    Cited Here
  • 67.

    Zou M, Toivanen R, Mitrofanova A, Floch N, Hayati S, et al. Transdifferentiation as a mechanism of treatment resistance in a mouse model of castration-resistant prostate cancer. Cancer Discov 2017;7:736 49
    Cited Here
  • 68.

    Dong B, Miao J, Wang Y, Luo W, Ji Z, et al. Single-cell analysis supports a luminal-neuroendocrine transdifferentiation in human prostate cancer. Commun Biol 2020;3:778
    Cited Here
  • 69.

    Ferguson A, Bhinder B, Conteduca V, Sigouros M, Sboner A, et al. Immunogenomic landscape of neuroendocrine prostate cancer (NEPC). J Clin Oncol 2019;37:224
    Cited Here
  • 70.

    Nappi L, Kesch C, Vahid S, Fazli L, Eigl BJ, et al. Immunogenomic landscape of neuroendocrine small cell prostate cancer. J Clin Oncol 2019;37:217
    Cited Here
  • 71.

    Brown LC, Halabi S, Somarelli JA, Humeniuk M, Wu Y, et al. A phase 2 trial of avelumab in men with aggressive-variant or neuroendocrine prostate cancer. Prostate Cancer Prostatic Dis 2022 doi: 10.1038/s41391-022-00524-7. [Online ahead of print]
    Cited Here
  • 72.

    Narayan V, Barber-Rotenberg JS, Jung IY, Lacey SF, Rech AJ, et al. PSMA-targeting TGFβ-insensitive armored CAR Tcells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat Med 2022;28:724 34
    Cited Here
  • 73.

    Petrylak DP, Kantoff P, Vogelzang NJ, Mega A, Fleming MT, et al. Phase 1 study of PSMA ADC, an antibody-drug conjugate targeting prostate-specific membrane antigen, in chemotherapy-refractory prostate cancer. Prostate 2019;79:604 13
    Cited Here
  • 74.

    Bakht MK, Derecichei I, Li Y, Ferraiuolo RM, Dunning M, et al. Neuroendocrine differentiation of prostate cancer leads to PSMA suppression. Endocr Relat Cancer 2018;26:131 46
    Cited Here
  • 75.

    Lee JK, Bangayan NJ, Chai T, Smith BA, Pariva TE, et al. Systemic surfaceome profiling identifies target antigens for immune-based therapy in subtypes of advanced prostate cancer. Proc Natl Acad Sci U S A 2018;115:E4473 82
    Cited Here
  • 76.

    Spranger S, Gajewski TF Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer 2018;18:139 47
    Cited Here
  • 77.

    Wellenstein MD, de Visser KE Cancer-cell-intrinsic mechanisms shaping the tumor immune landscape. Immunity 2018;48:399 416
  • 78.

    Kalbasi A, Ribas A Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20:25 39
  • 79.

    Ghoneim HE, Zamora AE, Thomas PG, Youngblood BA Cell-intrinsic barriers of T cell-based immunotherapy. Trends Mol Med 2016;22:1000 11
    Cited Here
  • 80.

    Spratt DE, Zumsteg ZS, Feng FY, Tomlins SA Translational and clinical implications of the genetic landscape of prostate cancer. Nat Rev Clin Oncol 2016;13:597 610
    Cited Here
  • 81.

    Armenia J, Wankowicz SA, Liu D, Gao J, Kundra R, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet 2018;50:645 51
    Cited Here
  • 82.

    Jamaspishvili T, Berman DM, Ross AE, Scher HI, De Marzo AM, et al. Clinical implications of PTEN loss in prostate cancer. Nat Rev Urol 2018;15:222 34
    Cited Here
  • 83.

    Garcia AJ, Ruscetti M, Arenzana TL, Tran LM, Bianci-Frias D, et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression. Mol Cell Biol 2014;34:2017 28
    Cited Here
  • 84.

    Maxwell PJ, Coulter J, Walker SM, McKechnie M, Neisen J, et al. Potentiation of inflammatory CXCL8 signalling sustains cell survival in PTEN-deficient prostate carcinoma. Eur Urol 2013;64:177 88
    Cited Here
  • 85.

    Armstrong CW, Maxwell PJ, Ong CW, Redmond KM, McCann C, et al. PTEN deficiency promotes macrophage infiltration and hypersensitivity of prostate cancer to IAP antagonist/radiation combination therapy. Oncotarget 2016;7:7885 98
    Cited Here
  • 86.

    Vidotto T, Saggioro FP, Jamaspishvili T, Chesca DL, Picanco de Albuquerque CG, et al. PTEN-deficient prostate cancer is associated with an immunosuppressive tumor microenvironment mediated by increased expression of IDO1 and infiltrating FoxP3+ T regulatory cells. Prostate 2019;79:969 79
    Cited Here
  • 87.

    Teroerde M, Nientiedt C, Duensing A, Hohenfellner M, Stenzinger A, et al. Revisiting the role of p53 in prostate cancer Bott SR, Ng KL Prostate Cancer Brisbane (AU) Exon Publications 2021 113 24
    Cited Here
  • 88.

    Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, et al. PDL1 regulation by p53 via miR-34. J Natl Cancer Inst 2016;108:djv303
    Cited Here
  • 89.

    Cooks T, Pateras IS, Jenkins LM, Patel KM, Robles AI, et al. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun 2018;9:771
    Cited Here
  • 90.

    Cooks T, Pateras IS, Tarcic O, Solomon H, Schetter AJ, et al. Mutant p53 prolongs NF-κB activation and promotes chronic inflammation and inflammation-associated colorectal cancer. Cancer Cell 2013;23:634 46
    Cited Here
  • 91.

    Bezzi M, Seitzer N, Ishikawa T, Reschke M, Chen M, et al. Diverse genetic-driven immune landscapes dictate tumor progression through distinct mechanisms. Nat Med 2018;24:165 75
    Cited Here
  • 92.

    Kazantseva M, Mehta S, Eiholzer RA, Gimenez G, Bowie S, et al. The Delta133p53beta isoform promotes an immunosuppressive environment leading to aggressive prostate cancer. Cell Death Dis 2019;10:631
    Cited Here
  • 93.

    Kaur HB, Lu J, Guedes LB, Maldonado L, Reitz L, et al. TP53 missense mutation is associated with increased tumor-infiltrating T cells in primary prostate cancer. Hum Pathol 2019;87:95 102
    Cited Here
  • 94.

    Qiu X, Boufaied N, Hallal T, Feit A, de Polo A, et al. MYC drives aggressive prostate cancer by disrupting transcriptional pause release at androgen receptor targets. Nat Commun 2022;13:2559
    Cited Here
  • 95.

    Rebello RJ, Pearson RB, Hannan RD, Furic L Therapeutic approaches targeting MYC-driven prostate cancer. Genes (Basel) 2017;8:71
    Cited Here
  • 96.

    Han H, Jain AD, Truica MI, Izquierdo-Ferrer J, Anker JF, et al. Small-molecule MYC inhibitors suppress tumor growth and enhance immunotherapy. Cancer Cell 2019;36:483 97.e15
    Cited Here
  • 97.

    Spranger S, Bao R, Gajewski TF Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015;523:231 5
    Cited Here
  • 98.

    Linch M, Goh G, Hiley C, Shanmugabavan Y, McGranahan N, et al. Intratumoural evolutionary landscape of high-risk prostate cancer: the PROGENY study of genomic and immune parameters. Ann Oncol 2017;28:2472 80
    Cited Here
  • 99.

    Boysen G, Barbieri CE, Prandi D, Blattner M, Chae SS, et al. SPOP mutation leads to genomic instability in prostate cancer. Elife 2015;4:e09207
    Cited Here
  • 100.

    Zhang D, Wang H, Sun M, Yang J, Zhang W, et al. Speckle-type POZ protein, SPOP, is involved in the DNA damage response. Carcinogenesis 2014;35:1691 7
  • 101.

    Kari V, Mansour WY, Raul SK, Baumgart SJ, Mund A, et al. Loss of CHD1 causes DNA repair defects and enhances prostate cancer therapeutic responsiveness. EMBO Rep 2016;17:1609 23
  • 102.

    Shenoy TR, Boysen G, Wang MY, Xu QZ, Guo W, et al. CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair. Ann Oncol 2017;28:1495 507
    Cited Here
  • 103.

    Zhu Y, Wen J, Huang G, Mittlesteadt J, Wen X, et al. CHD1 and SPOP synergistically protect prostate epithelial cells from DNA damage. Prostate 2021;81:81 8
    Cited Here
  • 104.

    Zhang J, Bu X, Wang H, Zhu Y, Geng Y, et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018;553:91 5
    Cited Here
  • 105.

    Zhao D, Lu X, Wang G, Lan Z, Liao W, et al. Synthetic essentiality of chromatin remodelling factor CHD1 in PTEN-deficient cancer. Nature 2017;542:484 8
    Cited Here
  • 106.

    Zhao D, Cai L, Lu X, Liang X, Li J, et al. Chromatin regulator CHD1 remodels the immunosuppressive tumor microenvironment in PTEN-deficient prostate cancer. Cancer Discov 2020;10:1374 87
    Cited Here
  • 107.

    Wu YM, Cieslik M, Lonigro RJ, Vats P, Reimers MA, et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell 2018;173:1770 82.e14
    Cited Here
  • 108.

    Su W, Han HH, Wang Y, Zhang B, Zhou B, et al. The polycomb repressor complex 1 drives double-negative prostate cancer metastasis by coordinating stemness and immune suppression. Cancer Cell 2019;36:139 55.e10
    Cited Here
  • 109.

    Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 2002;419:624 9
    Cited Here
  • 110.

    Morel KL, Sheahan AV, Burkhart DL, Baca SC, Boufaied N, et al. EZH2 inhibition activates a dsRNA-STING-interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer. Nat Cancer 2021;2:444 56
    Cited Here
Copyright © 2022 Copyright: © The Author(s)(2022)
View full text|Download PDF