Skip to main content
American Journal of Clinical and Experimental Urology logoLink to American Journal of Clinical and Experimental Urology
. 2022 Aug 15;10(4):210–233.

Prostate cancer immunotherapy: a review of recent advancements with novel treatment methods and efficacy

Ian Wang 1, Liankun Song 2, Beverly Y Wang 3, Arash Rezazadeh Kalebasty 4, Edward Uchio 4,5, Xiaolin Zi 2,4,5,6
PMCID: PMC9428569  PMID: 36051616

Abstract

Immunotherapy remains to be an appealing treatment option for prostate cancer with some documented promise. Prostate cancer is traditionally considered as an immunologically “cold” tumor with low tumor mutation burden, low expression of PD-L1, sparse T-cell infiltration, and a immunosuppressive tumor microenvironment (TME). Sipuleucel-T (Provenge) is the first FDA approved immunotherapeutic agent for the treatment of asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer (mCRPC); demonstrating a benefit in overall survival. However various clinical trials by immune checkpoint inhibitors (ICIs) and their combinations with other drugs have shown limited responses in mCRPC. Up to now, only a small subset of patients with mismatch repair deficiency/microsatellite instability high and CDK12 mutations can clinically benefit from ICIs and/or their combinations with other agents, such as DNA damage agents. The existence of a large heterogeneity in genomic alterations and a complex TME in prostate cancer suggests the need for identifying new immunotherapeutic targets. As well as designing personalized immunotherapy strategies based on patient-specific molecular signatures. There is also a need to adjust strategies to overcome histologic barriers such as tissue hypoxia and dense stroma. The racial differences of immunological responses between men of diverse ethnicities also merit further investigation to improve the efficacy of immunotherapy and better patient selection in prostate cancer.

Keywords: Prostate cancer, immunotherapy, immune checkpoint inhibitors, tumor microenvironment, African American, Race, European American

Introduction

Prostate cancer is the most commonly diagnosed cancer in men, and the second most diagnosed disease for men in the U.S. As of 2022, the estimated new cases of prostate cancer in the U.S is said to be 268,490 [1]. This makes up nearly 21% of all cancer cases in men. Alongside an estimated 34,500 deaths that year has made it the second most common form of cancer related death in the United States after lung and bronchus cancer [1]. With the advancement of detection methods such as the prostate health index, urine prostate cancer antigen 3 (PCA3) test, and magnetic resonance imaging (MRI) fusion prostate biopsy [2-4], diagnosis of this disease has greatly improved over the past few decades. However, the mortality rate of the disease remains very high despite modern day treatments.

A few forms of immunotherapy have become part of standard care over the past few years. Novel immunotherapy approaches utilize a wide variety of immune response mechanisms to target malignant cells. This treatment method has shown promising results in patients with aggressive cancers. Some of these agents have been able to produce deep and long-term remission in malignancies with otherwise limited treatment options. Advancements in immunology as well as the approval of drugs such as sipuleucel-T and ICIs provide a viable alternative treatment modality for castration-resistant prostate cancer (CRPC) to prevalent methods such as androgen suppression therapy and chemotherapy [5,6]. The goal of immunotherapy is to target cancer cells through the recognition by T-cells or antibodies; essentially encouraging an immune response to cancer, but it has been shown to be less effective against prostate cancer when compared to results from other cancers including non-small-cell lung cancer (NSCLC) [7], renal cell carcinoma (RCC) [8,9], urothelial cancer [10,11], head and neck cancer [12], and melanoma [13,14]. The strong immunosuppressive tumor microenvironment (TME), lower infiltration of T-cells, and lower mutation burden in prostate cancer have resulted in lowered efficacy of treatment through immunotherapy [15]. Nevertheless, a subset of prostate cancer exhibits immunogenic phenotype. A special subgroup of patients with high PD-L1 tumor expression, CDK12 mutations, high tumor mutational burden, or tumors with high microsatellite instability (MSI) and mismatch repair-deficient (dMMR) have recently demonstrated excellent responses to immune checkpoint inhibitors (ICIs) and/or their combinations with other agents [15,16]. Therefore, immunotherapy remains to be an appealing treatment option for prostate cancer to optimize the management of this disease. This review will summarize the current state of immunotherapy usage, including immune checkpoint blockade therapy, vaccine-based treatments, adoptive cell therapy and bispecific T cell engager (BiTE) therapy in prostate cancer treatment. In addition, we will discuss current mechanisms of resistance or responses to ICIs, and the immunological differences between African American (AA) and European American (EA) men with prostate cancer.

ICIs based therapy

Ipilimumab is the first FDA approved immune check point inhibitor. Ipilimumab is a monoclonal antibody that works to upregulate immune response by targeting the immune downregulating receptor, CTLA-4 [17]. Activated T cells induce CTLA-4 expression to send out inhibitory signal to T cells [18]. In addition, CTLA-4 is constitutively expressed in regulatory T cells to control cytotoxic T-cell activation [18]. When administered as a monotherapy, ipilimumab was shown to noticeably increase the ratio of regulator effector T lymphocytes present in the TME [17]. In a phase I trial, two out of fourteen patients with mCRPC who received a single intra-venous dose of Ipilimumab exhibited prostate specific antigen (PSA) decreases of > or =50% and treatment was well tolerated [19]. In another Phase I trial of tremelimumab (a humanized anti-CTLA-4 antibody) in combination with androgen deprivation using bicalutamide in PSA recurrent prostate cancer patients without radiographic evidence of metastatic disease, three out of eleven patients experienced an extension in PSA doubling time [20]. Phase I trial combining ipilimumab with a vaccine containing transgenes for PSA and for a triad of costimulatory molecules (PROSTVAC) in patients with mCRPC showed a PSA decline in 14 out of 24 (58%) chemotherapy-naïve patients [21]. A combination of evofosfamide, a prodrug that alleviates hypoxia, with ipilimumab resulted in 3 (16.7%) partial response and 12 (66.7%) stable disease in 18 patients with measurable disease at baseline [22]. These responsive patients had improved peripheral T-cell proliferation and increased intra-tumoral T-cell infiltration [22]. In a phase I trial of ipilimumab at escalating doses in combination with a fixed dose of GM-CSF, 24 patients with mCRPC were treated and three of six patients treated at the highest dose level had PSA declines of >50% [23]. The combined therapy induced the expansion of activated effector CD8 T cells and tumor-associated antigens specific T cells [23]. In phase I/II study in patients with metastatic CRPC (mCRPC), 50 patients received ipilimumab alone or as in combination with radiotherapy. Eight had PSA declines of ≥50%, one had complete response, and six had stable disease [24].

In a double-blind, phase 3 trial, 799 mCRPC patients were randomly assigned to palliative radiotherapy followed by ipilimumab or placebo [6]. There was a statistically significant improvement in progression-free survival but no statistically significant difference in overall survival (OS) between ipilimumab vs. placebo groups. However in long term follow-up study, OS rates in the ipilimumab versus placebo arms are 25.2% vs. 16.6% at 2 years, 15.3% vs. 7.9% at 3 years, 10.1% vs. 3.3% at 4 years, and 7.9% vs. 2.7% at 5 years. Approximately two to three times higher survival benefit after three years was identified in the ipilimumab arm [25]. Beer reported an increase in median progression-free survival in the ipilimumab arm (5.6 months) versus placebo arm (3.8 months) and a higher PSA response rate (23% in the ipilimumab arm vs. 8% in the placebo arm) in a trial of ipilimumab versus placebo as monotherapy in asymptomatic/oligo-symptomatic chemo-naïve mCRPC [26].

Nivolumab is a human IgG4 monoclonal antibody for blocking PD-1. PD-1 interaction with its ligand prevents activation of T cells from attacking the cancer [27-29]. In the phase II clinical trial CheckMate 650, which investigated combined effects of ipilimumab and nivolumab in patients with mCRPC who had developed resistance to androgen receptor (AR)-targeted therapies, the combination resulted in 25% of objective response rate and was associated with considerable side effects leading to discontinuation of the therapy in the population [29]. In another phase II trial of nivolumab and ipilimumab combination, patients with ARV7 positive mCRPC were treated with or without enzalutamide. In the arm without enzalutamide, there was 13% (2/15) PSA response, and the objective response rate (ORR) was shown to be 25% (2/8). In the arm with enzalutamide, PSA response rate and ORR were shown to be 0% (0/15) and 0% (0/9) in those with measurable disease. 20% (3/15) patients without enzalutamide and 26.7% (4/15) patients with enzalutamide reached a durable progression-free survival more than two years. The results did not justify further studies in unselected patients [30].

Pembrolizumab is an anti-PD1 antibody. In a multiple cohort phase II trial (the KEYNOTE-199), a pembrolizumab monotherapy was utilized in 258 patients with Response Evaluation Criteria in Solid Tumours (RECIST)-measurable and bone-predominant mCRPC who were previously treated with docetaxel and targeted endocrine therapy. This study showed an ORR of 5% in PD-L1-positive RECIST-measurable patients and 3% in PD-L1-negative RECIST-measurable. Median OS was 9.5 months in PD-L1-positive RECIST-measurable patients, 7.9 months in PD-L1-negative RECIST-measurable patients, and 14.1 months in patients with bone-predominant disease regardless of PD-L1 expression [31]. Another nonrandomized phase Ib KEYNOTE-028 trial of pembrolizumab in PD-L1-positive, mCRPC patients who received at least two prior therapies showed an ORR of 17.4%, median progression-free survival (PFS) and OS of 3.5 and 7.9 months, respectively [32].

In the Phase 1b/2 KEYNOTE-365 study (NCT02861573), pembrolizumab plus docetaxel and prednisone demonstrated a 34% PSA response rate, 23% ORR, 8.5 months of median radiographic PFS (rPFS), and 20.2 months of OS in chemotherapy-naïve mCRPC patients who were treated with abiraterone or enzalutamide before [33]. The multicenter, randomized, double-blind Phase III study, KEYNOTE-921, is now ongoing for further evaluating the therapeutic efficacies of pembrolizumab plus docetaxel and prednisone or prednisolone versus a placebo plus docetaxel in this patient population with primary endpoints of OS and rPFS [34].

A single-arm phase II trial of pembrolizumab in combination with enzalutamide was carried out in 28 men with mCRPC progressing on enzalutamide alone. The trial achieved a PSA response rate of 18% (5/28), ORR of 25% (3/12), median PSA PFS of 3.8 months, OS of 21.9 months in all patients and OS of 41.7 months in the responders [35]. Among the three responders, one is MSI high, and none had detectable PD-L1 expression in their baseline biopsy tissues [35]. A multicenter, randomized, double-blind, Phase III KEYNOTE-641 study is now ongoing to further evaluate the efficacy and safety of pembrolizumab plus enzalutamide versus enzalutamide plus placebo in estimated 1200 patients with mCRPC (NCT03834493) [36]. Similar studies have been performed with other ICIs and some of the phase III trials are being completed [37].

Phase I and II trials of pembrolizumab in combination with ADXS31-142 [a cancer vaccine containing a live-attenuated strain of the Gram-positive bacterium Listeria monocytogenes encoding a fusion protein consisting of PSA and a fragment of the immunostimulant listeriolysin O (LLO) protein], MVI-816 (a DNA vaccine encoding prostatic acid phosphatase) or cryotherapy were carried out in patients with progressive mCPRC or newly diagnosed oligometastatic hormone-sensitive prostate cancer [38-41]. These trials observed durable responses of the combined therapies in a subset of prostate cancer patients [38-41]. However future randomized clinical trials are needed to validate these findings.

The usage of atezolizumab, avelumab, and durvalumab to target PD-L1 and to block the interaction of PD-L1 with the PD-1 has also been explored as an option in treatment of advanced prostate cancer [42-49]. In the randomized phase III trial IMbassador 250 (no. NCT03016312), atezolizumab in combination with enzalutamide was compared to enzalutamide alone in 759 patients with mCRPC or locally advanced CRPC patients who had progressed on abiraterone, and docetaxel did not reach the primary endpoint for a better OS in the combination arm [43]. The results also suggested that atezolizumab plus enzalutamide might be useful in selected patients with pre-existing immunity, such as high PDL1 expression and high levels of CD8+ T cells [43]. A phase II study of avelumab with stereotactic ablative body radiotherapy in 31 men with progressive mCRPC treated previously with anti-androgen agents exhibited an ORR of 31%, rPFS of 8.4 months and median OS of 14.1 months [44]. In another phase II trial of avelumab in 15 men with progressive neuroendocrine or aggressive-variant metastatic prostate cancer (NEPC/AVPC) one man (6.7%) with MSH2 somatic mutation and MSI-high NEPC achieved complete remission for 2 years [48]. Seventeen patients with previously treated mCRPC with or without alterations in DDR genes who received durvalumab and olaparib combination were reported to have median rPFS of 16.1 months and a radiographic and/or PSA response of 53% [49].

Other trials for testing PD-1 blockade in combination with anti-IL6, TGF-β blockage, and IDO1 inhibitor, as well as other immune checkpoint targets, such as B7-H3 inhibitors enoblituzumab, LAG-3, OX40, and 4-1BBL are in various stages of clinical development for phase I and II trials in mCRPC [50-55] (NCT03821246, NCT02628535, NCT02923180, NCT01391143). Clinical trials of a variety of prostate cancer immune therapies were summarized as Table 1.

Table 1.

Clinical Trials of varying treatment plans for mCRPC and other forms of carcinoma

Treatment N Target Dosing Interval Data Collection method Results Article
Ipilimumab 399 to 400 mCRPC that has progressed post docetaxel chemo therapy 10 mg/kg every 3 weeks Intention-to-treat analysis No significant difference [6]
Nivolumab 854 Non-small-lung cancer 3 mg/kg every 2 weeks Kaplan-Meier method 9%-15% increase in overall survival rates compared to docetaxel [7]
Nivolumab 821 Advanced clear cell renal-cell carcinoma 3 mg/kg every 2 weeks Kaplan-Meier method Roughly 5 month increase in survival [8]
Nivolumab plus Ipilimumab 1096 Advanced clear cell renal-cell carcinoma 1 mg/kg every 3 weeks RECIST evaluation 15% increased survival rate [9]
Pembrolizumab alone or with chemotherapy 1010 Advanced urothelial carcinoma 200 mg every 3 weeks Comparisons of non-inferiority and superiority No significant difference [10]
Ramucirumab 530 Advanced or metastatic urothelial carcinoma 10 mg/kg every 3 weeks Intention-to-treat analysis Average 1.5 month increased survival [11]
Avelumab 697 Advanced squamous cell carcinoma of head and neck 10 mg/kg every 2 weeks Response Evaluation Criteria in Solid Tumors No significant difference [12]
Ipilimumab and Nivolumab 14 Melanoma 3 dose per kg every 3 weeks ECOG 8.9 month OS vs. 2.9 months [13]
Tremelimumab 11 PSA recurrent prostate cancer 150 mg of bicalutamide for 28 days followed by temelimumab on 29th day Flow cytometric analysis No significant adverse effects reported [20]
Ipilimumab with PSA transgene vaccine 30 mCRPC Varying doses of ipilimumab every 2 weeks with monthly vaccination booster Flow cytometry Trending towards associations of longer overall survival with no conclusive data [21]
And Kaplan-Meier method
Evofosfamide with Ipilimumab 22 Patients with mCRPC, pancreatic cancer, and/or head and neck cancer 400-640 mg/m2 evofosfamide and 3 mg/kg ipilumumab every 3 weeks RECIST Evaluation No significant observations [22]
ECOG evaluation
CTLA4 blockade with GM-CSF combination 24 mCRPC Escalating doses of ipilumumab with fixed dose of GM-CSF given every 4 weeks Flow cytometry >50% PSA decline in 3 patients with no significant observations in the rest [23]
Ipilimumab 50 mCRPC Varying doses of ipilimumab from 3-10 mg/kg every 3 weeks RECIST >50% PSA declines amongst some patients receiving 10 mg/kg doses [24]
Ipilimumab 799 mCRPC One dose of radiotherapy followed by 10 mg/kg ipilumumab every 3 weeks Two sided log-rank test stratified by ECOG Overall increased survival rates for patients given ipilumumab [25]
Ipilimumab 598 Asymptomatic mCRPC 10 mg/kg every 3 weeks Two sided log-rank test stratified by ECOG No significant difference in overall survival rates [26]
Nivolumab plus Ipilimumab 78 mCRPC 1 mg/kg nivolumab + 3 mg/kg ipilimumab followed by 480 mg nivolumab every 4 weeks ECOG Reported consistent safety [28]
Nivolumab plus Ipilimumab 15 AR-V7 expressing mCRPC 1 mg/kg ipilimumab + 3 mg/kg nivolumab every 3 weeks ECOG No significant observations [30]
Pembrolizumab 258 mCRPC 200 mg every 3 weeks RECIST Median overall survival rate of 14.1 months with acceptable safety [31]
ECOG
Pembrolizumab 23 Advanced prostate adenocarcinoma 10 mg/kg every 2 weeks RECIST Overall survival of 7.9 months [32]
Pembrolizumab plus Docetaxel and Prednisone 104 mCRPC 200 mg pembrolizumab and 75 mg/m2 docetaxel every 3 weeks, 5 mg prednisone BID RECIST Overall survival of 29.2 months, reported acceptable safety [33]
Pembrolizumab plus Docetaxel ~1000 mCRPC Every 3 weeks RECIST Ongoing phase 3 trial [34]
TFST
Pembrolizumab plus Enzalutamide 28 mCRPC 200 mg pembrolizumab every 3 weeks with 4 doses with enzalutamide RECIST Overall survival of 41.7 months [35]
Pembrolizumab plus Enzalutamide ~1200 mCRPC 200 mg pembrolizumab every 3 weeks 160 mg/day enzalutamide PCWG3 modified RECIST Ongoing phase 3 trial [36]
Pembrolizumab plus MV1-816 25 mCRPC Every 3 weeks RECIST Overall survival of 22.9 months [38]
pTVG-HP (MVI-816) Vaccine 99 mCRPC 200 μg 6 times every 2 weeks, then quarterly for 2 years PCWG3 modified RECIST No significant change [39]
Pembrolizumab plus ADXS31-142 37 mCRPC 200 mg with monotherapy every 3 weeks RECIST Overall survival of 16.0 months [40]
Pembrolizumab with Cryotherapy 12 mCRPC 200 mg every 3 weeks with eight months cryotherapy Kaplan Meier Overall survival of 17.5 months [41]
Atezolizumab 35 mCRPC Every 3 weeks Kaplan Meier Overall survival of 14.7 months with acceptable safety profile [42]
Atezolizumab with Ezalutamide 759 mCRPC Every 3 weeks - Ongoing study [43]
Atezolizumab with Radium-223 45 mCRPC 840 mg every 2 weeks, Radium-223 every 4 weeks RECIST Overall survival of 16.3 months [44]
Atezolizumab with Sipuleucel-T 37 mCRPC 1200 mg azetolizumab every 3 weeks, sipuleucel-T every 2 weeks RECIST Overall survival of 23.6 months [45]
Atezolizumab with Cabozantinib 580 mCRPC 1200 mg atezoliumab followed by 40 mg cabozantinib PO QD RECIST Ongoing study [46]
Avelumab with Stereotactive Ablative Body Radiotherapy 31 mCRPC 10 mg/kg every 2 weeks for 24 weeks Clopper-Pearson Overall survival of 14.1 months [47]
Avelumab 15 mCRPC 10 mg/kg every 2 weeks RECIST Overall survival of 7.4 months [48]
Durvalumab with Olaparib 17 mCRPC 1500 mg durvalumab every 4 weeks, 300 mg olaparib PO BID RECIST Overall survival of 16.1 months [49]
Ieramilimab plus spartalizumab 255 Advanced or metastatic tumors 1 mg/kg every 2 weeks RECIST Acceptable safety profile [53]
RhoC Vacccine 22 Prostate cancer 9.1 mg every 2 weeks for 6 cycles followed by every 4 weeks for 5 cycles Flow cytometry CD4 T-Cell response lasting 10 months and generally well tolerated [54]
Nivolumab plus Ipilimumab 90 mCRPC 1 mg/kg nivolumab and 3 mg/kg ipilimumab IV followed by 480 mg nivolumab every 4 weeks RECIST Overall survival of 19.0 months [80]

Current mechanisms underly ICIs responses in prostate cancer

A small subset (3-5%) of mCRPC patients with a microsatellite instability (MSI) and dMMR phenotype exhibit high-tumoral mutation burden and higher levels of tumor infiltrating lymphocytes (TILs). It has been reported that some cancer patients with dMMR, including mCRPC, colorectal, and endometrial cancers display exceptional responses to the anti-PD-1 pembrolizumab [56-58]. These results led to the approval of pembrolizumab by FDA for the treatment of all MMR-deficient metastatic tumors, which was based on a predictive biomarker alone, but not on tumor histology.

Biallelic loss of CDK12 represents another novel subtype of prostate cancer, which holds significant promise for immunotherapy and is genetically, transcriptionally, and phenotypically distinct from tumors with homologous recombination repair defects (HRD) and dMMR [59,60]. CDK12 mutations occur in 2%-4% of primary prostate cancers and in 4.7%-11% of mCRPCs and associated with a high rate of metastases and short overall survival [61-67]. CDK12-mutated prostate cancer is characterized with focal tandem duplications (FTDs) leading to increased gene fusions and genomic rearrangements, the formation of fusion-related immunogenic neoantigens, and increased tumor-infiltrating lymphocytes and/or clonal expansion [59,60,68]. Wu [67] reported that 2 of 4 patients with biallelic CDK12 mutation exhibited an exceptional PSA response to anti-PD1 antibody. Other studies also found exceptional responses of two mCRPC patients to DNA-damaging therapies (bipolar androgen therapy that consist of periodical oscillation between castration levels and supraphysiological levels of testosterone and radium-223) after or in combination with immunotherapy (nivolumab and sipuleucel-T) [69,70]. The anecdotal evidence suggests that a subset of prostate cancer patients with CDK12 mutations may favorably respond to PD-1 immunotherapy. However, recent studies using immunohistochemistry analysis of TIL landscape revealed that human prostate tumors with biallelic CDK12 aberrations were predominantly enriched for immunosuppressive CD4+FOXP3- T lymphocytes but not for CD4+FOXP3+ or CD8+TILs [71,72]. This result suggests that immunotherapeutic strategies for better address the immunosuppressive tumor microenvironment are needed.

Other molecular alterations in prostate cancer may also affect the immune responses, which are clinically relevant. Calagua [73] reported that prostate tumors in a subset of aggressive localized prostate cancer cases express PD-L1 along with a high density of tumor infiltrating lymphocytes but without high microsatellite instability or CDK12 alterations. Exhausted progenitor CD8+ T cells and differentiated effector T cells as indicated by positive PD-1 and transcription factor TCF1 (encoded by TCF7) staining were founded in the tumor infiltrating lymphocytes, which are expandable by ICIs [74,75]. Areas within tumor tissue with MHC-II+ cells and CD8+TCF1+ T cells were also identified to be comparable with prostate cancer cases with dMMR, suggesting the existence of sufficient antigen-presenting cells (APC) niches. In addition, genomic losses of RB1, BRCA2, and CHD1 are common features of this subset of prostate cancer cases with potential immunogenicity.

Speckle-type pox virus and zinc finger protein (SPOP) are mutated in about 6-15% of localized and metastatic prostate cancer [76,77]. Evolution from SPOP-mutated to CHD1-deleted prostate cancer is considered a unique molecular subtype of prostate cancer [78]. SPOP is a component of a cullin-RING-based BCR (BTB-CUL3-RBX1) E3 ubiquitin-protein ligase complex that promotes the ubiquitination and degradation of PD-L1 [79]. Expression of Mutated SPOP would stabilize PD-L1. Prostate tumors with SPOP mutation exhibited increased PD-L1 expression and fewer tumor TILs [79]. Kaur also reported that homologous recombination deficiency scores but not pathogenic germline BRCA2- and ATM-mutations were associated with higher numbers of TIL, including both cytotoxic and regulatory T-cells. It is also interesting to test novel immunotherapeutic strategies in SPOP mutant prostate cancer.

Potentially underlying mechanisms for resistance to ICIs in prostate cancer

Most prostate cancer patients are resistant to immunotherapies, especially to immune checkpoint inhibitors. It has been estimated that about 5%-17% of unselected mCRPC patients respond to pembrolizumab monotherapy [29]. An ORR of 10% and 26% was only observed in mCRPC patients with and without previous taxane-based chemotherapy, respectively, after nivolumab plus ipilimumab treatment in a mCRPC phase II trial (CHECK MATE-650) [79]. A lower infiltration of T-cells, Low tumor mutational burden (TMB), low PD-L1 expression, and immunosuppressive TME have been considered as major hindrances of successful immunotherapy in prostate cancer.

TMB is the number of non-synonymous mutations present per megabase (mut/Mb) and has been used as biomarker for predicting response to ICIs. Although prostate cancer is characterized with a high rate of genomic instability and chromosomal rearrangements, TMBs in both localized and metastatic prostate cancer were estimated to be about 0.7~1.0 and 2.3~4.4 per Mb, respectively, which are significantly lower compared to other ICIs responsive cancers, such as bladder (7.1 per Mb) and melanoma (12.1 per Mb) [80-83]. Only 3-8.3% of metastatic prostate cancer tumors have a high TMB [84-86]. A low TMB in prostate cancer was associated with fewer mutated peptides (35 mutated peptides in prostate cancer versus 197 and 276 mutated proteins in lung adenocarcinoma and melanoma) [87,88], suggesting a poor collection of neoepitopes in prostate tumor may lead to less immune cell attraction to the tumor sites, epitope-MHC interactions and activation of TILs by APC. Therefore, a low TMB represents a significant hurdle for improving the efficacy of ICIs based immunotherapies in prostate cancer.

Compared to high-responsive tumors, such as melanoma and non-small cell lung carcinoma, for immunotherapies, the tumor immune microenvironment (TIME) in prostate cancer is generally featured with low frequency of TILs and high frequency of tumor-associated macrophages (TAMs). A unique and highly complex TIME in prostate cancer consists in different types of immune cells, working together to resist T-cell infiltration, even under treatment of ICIs [89]. Immune cell types estimated by deconvolution of RNA sequencing data from The Cancer Genome Atlas (TCGA) using by CIBERSORT algorithm revealed that total infiltrating T cells and mast cells were relatively less and infiltrating B cells, nature killer (NK) cells, macrophages and neutrophils are more compared with benign tissues [90]. Fewer infiltrating CD8+ T cells were consistently identified in prostate tumor tissues in several studies [90-92].

Local infiltration of CD8+ T cells in several types of tumors was shown to be associated with clinical benefits of ICIs with improved survival of cancer patients [93-97]. However a role of intratumoral CD8+ T cell density for predicting the clinical outcomes of prostate cancer patients remains debatable [98]. Previous studies have shown that a higher intratumoral CD8+ T cell density was associated with a poor prognosis of prostate cancer patients [99,100]. However most recent studies indicated that high levels of CD8+ T cell infiltration in radical prostatectomy specimens predicted a better survival and lower risks of biochemical recurrence and metastasis of prostate cancer patients [101,102].

In addition, increased number of immunosuppressive cells including TAMs, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSC) affect the antitumor response of CD8+ T cells [103-110]. Macrophages consist of 30 to 50% of infiltrating immune cells in tumor microenvironment. A subset of macrophages, such as M2-tumor associated macrophages (M2-TAMs) have shown to promote resistances to immunotherapy, chemotherapy, and radiotherapy through secretion of soluble factors and remodeling of cell matrix for promoting proliferation, angiogenesis, immunosuppression and tumor cell migration and invasion [103-110]. Several studies have shown that increased M2-TAM infiltration in prostate cancer TME was associated with worst clinicopathological features and prognosis or with more aggressive diseases with an estimated odds ratio of 1.93 (95% confidence interval: 1.23-3.03) [103-110]. Inhibition of androgen receptor (AR) signaling induces major changes in the immune landscape of prostate tumors, including increased infiltration of TAMs [111-116]. The increased numbers of TAMs in AR signaling inhibitors treated tumors predicted tumor recurrence and treatment resistance [111-116]. TAMs express PD-L1 and PD-1 leading to a decreased phagocytosis activity [118,119]. The decreased phagocytosis activity can be rescued by PD-1/PD-L1 blockade. M1 macrophages that are often stimulated by LPS or IFN-γ, etc. produce pro-inflammatory cytokines. Anti-PD-1/PD-L1 blockade induced an M1 macrophage polarization to reduce tumor burden [120,121]. MMR-deficient prostate cancer has been shown to have higher densities of TILs compared to MMR-proficient tumors [122]. Sena [123] reported that MMR-deficient prostate cancer with parenchymal brain metastases exhibited very few CD8+ tumor-infiltrating lymphocytes but highly enriched macrophages. Studies also showed that reduction of Tregs by anti-CTLA-4 antibodies in tumors was associated with tumor regression and dependent on FcγRIV-expressing macrophage-mediated cell depletion [124-128]. Taken together, these studies suggest that macrophages may also significantly contribute to the efficacies of ICIs based immunotherapies.

In addition, prostate tumors expressed high levels of chemokines, CCL2, CCL22, and CXCL12, to attract MDSCs, Tregs, and low levels of CTL/NK/Th1 cells-recruiting chemokines (CCL5, CXCL9, CXCL10). MDSCs also emerge in the context of castration resistance (129). MDSCs could be additional mechanism of resistance to ICIs in advanced CRPC.

The blockade of PD-1 and PD-L1 between CD8+ T cells and tumor cells is expected to restore antitumor responses induced by tumor-infiltrating CD8+ T cells [130]. Therefore, low level of PD-L1 expression in prostate cancer could also significantly limit efficacy of anti-PD1 based immunotherapy [131]. Due to tumor heterogeneity and different antibody clone selection, immunohistochemistry protocols, and scoring system, there is a large variability of PD-L1 expression in prostate tumor tissues. One study reported that 29% of acinar prostate cancer, 7% ductal prostate carcinoma and 46% of neuroendocrine carcinomas were PD-L1-positive [132-148]. While different cut-off values for positivity were used, 1369/4708 (29%) prostate cancer cases were overall positive. 687/2676 (26%) cases were PD-L1-positive if at least ≥1% of tumor cells were counted as positive, 93/1062 (9%) were positive for ≥5% of stained tumor cells, and 9/523 (2%) cases expressed PD-L1 on >50% of tumor cells [132-148]. In contrast to lower levels of PD-L1, the levels of PD-L2 expression were significantly higher across all 9393 radical prostatectomy samples. [149]. In addition, other immune checkpoints or targets may be clinically relevant in prostate cancer such as v-domain Ig suppressor of T-cell activation (VISTA), Poly (ADP-ribose) polymerase (PARP), MSH2 and MSH6 mutations, etc. [150-155]. To improve the immunogenicity of the “cold” prostate tumor cells, multiple targeting and combination approaches may be needed.

Racial difference in immune response in prostate cancer

Although prostate cancer is diagnosed at an earlier age and more aggressive in AA men [156-158], accumulating evidence suggest that AA prostate cancer respond favorably to immunotherapies, and specifically to cancer vaccines [159,160]. In the PROCEED trial/registry, AA men with mCRPC who were treated with the cancer vaccine, Sipuleucel-T, had a median nine-month of overall survival advantage over EA men [159,160]. Hawley [161] compared levels of circulating immune markers in AA and EA men with mCRPC who received sipuleucel-T. The results showed that AA men had statistically significantly higher levels of Th2-type (IL-4, IL-10, and IL-13) and inflammatory cytokines (IL-2, IL-12, and IL-6) compared with prostate-specific antigen-matched EA men both at baseline and 52 weeks after sipuleucel-T and that there are no differences in the antigen-specific T-cell response and the humoral responses to the immunizing antigen PA2024 and select secondary antigens.

A study performed by Calagua [73] showed a significant association between AA and PD-L1 overexpression in prostate cancer (26% in AA vs. 12% in EA), suggesting that AA men with prostate cancer may respond more favorably to anti-PD1 based immunotherapies. AA men were also shown to have lower DNA damage repair (DDR) activity compared to EA men [162-165]. Defective DNA damage responses are associated with improved radiation response and tumor immunogenicity [46,166], which may implicate a combined approach of radiotherapy and immune therapy in AA men.

Prominent differences in tumor immunobiology between AA vs. EA men have been reported in several independent gene expression profiling studies [167-169]. For example, the gene expression study by Wallace revealed significantly different profiles of immune-related genes when comparing 69 tumors from AA and EA patients; autoimmune disease modulators, such as PTPN22 and components of the HLA complex, and key genes in antigen presentation were expressed at higher levels in AA tumors [168]. Kinseth [169] examined the differences in gene expression between AA and EA PCa by matching for age and pathological stage or Gleason scores as well as tumor-cell content and stroma-cell content. Striking differences in gene expression were observed in the stroma of AA patients relative to EA; 1016 genes with significant differences between the expression of EA and AA patients were observed. The vast majority (82%) of significant differences were downregulated. The down-regulated genes included many immune cell modulators compared to EA stroma such as interleukins (IL) -2, -4, -5, -6, -7, -10, -13, -15 and -22. Emerging data also have shown that levels of cytokines, IFNα, IFNγ, and TNFα signaling, ILs, and epithelial to mesenchymal (EMT) transition signaling, as well as tumor infiltrating lymphocytes were elevated in AA men, which suggest uniquely inflammatory phenotype in AA prostate cancer [163]. Weiner identified an enrichment of plasma cells in primary prostate tumors of AA men or men of African ancestry and elevated expression of NK cell activity markers and IgG [170], suggesting role of plasma cells in immune responses of AA men. Awasthi and his colleagues [163] have analyzed whole transcriptome data from the Decipher GRID registry and found that differentially expressed genes in major immune pathways were significantly enriched in AA compared to EA prostate cancer. In addition, IFN-induced transmembrane protein 3 (IFITM3) was validated in TCGA data base as a biomarker that was significantly associated with increased risk of prostate cancer recurrence for AA men. Tang [171] reported that AA prostate cancer patients with IFNL4 rs368234815-DG and IFNL4 rs12979860-T germline variants have poorer overall survival after prostatectomy surgery and that IFNL4 rs368234815-DG germline variant was associated with IFN-related DNA damage resistance signature.

Although many clinical trials of ICIs or in combination with other therapeutic approaches are ongoing, AA men are currently underrepresented in most of the clinical trials [172,173]. There is a need for enhancing accrual of more AA men to clinical trials via prespecified enrolling clinical sites and by overcoming socioeconomic and cultural barriers [172,173]. Clinical trials that specifically focused on the treatment response of AA men with metastatic prostate cancer will greatly facilitate our understanding of racial differences in the immune response. There are ongoing NCI efforts with sponsoring trials for enrollment of minority groups of patients.

Vaccine based treatments

The majority of immunotherapy vaccinations available for prostate cancer can be considered to be experimental. Sipuleucel-T currently the only FDA approved vaccine designated for usage towards prostate cancer and be said the most effective in clinical usage [174,175]. Sipuleucel-T is an autologous active cellular immunotherapy vaccine that primarily consists of autologous peripheral-blood mononuclear cells with APCs which are activated when exposed to PA2024, a recombinant fusion protein of PAP and costimulatory GM-CSF. A phase III trial (IMPACT: NCT00065442) in mCRPC patients demonstrated an improved OS by 4.1 months and a 22% reduction of relative mortality risk. However, only minimal antitumor responses were observed [175].

DNA vaccines have been examined largely in animals as a potential treatment for cancer. Their use in human remains controversial given the risk vs. benefit [176]. They offer a new approach over other anti-tumor vaccinations in terms of ease of use and the absence of infectious agents. Presently various phase 1 clinic trials are underway for prostate cancer DNA, namely NCT02411786 by Madison Vaccines Inc which encodes androgen receptors pTVG-AR, MVI-118 [177]. INO-5150 by Inovio Pharmceuticals is a dual-antigen DNA vaccine that utilizes parts of prostate-specific membrane antigen and the prostate specific antigen, which underwent phase I/II trial in biochemically relapsed prostate cancer patients [178].

PROSTVAC is an off the shelf vaccine that uses a recombinant strain of vaccinia paired with foxpowl vector boosts, transgenes, and co-stimulatory molecules to elicit an immune response from the body [179]. Patients that were treated with PROSTVAC have shown an increase in PSA-specific T-cell levels [179]. Two phase II studies of PROSTVAC have demonstrated its potential efficacy in treating mCRPC patients. One hundred twenty-five patients with mCRPC and a Gleason score of ≤7 were randomly given either PROSTVAC, or a placebo [180]. Patients treated with PROSTVAC demonstrated a median survival rate of 24.4 months; while patients treated with the placebo had a survival rate of 16.3 months [180]. A recent phase III study of PROSTVAC was conducted to follow up on this hypothesis but did not show any significant clinical benefit [181,182]. The vaccine was reported to be well tolerated and eliciting an immune response, however there was minimal survival benefit [182].

GVAX uses the whole prostate cancer cell genetically modified to secrete the immune stimulatory cytokine granulocyte-macrophage colony-stimulating factor and allows the tumor cells themselves to be used as the antigen source for immunotherapy [183]. GVAX has shown to be a safe and potent cytokine and has elicited a high immune response dependent on the dosage. Patients only exhibited flu-like symptoms and a fever when treated. However, considering several failed phase 3 trials of this vaccination, further experimentation on it has largely been abandoned [184,185].

Adoptive cell therapy

Adoptive Cell Therapy (ACT) has been shown to be effective in treating metastatic melanoma [186,187]. This treatment involves the usage of T-lymphocytes specifically engineered to target specific viruses or tumors. Through the isolation and modification of patient T-lymphocytes with specific antigen receptors, followed by subsequent reinfusion, it is possible for patients produce an immunization-like response towards specific cancer antigens. Chimeric antigen receptors (CAR) allow for the production of artificial T-cell receptors for the purposes of ACT [188].

Epithelial cell adhesion molecule (EpCAM) targeting T-cells modified with CAR have shown to be effective in a wide range of cancer immunotherapies involving this stem cell antigen [189]. Studies performed on human prostate cancer cells with low expression levels of EpCAM have shown significant effectiveness in inhibiting tumor growth both in vitro and in vito [189]. The Natural Killer Group 2D (NKG2D) receptor has also been shown to a promising target for CAR T-cell therapy. When paired with the IL-7 gene it is shown to be effective in prostate cancer treatment [190]. To target prostate cancer more specifically, CAR-T cells were commonly generated against prostate-specific membrane antigen (PSMA). In a first in-human phase 1 trial of PSMA targeting CAR T cells armored with a dominant-negative TGF-β receptor (NCT03089203) in CRPC patients, 5 of 13 patients developed cytokine-release (CRS) at grade 2 or higher and 4 had decreases of ≥30% in PSA. One patient reached a >98% reduction in PSA with evidence of substantial clonal CAR T cell expansion. However, this one patient developed enterococcal sepsis 30 days after infusion, leading to multi-organ dysfunction and death [191]. Another ongoing clinical trial of PSMA-targeting CAR in mCRPC patients, 3 of 9 patients reached decreases of >50% in PSA and improvements in PSMA positron-emission-tomography imaging [192]. Three patients had CRS of grades 1-2 [192]. One patient experienced a complete clearance of measurable disease for over 5 months [192]. The results from these studies are promising, but CAR-T cell therapy still face many challenges or difficulties. Overcoming the immunosuppressive TME that are enriched with immunosuppressive cytokines and growth factors, TAMs, Treg, and MDSC and potential toxicities are some of the major limitations in CAR-T therapy.

Another method of ACT can be seen in TIL therapy, which involves the examination of specific lymphocytes located around the tumor. T-cells that best identify malignant tumor cells are treated and encouraged to proliferate around the tumor. Due to the T-cell exclusive nature of prostate cancer, it is often challenging to effectively incorporate TILs based immunotherapy into prostate cancer treatments [193]. This can be attributed to a lack of genomic complexity within prostate cancer cells compared to other cancers [194,195]. Recent experiments on TILs in prostate cancer have indicated the potential for the viable extraction of functional and tumor reactive TIL within prostate cancer. In a study, twenty-eight prostate-TIL cultures were successfully extracted and expanded in a laboratory setting. The extracted TIL displayed an expansion frequency of roughly 50% across all samples. Analysis revealed a clear expression of chemokine receptors after expansion. Further studies into this form of therapy can potentially open more modalities in patient treatment [196].

A major challenge in CAR T-cell and TILs therapy is proliferating for long periods of time in immunosuppressive environments. As such, there is a push towards research that increases survival rates of CAR T-cells; notably through the incorporation of the TILs 4-1BB and CD137 receptor respectively [197,198]. The inclusion of such therapies into patient care provides an efficient treatment method that suffers from fewer side effects associated with other methods of cancer treatment.

Bispecific T cell engager

Bispecific T cell engager (BiTE) antibodies can target PSMA on prostate cancer cells and engage T cell via CD3 receptor leading to T cell activation. AMG 212 (pasotuxizumab) demonstrated encouraging results in a phase I trial with dose-dependent PSA reductions and measurable tumor responses in approximately one-third of the 68 patients who were enrolled after progression on androgen deprivation therapy with abiraterone or enzalutamide and at least one taxane chemotherapy [199]. Limitation of this study included development of drug-neutralizing antibodies with subcutaneous injection and short serum half-life of the molecule. In order to overcome these limitations, AMG212 has been modified to AMG 160 with ongoing studies using IV formulation (NCT04631601) and half-life extended BiTE molecule [200].

Other target candidates on prostate cancer cells for BiTE therapy are being evaluated including the six-transmembrane epithelial antigen of prostate (STEAP), human carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5, also known as CEA) and delta-like protein 3 (DLL3), which are upregulated in different subtypes of prostate cancer [201,202].

Conclusion

Prostate cancer exhibits many immunosuppressive characteristics associated with TME, low TMB, low expression of PD-L1 and sparse T-cell infiltration and therefore, prostate cancer has been considered as an immunologically “cold” tumor. Nevertheless, immunotherapy remains to be a promising treatment at least in a subset of prostate cancer patients. Prostate cancer tumors with high MSI/dMMR or CDK12 mutations can be more responsive to ICIs in clinics. Clinical data suggests higher degree of benefit in AA patients with prostate cancer treated with Sipuleucel-T. CAR-T therapy and BiTEs have shown some early encouraging clinical results with ongoing clinical trials evaluating the role of these treatments. Understanding resistant mechanisms to ICIs related to prostate TME and identification of new immune targets, could bring a new promising immune therapeutic approach for advanced prostate cancer and lead to extension of patent’s lifespan.

Acknowledgements

This work was supported in part by NIH award 1R01CA260351-01A1, DOD/Prostate Cancer Research award PC181016 and VA merit award I01BX005105 (to X. Zi.).

Disclosure of conflict of interest

None.

References

  • 1.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics. CA Cancer J Clin. 2022;72:7–33. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
  • 2.Loeb S, Catalona WJ. The prostate health index: a new test for the detection of prostate cancer. Ther Adv Urol. 2014;6:74–7. doi: 10.1177/1756287213513488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Askari M, Yazdani A, Yazdani M, Izadpanahi MH. Serum levels of total and urine level of PCA3 in patients with benign prostatic hyperplasia and prostate cancer. Am J Clin Exp Urol. 2020;8:43–47. [PMC free article] [PubMed] [Google Scholar]
  • 4.Siddiqui MM, Rais-Bahrami S, Turkbey B, George AK, Rothwax J, Shakir N, Okoro C, Raskolnikov D, Parnes HL, Linehan WM, Merino MJ, Simon RM, Choyke PL, Wood BJ, Pinto PA. Comparison of MR/ultrasound fusion-guided biopsy with ultrasound-guided biopsy for the diagnosis of prostate cancer. JAMA. 2015;313:390–7. doi: 10.1001/jama.2014.17942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kantoff PW, Higano CS, Shore ND, Berger R, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer RD, Sims RB, Xu Y, Frohlich MW, Schellhammer PF. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;36:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 6.Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, Krainer M, Houede N, Santos R, Mahammedi H, Ng S, Maio M, Franke FA, Sundar S, Agarwal N, Bergman AM, Ciuleanu TE, Korbenfeld E, Sengeløv L, Hansen S, Logothetis C, Beer TM, McHenry MB, Gagnier P, Liu D, Gerritsen WR. 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. doi: 10.1016/S1470-2045(14)70189-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, Poddubskaya E, Borghaei H, Felip E, Paz-Ares L, Pluzanski A, Reckamp KL, Burgio MA, Kohlhäeufl M, Waterhouse D, Barlesi F, Antonia S, Arrieta O, Fayette J, Crinò L, Rizvi N, Reck M, Hellmann MD, Geese WJ, Li A, Blackwood-Chirchir A, Healey D, Brahmer J, Eberhardt WEE. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase iii trials (CheckMate 017 and CheckMate 057) J. Clin. Oncol. 2017;35:3924–3933. doi: 10.1200/JCO.2017.74.3062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–13. doi: 10.1056/NEJMoa1510665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm MO, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277–1290. doi: 10.1056/NEJMoa1712126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Powles T, Csőszi T, Özgüroğlu M, Matsubara N, Géczi L, Cheng SY, Fradet Y, Oudard S, Vulsteke C, Morales Barrera R, Fléchon A, Gunduz S, Loriot Y, Rodriguez-Vida A, Mamtani R, Yu EY, Nam K, Imai K, Homet Moreno B, Alva A. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): a randomised, open-label, phase 3 trial. Lancet Oncol. 2021;22:931–945. doi: 10.1016/S1470-2045(21)00152-2. [DOI] [PubMed] [Google Scholar]
  • 11.Petrylak DP, de Wit R, Chi KN, Drakaki A, Sternberg CN, Nishiyama H, Castellano D, Hussain S, Fléchon A, Bamias A, Yu EY, van der Heijden MS, Matsubara N, Alekseev B, Necchi A, Géczi L, Ou YC, Coskun HS, Su WP, Hegemann M, Percent IJ, Lee JL, Tucci M, Semenov A, Laestadius F, Peer A, Tortora G, Safina S, Del Muro XG, Rodriguez-Vida A, Cicin I, Harputluoglu H, Widau RC, Liepa AM, Walgren RA, Hamid O, Zimmermann AH, Bell-McGuinn KM, Powles T. Ramucirumab plus docetaxel versus placebo plus docetaxel in patients with locally advanced or metastatic urothelial carcinoma after platinum-based therapy (RANGE): a randomised, double-blind, phase 3 trial. Lancet. 2017;390:2266–2277. doi: 10.1016/S0140-6736(17)32365-6. [DOI] [PubMed] [Google Scholar]
  • 12.Lee NY, Ferris RL, Psyrri A, Haddad RI, Tahara M, Bourhis J, Harrington K, Chang PM, Lin JC, Razaq MA, Teixeira MM, Lövey J, Chamois J, Rueda A, Hu C, Dunn LA, Dvorkin MV, De Beukelaer S, Pavlov D, Thurm H, Cohen E. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo controlled, multicentre, phase 3 trial. Lancet Oncol. 2021;22:450–462. doi: 10.1016/S1470-2045(20)30737-3. [DOI] [PubMed] [Google Scholar]
  • 13.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, de la Cruz Merino L, Khattak MA, Schadendorf D, Long GV, Ascierto PA, Mandala M, De Galitiis F, Haydon A, Dummer R, Grob JJ, Robert C, Carlino MS, Mohr P, Poklepovic A, Sondak VK, Scolyer RA, Kirkwood JM, Chen K, Diede SJ, Ahsan S, Ibrahim N, Eggermont AMM KEYNOTE-716 Investigators. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet. 2022;399:1718–1729. doi: 10.1016/S0140-6736(22)00562-1. [DOI] [PubMed] [Google Scholar]
  • 15.Bilusic M, Madan RA, Gulley JL. Immunotherapy of prostate cancer: facts and hopes. Clin Cancer Res. 2017;23:6764–6770. doi: 10.1158/1078-0432.CCR-17-0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Markowski MC, Shenderov E, Eisenberger MA, Kachhap S, Pardoll DM, Denmeade SR, Antonarakis ES. Extreme responses to immune checkpoint blockade following bipolar androgen therapy and enzalutamide in patients with metastatic castration resistant prostate cancer. Prostate. 2020;80:407–411. doi: 10.1002/pros.23955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fellner C. Ipilimumab (Yervoy) prolongs survival in advanced melanoma: serious side effects and a hefty price tag may limit its use. P T. 2012;37:503–512. [PMC free article] [PubMed] [Google Scholar]
  • 18.Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, Baker J, Jeffrey LE, Kaur S, Briggs Z, Hou TZ, Futter CE, Anderson G, Walker LS, Sansom DM. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332:600–3. doi: 10.1126/science.1202947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Small EJ, Tchekmedyian NS, Rini BI, Fong L, Lowy I, Allison JP. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin Cancer Res. 2007;13:1810–5. doi: 10.1158/1078-0432.CCR-06-2318. [DOI] [PubMed] [Google Scholar]
  • 20.McNeel DG, Smith HA, Eickhoff JC, Lang JM, Staab MJ, Wilding G, Liu G. Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer. Cancer Immunol Immunother. 2012;61:1137–1147. doi: 10.1007/s00262-011-1193-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jochems C, Tucker JA, Tsang KY, Madan RA, Dahut WL, Liewehr DJ, Steinberg SM, Gulley JL, Schlom J. A combination trial of vaccine plus ipilimumab in metastatic castration-resistant prostate cancer patients: immune correlates. Cancer Immunol Immunother. 2014;63:407–18. doi: 10.1007/s00262-014-1524-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hegde A, Jayaprakash P, Couillault CA, Piha-Paul S, Karp D, Rodon J, Pant S, Fu S, Dumbrava EE, Yap TA, Subbiah V, Bhosale P, Coarfa C, Higgins JP, Williams ET, Wilson TF, Lim J, Meric-Bernstam F, Sumner E, Zain H, Nguyen D, Nguyen LM, Rajapakshe K, Curran MA, Hong DS. A phase i dose-escalation study to evaluate the safety and tolerability of evofosfamide in combination with Ipilimumab in advanced solid malignancies. Clin Cancer Res. 2021;27:3050–3060. doi: 10.1158/1078-0432.CCR-20-4118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fong L, Kwek SS, O’Brien S, Kavanagh B, McNeel DG, Weinberg V, Lin AM, Rosenberg J, Ryan CJ, Rini BI, Small EJ. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609–15. doi: 10.1158/0008-5472.CAN-08-3529. [DOI] [PubMed] [Google Scholar]
  • 24.Slovin SF, Higano CS, Hamid O, Tejwani S, Harzstark A, Alumkal JJ, Scher HI, Chin K, Gagnier P, McHenry MB, Beer TM. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013;24:1813–1821. doi: 10.1093/annonc/mdt107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fizazi K, Drake CG, Beer TM, Kwon ED, Scher HI, Gerritsen WR, Bossi A, den Eertwegh AJMV, Krainer M, Houede N, Santos R, Mahammedi H, Ng S, Danielli R, Franke FA, Sundar S, Agarwal N, Bergman AM, Ciuleanu TE, Korbenfeld E, Sengeløv L, Hansen S, McHenry MB, Chen A, Logothetis C CA184-043, Investigators. Final analysis of the Ipilimumab versus placebo following radiotherapy phase III trial in postdocetaxel metastatic castration-resistant prostate cancer identifies an excess of long-term survivors. Eur Urol. 2020;78:822–830. doi: 10.1016/j.eururo.2020.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, Ganju V, Polikoff J, Saad F, Humanski P, Piulats JM, Gonzalez Mella P, Ng SS, Jaeger D, Parnis FX, Franke FA, Puente J, Carvajal R, Sengeløv L, McHenry MB, Varma A, van den Eertwegh AJ, Gerritsen W. 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. doi: 10.1200/JCO.2016.69.1584. [DOI] [PubMed] [Google Scholar]
  • 27.Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, Zhao H, Chen J, Chen H, Efstathiou E, Troncoso P, Allison JP, Logothetis CJ, Wistuba II, Sepulveda MA, Sun J, Wargo J, Blando J, Sharma P. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23:551–5. doi: 10.1038/nm.4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sharma P, Pachynski RK, Narayan V, Flechon A, Gravis G, Galsky MD, Mahammedi H, Patneik A, Subudhi SK, Ciprotti M, Simsek B, Saci A, Hu Y, Han GC, Fizazi K. Initial results from a phase II study of nivolumab (NIVO) plus ipilimumab (IPI) for the treatment of metastatic castration-resistant prostate cancer (mCRPC; CheckMate 650) J. Clin. Oncol. 2019;37:142. [Google Scholar]
  • 29.Anti-PD-1-CTLA4 combo hits prostate cancer. Cancer Discov. 2019;9:569–70. doi: 10.1158/2159-8290.CD-NB2019-039. [DOI] [PubMed] [Google Scholar]
  • 30.Shenderov E, Boudadi K, Fu W, Wang H, Sullivan R, Jordan A, Dowling D, Harb R, Schonhoft J, Jendrisak A, Carducci MA, Eisenberger MA, Eshleman JR, Luo J, Drake CG, Pardoll DM, Antonarakis ES. Nivolumab plus ipilimumab, with or without enzalutamide, in AR-V7-expressing metastatic castration-resistant prostate cancer: a phase-2 nonrandomized clinical trial. Prostate. 2021;81:326–338. doi: 10.1002/pros.24110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, Vaishampayan U, Berger R, Sezer A, Alanko T, de Wit R, Li C, Omlin A, Procopio G, Fukasawa S, Tabata KI, Park SH, Feyerabend S, Drake CG, Wu H, Qiu P, Kim J, Poehlein C, de Bono JS. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J. Clin. Oncol. 2020;38:395–405. doi: 10.1200/JCO.19.01638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hansen AR, Massard C, Ott PA, Haas NB, Lopez JS, Ejadi S, Wallmark JM, Keam B, Delord JP, Aggarwal R, Gould M, Yang P, Keefe SM, Piha-Paul SA. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol. 2018;29:1807–1813. doi: 10.1093/annonc/mdy232. [DOI] [PubMed] [Google Scholar]
  • 33.Yu EY, Kolinsky MP, Berry WR, Retz M, Mourey L, Piulats JM, Appleman LJ, Romano E, Gravis G, Gurney H, Bögemann M, Emmenegger U, Joshua AM, Linch M, Sridhar S, Conter HJ, Laguerre B, Massard C, Li XT, Schloss C, Poehlein CH, de Bono JS. Pembrolizumab plus docetaxel and prednisone in patients with metastatic castration-resistant prostate cancer: long-term results from the phase 1b/2 KEYNOTE-365 Cohort B study. Eur Urol. 2022;82:22–30. doi: 10.1016/j.eururo.2022.02.023. [DOI] [PubMed] [Google Scholar]
  • 34.Petrylak DP, Ratta R, Gafanov R, Facchini G, Piulats JM, Kramer G, Flaig TW, Chandana SR, Li B, Burgents J, Fizazi K. Phase III study of pembrolizumab plus docetaxel for metastatic castration-resistant prostate cancer. Future Oncol. 2021;17:3291–3299. doi: 10.2217/fon-2020-1133. [DOI] [PubMed] [Google Scholar]
  • 35.Graff JN, Beer TM, Alumkal JJ, Slottke RE, Redmond WL, Thomas GV, Thompson RF, Wood MA, Koguchi Y, Chen Y, Latour E, Bergan RC, Drake CG, Moran AE. A phase II single-arm study of pembrolizumab with enzalutamide in men with metastatic castration-resistant prostate cancer progressing on enzalutamide alone. J Immunother Cancer. 2020;8:e000642. doi: 10.1136/jitc-2020-000642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Graff JN, Liang LW, Kim J, Stenzl A. A Phase III study of pembrolizumab plus enzalutamide for metastatic castration-resistant prostate cancer. Future Oncol. 2021;17:3017–3026. doi: 10.2217/fon-2020-1008. [DOI] [PubMed] [Google Scholar]
  • 37.Fizazi K, González Mella P, Castellano D, Minatta JN, Rezazadeh Kalebasty A, Shaffer D, Vázquez Limón JC, Sánchez López HM, Armstrong AJ, Horvath L, Bastos DA, Amin NP, Li J, Unsal-Kacmaz K, Retz M, Saad F, Petrylak DP, Pachynski RK. Nivolumab plus docetaxel in patients with chemotherapy-naïve metastatic castration-resistant prostate cancer: results from the phase II CheckMate 9KD trial. Eur J Cancer. 2022;160:61–71. doi: 10.1016/j.ejca.2021.09.043. [DOI] [PubMed] [Google Scholar]
  • 38.McNeel DG, Eickhoff JC, Wargowski E, Johnson LE, Kyriakopoulos CE, Emamekhoo H, Lang JM, Brennan MJ, Liu G. Phase 2 trial of T-cell activation using MVI-816 and pembrolizumab in patients with metastatic, castration-resistant prostate cancer (mCRPC) J Immunother Cancer. 2022;10:e004198. doi: 10.1136/jitc-2021-004198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.McNeel DG, Eickhoff JC, Johnson LE, Roth AR, Perk TG, Fong L, Antonarakis ES, Wargowski E, Jeraj R, Liu G. Phase II trial of a DNA vaccine encoding prostatic acid phosphatase (pTVG-HP [MVI-816]) in patients with progressive, nonmetastatic, castration-sensitive prostate cancer. J. Clin. Oncol. 2019;37:3507–3517. doi: 10.1200/JCO.19.01701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stein MN, Fong L, Tutrone R, Mega A, Lam ET, Parsi M, Vangala S, Gutierrez AA, Haas NB. ADXS31-142 immunotherapy ± pembrolizumab treatment for metastatic castration-resistant prostate cancer: open-label phase I/II KEYNOTE-046 study. Oncologist. 2022;27:453–461. doi: 10.1093/oncolo/oyac048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ross AE, Hurley PJ, Tran PT, Rowe SP, Benzon B, Neal TO, Chapman C, Harb R, Milman Y, Trock BJ, Drake CG, Antonarakis ES. A pilot trial of pembrolizumab plus prostatic cryotherapy for men with newly diagnosed oligometastatic hormone-sensitive prostate cancer. Prostate Cancer Prostatic Dis. 2020;23:184–193. doi: 10.1038/s41391-019-0176-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Petrylak DP, Loriot Y, Shaffer DR, Braiteh F, Powderly J, Harshman LC, Conkling P, Delord JP, Gordon M, Kim JW, Sarkar I, Yuen K, Kadel EE 3rd, Mariathasan S, O’Hear C, Narayanan S, Fassò M, Carroll S, Powles T. Safety and clinical activity of Atezolizumab in patients with metastatic castration-resistant prostate cancer: a phase I study. Clin Cancer Res. 2021;27:3360–3369. doi: 10.1158/1078-0432.CCR-20-1981. [DOI] [PubMed] [Google Scholar]
  • 43.Powles T, Yuen KC, Gillessen S, Kadel EE 3rd, Rathkopf D, Matsubara N, Drake CG, Fizazi K, Piulats JM, Wysocki PJ, Buchschacher GL Jr, Alekseev B, Mellado B, Karaszewska B, Doss JF, Rasuo G, Datye A, Mariathasan S, Williams P, Sweeney CJ. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat Med. 2022;28:144–153. doi: 10.1038/s41591-021-01600-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Fong L, Morris MJ, Sartor O, Higano CS, Pagliaro L, Alva A, Appleman LJ, Tan W, Vaishampayan U, Porcu R, Tayama D, Kadel EE 3rd, Yuen KC, Datye A, Armstrong AJ, Petrylak DP. A Phase Ib study of Atezolizumab with Radium-223 Dichloride in men with metastatic castration-resistant prostate cancer. Clin Cancer Res. 2021;27:4746–4756. doi: 10.1158/1078-0432.CCR-21-0063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dorff T, Hirasawa Y, Acoba J, Pagano I, Tamura D, Pal S, Zhang M, Waitz R, Dhal A, Haynes W, Shon J, Scholz M, Furuya H, Chan OTM, Huang J, Rosser C. Phase Ib study of patients with metastatic castrate-resistant prostate cancer treated with different sequencing regimens of atezolizumab and sipuleucel-T. J Immunother Cancer. 2021;9:e002931. doi: 10.1136/jitc-2021-002931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Agarwal N, Azad A, Carles J, Chowdhury S, McGregor B, Merseburger AS, Oudard S, Saad F, Soares A, Benzaghou F, Kerloeguen Y, Kimura A, Mohamed N, Panneerselvam A, Wang F, Pal S. A phase III, randomized, open-label study (CONTACT-02) of cabozantinib plus atezolizumab versus second novel hormone therapy in patients with metastatic castration-resistant prostate cancer. Future Oncol. 2022;18:1185–1198. doi: 10.2217/fon-2021-1096. [DOI] [PubMed] [Google Scholar]
  • 47.Kwan EM, Spain L, Anton A, Gan CL, Garrett L, Chang D, Liow E, Bennett C, Zheng T, Yu J, Dai C, Du P, Jia S, Fettke H, Abou-Seif C, Kothari G, Shaw M, Parente P, Pezaro C, Tran B, Siva S, Azad AA. Avelumab combined with stereotactic ablative body radiotherapy in metastatic castration-resistant prostate cancer: the phase 2 ICE-PAC clinical trial. Eur Urol. 2022;81:253–262. doi: 10.1016/j.eururo.2021.08.011. [DOI] [PubMed] [Google Scholar]
  • 48.Brown LC, Halabi S, Somarelli JA, Humeniuk M, Wu Y, Oyekunle T, Howard L, Huang J, Anand M, Davies C, Patel P, Staats J, Weinhold KJ, Harrison MR, Zhang T, George DJ, Armstrong AJ. A phase 2 trial of avelumab in men with aggressive-variant or neuroendocrine prostate cancer. Prostate Cancer Prostatic Dis. 2022:1–8. doi: 10.1038/s41391-022-00524-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Karzai F, VanderWeele D, Madan RA, Owens H, Cordes LM, Hankin A, Couvillon A, Nichols E, Bilusic M, Beshiri ML, Kelly K, Krishnasamy V, Lee S, Lee MJ, Yuno A, Trepel JB, Merino MJ, Dittamore R, Marté J, Donahue RN, Schlom J, Killian KJ, Meltzer PS, Steinberg SM, Gulley JL, Lee JM, Dahut WL. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer. 2018;6:141. doi: 10.1186/s40425-018-0463-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhao D, Cai L, Lu X, Liang X, Li J, Chen P, Ittmann M, Shang X, Jiang S, Li H, Meng C, Flores I, Song JH, Horner JW, Lan Z, Wu CJ, Li J, Chang Q, Chen KC, Wang G, Deng P, Spring DJ, Wang YA, DePinho RA. Chromatin regulator CHD1 remodels the immunosuppressive tumor microenvironment in PTEN-deficient prostate cancer. Cancer Discov. 2020;10:1374–1387. doi: 10.1158/2159-8290.CD-19-1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Majidpoor J, Mortezaee K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin Immunol. 2021;226:108707. doi: 10.1016/j.clim.2021.108707. [DOI] [PubMed] [Google Scholar]
  • 52.Jafari S, Molavi O, Kahroba H, Hejazi MS, Maleki-Dizaji N, Barghi S, Kiaie SH, Jadidi-Niaragh F. Clinical application of immune checkpoints in targeted immunotherapy of prostate cancer. Cell Mol Life Sci. 2020;77:3693–3710. doi: 10.1007/s00018-020-03459-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Schöffski P, Tan DSW, Martín M, Ochoa-de-Olza M, Sarantopoulos J, Carvajal RD, Kyi C, Esaki T, Prawira A, Akerley W, De Braud F, Hui R, Zhang T, Soo RA, Maur M, Weickhardt A, Krauss J, Deschler-Baier B, Lau A, Samant TS, Longmire T, Chowdhury NR, Sabatos-Peyton CA, Patel N, Ramesh R, Hu T, Carion A, Gusenleitner D, Yerramilli-Rao P, Askoxylakis V, Kwak EL, Hong DS. Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) ± anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J Immunother Cancer. 2022;10:e003776. doi: 10.1136/jitc-2021-003776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schuhmacher J, Heidu S, Balchen T, Richardson JR, Schmeltz C, Sonne J, Schweiker J, Rammensee HG, Thor Straten P, Røder MA, Brasso K, Gouttefangeas C. Vaccination against RhoC induces long-lasting immune responses in patients with prostate cancer: results from a phase I/II clinical trial. J Immunother Cancer. 2020;8:e001157. doi: 10.1136/jitc-2020-001157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhu H, Wang M, Du Y, Liu X, Weng X, Li C. 4-1BBL has a possible role in mediating castration-resistant conversion of prostate cancer via up-regulation of androgen receptor. J Cancer. 2019;10:2464–2471. doi: 10.7150/jca.29648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 2019;25:3753–3758. doi: 10.1158/1078-0432.CCR-18-4070. [DOI] [PubMed] [Google Scholar]
  • 57.Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, Rathkopf D, Morris MJ, Danila DC, Slovin SF, Carbone E, Barnett ES, Hullings M, Hechtman JF, Zehir A, Shia J, Jonsson P, Stadler ZK, Srinivasan P, Laudone VP, Reuter V, Wolchok JD, Socci ND, Taylor BS, Berger MF, Kantoff PW, Sawyers CL, Schultz N, Solit DB, Gopalan A, Scher HI. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 2019;5:471–478. doi: 10.1001/jamaoncol.2018.5801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Aguiar-Ibáñez R, Hardern C, van Hees F, Lee D, Patel A, Chhabra N, Baluni G, Amonkar M, Lai Y, Xu R, Massaad R, Fogelman D. Cost-effectiveness of pembrolizumab for the first-line treatment of patients with unresectable or metastatic MSI-H/dMMR colorectal cancer in the United States. J Med Econ. 2022;25:469–480. doi: 10.1080/13696998.2022.2043634. [DOI] [PubMed] [Google Scholar]
  • 59.Bajrami I, Frankum JR, Konde A, Miller RE, Rehman FL, Brough R, Campbell J, Sims D, Rafiq R, Hooper S, Chen L, Kozarewa I, Assiotis I, Fenwick K, Natrajan R, Lord CJ, Ashworth A. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 2014;74:287–97. doi: 10.1158/0008-5472.CAN-13-2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu H, Liu K, Dong Z. Targeting CDK12 for cancer therapy: function, mechanism, and drug discovery. Cancer Res. 2021;81:18. doi: 10.1158/0008-5472.CAN-20-2245. [DOI] [PubMed] [Google Scholar]
  • 61.Warner E, Herberts C, Fu S, Yip S, Wong A, Wang G, Ritch E, Murtha AJ, Vandekerkhove G, Fonseca NM, Angeles A, Beigi A, Schönlau E, Beja K, Annala M, Khalaf D, Chi KN, Wyatt AW. BRCA2, ATM, and CDK12 defects differentially shape prostate tumor driver genomics and clinical aggression. Clin Cancer Res. 2021;27:1650–1662. doi: 10.1158/1078-0432.CCR-20-3708. [DOI] [PubMed] [Google Scholar]
  • 62.Nizialek E, Lim SJ, Wang H, Isaacsson Velho P, Yegnasubramanian S, Antonarakis ES. Genomic profiles and clinical outcomes in primary versus secondary metastatic hormone-sensitive prostate cancer. Prostate. 2021;81:572–579. doi: 10.1002/pros.24135. [DOI] [PubMed] [Google Scholar]
  • 63.Necchi A, Cucchiara V, Grivas P, Bratslavsky G, Jacob J, Spiess PE, Sokol ES, Killian JK, Lin D, Ramkissoon S, Huang RSP, Madison RW, Venstrom JM, Schrock AB, Danziger N, Decker B, Gjoerup O, Graf RP, Oxnard GR, Tukachinsky H, Ross JS. Contrasting genomic profiles from metastatic sites, primary tumors, and liquid biopsies of advanced prostate cancer. Cancer. 2021;127:4557–4564. doi: 10.1002/cncr.33865. [DOI] [PubMed] [Google Scholar]
  • 64.Chung JH, Dewal N, Sokol E, Mathew P, Whitehead R, Millis SZ, Frampton GM, Bratslavsky G, Pal SK, Lee RJ, Necchi A, Gregg JP, Lara P Jr, Antonarakis ES, Miller VA, Ross JS, Ali SM, Agarwal N. Prospective comprehensive genomic profiling of primary and metastatic prostate tumors. JCO Precis Oncol. 2019;3:PO.18.00283. doi: 10.1200/PO.18.00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Viswanathan SR, Ha G, Hoff AM, Wala JA, Carrot-Zhang J, Whelan CW, Haradhvala NJ, Freeman SS, Reed SC, Rhoades J, Polak P, Cipicchio M, Wankowicz SA, Wong A, Kamath T, Zhang Z, Gydush GJ, Rotem D PCF/SU2C International Prostate Cancer Dream Team. Love JC, Getz G, Gabriel S, Zhang CZ, Dehm SM, Nelson PS, Van Allen EM, Choudhury AD, Adalsteinsson VA, Beroukhim R, Taplin ME, Meyerson M. Structural alterations driving castration-resistant prostate cancer revealed by linked-read genome sequencing. Cell. 2018;174:433–447. e19. doi: 10.1016/j.cell.2018.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Koga Y, Song H, Chalmers ZR, Newberg J, Kim E, Carrot-Zhang J, Piou D, Polak P, Abdulkadir SA, Ziv E, Meyerson M, Frampton GM, Campbell JD, Huang FW. Genomic profiling of prostate cancers from men with African and European ancestry. Clin Cancer Res. 2020;26:4651–4660. doi: 10.1158/1078-0432.CCR-19-4112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wu YM, Cieślik M, Lonigro RJ. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell. 2018;173:1770–1782. e14. doi: 10.1016/j.cell.2018.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Antonarakis ES, Isaacsson Velho P, Fu W. CDK12-altered prostate cancer: clinical features and therapeutic outcomes to standard systemic therapies, poly (ADPribose) polymerase inhibitors, and PD-1 inhibitors. JCO Precis Oncol. 2020;4:370–381. doi: 10.1200/PO.19.00399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gongora ABL, Marshall CH, Velho PI, Lopes CDH, Marin JF, Camargo AA, Bastos DA, Antonarakis ES. Extreme responses to a combination of DNA-damaging therapy and immunotherapy in CDK12-Altered metastatic castration-resistant prostate cancer: a potential therapeutic vulnerability. Clin Genitourin Cancer. 2022;20:183–188. doi: 10.1016/j.clgc.2021.11.015. [DOI] [PubMed] [Google Scholar]
  • 70.Markowski MC, Shenderov E, Eisenberger MA, Kachhap S, Pardoll DM, Denmeade SR, Antonarakis ES. Extreme responses to immune checkpoint blockade following bipolar androgen therapy and enzalutamide in patients with metastatic castration resistant prostate cancer. Prostate. 2020;80:407–411. doi: 10.1002/pros.23955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rescigno P, Gurel B, Pereira R, Crespo M, Rekowski J, Rediti M, Barrero M, Mateo J, Bianchini D, Messina C, Fenor de la Maza MD, Chandran K, Carmichael J, Guo C, Paschalis A, Sharp A, Seed G, Figueiredo I, Lambros M, Miranda S, Ferreira A, Bertan C, Riisnaes R, Porta N, Yuan W, Carreira S, de Bono JS. Characterizing CDK12-mutated prostate cancers. Clin Cancer Res. 2021;27:566–574. doi: 10.1158/1078-0432.CCR-20-2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Elliott A, Zhang J, Zhang Q, Swensen J, Martin D, Xiu J, Geynisman DM, Vaena D, Herzog TJ, Holloway RW, El-Deiry WS, Spetzler D, Heath E, Stafford P, Korn WM. Predicted immunogenicity of CDK12 Biallelic loss-of-function tumors Varies across cancer types. J Mol Diagn. 2021;23:1761–1773. doi: 10.1016/j.jmoldx.2021.08.010. [DOI] [PubMed] [Google Scholar]
  • 73.Calagua C, Ficial M, Jansen CS, Hirz T, Del Balzo L, Wilkinson S, Lake R, Ku AT, Voznesensky O, Sykes DB, Saylor PJ, Ye H, Signoretti S, Kissick H, Sowalsky AG, Balk SP, Einstein DJ. A subset of localized prostate cancer displays an immunogenic phenotype associated with losses of key tumor suppressor genes. Clin Cancer Res. 2021;27:4836–4847. doi: 10.1158/1078-0432.CCR-21-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, Sharpe AH, Freeman GJ, Germain RN, Nakaya HI, Xue H, Ahmed R. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537:417–21. doi: 10.1038/nature19330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Siddiqui I, Schaeuble K, Chennupati V, Marraco SAF, Calderon-Copete S, Ferreira DP, Carmona SJ, Scarpellino L, Gfeller D, Pradervand S, Luther SA, Speiser DE, Held W. Intratumoral Tcf1+ PD-1+ CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity. 2019;50:195–211. doi: 10.1016/j.immuni.2018.12.021. [DOI] [PubMed] [Google Scholar]
  • 76.Liu D, Augello MA, Grbesa I, Prandi D, Liu Y, Shoag JE, Karnes RJ, Trock BJ, Klein EA, Den RB, Demichelis F, Davicioni E, Sbonder A, Barbieri CE. Tumor subtype defines distinctpathways of molecular and clinical progression in primary prostatecancer. J Clin Invest. 2021;131:10. doi: 10.1172/JCI147878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Boysen G, Rodrigues DN, Rescigno P, Seed G, Dolling D, Riisnaes R, Crespo M, Zafeiriou Z, Sumanasuriya S, Bianchini D, Hunt J, Moloney D, Perez-Lopez R, Tunariu N, Miranda S, Figueiredo I, Ferreira A, Christova R, Gil V, Aziz S, Bertan C, de Oliveira FM, Atkin M, Clarke M, Goodall J, Sharp A, MacDonald T, Rubin MA, Yuan W, Barbieri CE, Carreira S, Mateo J, de Bono J. SPOP-Mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin Cancer Res. 2018;24:5585–5593. doi: 10.1158/1078-0432.CCR-18-0937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhao D, Cai L, Lu X, Liang X, Li J, Chen P, Ittman M, Shang X, Jiang S, Li H, Meng C, Flores I, Song JH, Horner JW, Lan Z, Wu C, Li J, Chang Q, Chen K, Wang G, Deng P, Spring DJ, Wang YA, DePinho RA. Chromatin regulator CHD1 remodels the immunosuppressive tumor microenvironment in PTEN-deficient prostate cancer. Cancer Discov. 2020;10:1374–87. doi: 10.1158/2159-8290.CD-19-1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, Tan Y, Ci Y, Wu F, Dai X, Guo J, Huang Y, Fan C, Ren S, Sun Y, Freeman GJ, Sicinski P, Wei W. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature. 2018;553:91–95. doi: 10.1038/nature25015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, Mahammedi H, Patnaik A, Subudhi SK, Ciprotti M, Simsek B, Saci A, Hu Y, Han GC, Fizazi K. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell. 2020;38:489–499. e3. doi: 10.1016/j.ccell.2020.08.007. [DOI] [PubMed] [Google Scholar]
  • 81.Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, Kaley TJ, Kendall SM, Motzer RJ, Hakimi AA, Voss MH, Russo P, Rosenberg J, Iyer G, Bochner BH, Bajorin DF, Al-Ahmadie HA, Chaft JE, Rudin CM, Riely GJ, Baxi S, Ho AL, Wong RJ, Pfister DG, Wolchok JD, Barker CA, Gutin PH, Brennan CW, Tabar V, Mellinghoff IK, DeAngelis LM, Ariyan CE, Lee N, Tap WD, Gounder MM, D’Angelo SP, Saltz L, Stadler ZK, Scher HI, Baselga J, Razavi P, Klebanoff CA, Yaeger R, Segal NH, Ku GY, DeMatteo RP, Ladanyi M, Rizvi NA, Berger MF, Riaz N, Solit DB, Chan TA, Morris LGT. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–206. doi: 10.1038/s41588-018-0312-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Tapia-Laliena MA, Korzeniewski N, Hohenfellner M, Duensing S. High-risk prostate cancer: a disease of genomic instability. Urol Oncol. 2014;32:1101–7. doi: 10.1016/j.urolonc.2014.02.005. [DOI] [PubMed] [Google Scholar]
  • 83.Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, Foye A, Kothari V, Perry MD, Bailey AM, Playdle D, Barnard TJ, Zhang L, Zhang J, Youngren JF, Cieslik MP, Parolia A, Beer TM, Thomas G, Chi KN, Gleave M, Lack NA, Zoubeidi A, Reiter RE, Rettig MB, Witte O, Ryan CJ, Fong L, Kim W, Friedlander T, Chou J, Li H, Das R, Li H, Moussavi-Baygi R, Goodarzi H, Gilbert LA, Lara PN Jr, Evans CP, Goldstein TC, Stuart JM, Tomlins SA, Spratt DE, Cheetham RK, Cheng DT, Farh K, Gehring JS, Hakenberg J, Liao A, Febbo PG, Shon J, Sickler B, Batzoglou S, Knudsen KE, He HH, Huang J, Wyatt AW, Dehm SM, Ashworth A, Chinnaiyan AM, Maher CA, Small EJ, Feng FY. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell. 2018;174:758–769. e9. doi: 10.1016/j.cell.2018.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, Antipin Y, Mitsiades N, Landers T, Dolgalev I, Major JE, Wilson M, Socci ND, Lash AE, Heguy A, Eastham JA, Scher HI, Reuter VE, Scardino PT, Sander C, Sawyers CL, Gerald W. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. doi: 10.1016/j.ccr.2010.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, Montgomery B, Taplin ME, Pritchard CC, Attard G, Beltran H, Abida W, Bradley RK, Vinson J, Cao X, Vats P, Kunju LP, Hussain M, Feng FY, Tomlins SA, Cooney KA, Smith DC, Brennan C, Siddiqui J, Mehra R, Chen Y, Rathkopf DE, Morris MJ, Solomon SB, Durack JC, Reuter VE, Gopalan A, Gao J, Loda M, Lis RT, Bowden M, Balk SP, Gaviola G, Sougnez C, Gupta M, Yu EY, Mostaghel EA, Cheng HH, Mulcahy H, True LD, Plymate SR, Dvinge H, Ferraldeschi R, Flohr P, Miranda S, Zafeiriou Z, Tunariu N, Mateo J, Perez-Lopez R, Demichelis F, Robinson BD, Schiffman M, Nanus DM, Tagawa ST, Sigaras A, Eng KW, Elemento O, Sboner A, Heath EI, Scher HI, Pienta KJ, Kantoff P, de Bono JS, Rubin MA, Nelson PS, Garraway LA, Sawyers CL, Chinnaiyan AM. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–1228. doi: 10.1016/j.cell.2015.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, Asangani IA, Ateeq B, Chun SY, Siddiqui J, Sam L, Anstett M, Mehra R, Prensner JR, Palanisamy N, Ryslik GA, Vandin F, Raphael BJ, Kunju LP, Rhodes DR, Pienta KJ, Chinnaiyan AM, Tomlins SA. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487:239–43. doi: 10.1038/nature11125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kumar A, White TA, MacKenzie AP, Clegg N, Lee C, Dumpit RF, Coleman I, Ng SB, Salipante SJ, Rieder MJ, Nickerson DA, Corey E, Lange PH, Morrissey C, Vessella RL, Nelson PS, Shendure J. Exome sequencing identifies a spectrum of mutation frequencies in advanced and lethal prostate cancers. Proc Natl Acad Sci U S A. 2011;108:17087–92. doi: 10.1073/pnas.1108745108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Castle JC, Uduman M, Pabla S, Stein RB, Buell JS. Mutation-derived neoantigens for cancer immunotherapy. Front Immunol. 2019;10:18. doi: 10.3389/fimmu.2019.01856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wang TY, Wang L, Alam SK, Hoeppner LH, Yang R. ScanNeo: identifying indel-derived neoantigens using RNA-Seq data. Bioinformatics. 2019;35:4159–4161. doi: 10.1093/bioinformatics/btz193. [DOI] [PubMed] [Google Scholar]
  • 90.Martin AM, Nirschl TR, Nirschl CJ, Francica BJ, Kochel CM, van Bokhoven A, Meeker AK, Lucia MS, Anders RA, DeMarzo AM, Drake CG. Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis. 2015;18:325–32. doi: 10.1038/pcan.2015.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wu Z, Chen H, Luo W, Zhang H, Li G, Zeng F, Deng F. The landscape of immune cells infiltrating in prostate cancer. Front Oncol. 2020;10:517637. doi: 10.3389/fonc.2020.517637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Andersen LB, Nørgaard M, Rasmussen M, Fredsøe J, Borre M, Ulhøi BP, Sørensen KD. Immune cell analyses of the tumor microenvironment in prostate cancer highlight infiltrating regulatory T cells and macrophages as adverse prognostic factors. J Pathol. 2021;255:155–165. doi: 10.1002/path.5757. [DOI] [PubMed] [Google Scholar]
  • 93.Rui X, Shao S, Wang L, Leng J. Identification of recurrence marker associated with immune infiltration in prostate cancer with radical resection and build prognostic nomogram. BMC Cancer. 2019;19:1179. doi: 10.1186/s12885-019-6391-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, Ellis IO, Green AR. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J. Clin. Oncol. 2011;29:1949–1955. doi: 10.1200/JCO.2010.30.5037. [DOI] [PubMed] [Google Scholar]
  • 95.Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K. Intraepithelial CD8 + tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 97.Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, West AN, Carmona M, Kivork C, Seja E, Cherry G, Gutierrez AJ, Grogan TR, Mateus C, Tomasic G, Glaspy JA, Emerson RO, Robins H, Pierce RH, Elashoff DA, Robert C, Ribas A. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, Kohrt HE, Horn L, Lawrence DP, Rost S, Leabman M, Xiao Y, Mokatrin A, Koeppen H, Hegde PS, Mellman I, Chen DS, Hodi FS. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vitkin N, Nersesian S, Siemens DR, Koti M. The tumor immune contexture of prostate cancer. Front Immunol. 2019;10:603. doi: 10.3389/fimmu.2019.00603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zhao SG, Lehrer J, Chang SL, Das R, Erho N, Liu Y, Sjöström M, Den RB, Freedland SJ, Klein EA, Karnes RJ, Schaeffer EM, Xu M, Speers C, Nguyen PL, Ross AE, Chan JM, Cooperberg MR, Caroll PR, Davicioni E, Fong L, Spratt DE, Feng FY. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J Nat Cancer Inst. 2019;111:301–310. doi: 10.1093/jnci/djy141. [DOI] [PubMed] [Google Scholar]
  • 101.Ness N, Andersen S, Valkov A, Nordby Y, Donnem T, Al-Saad S, Busund LT, Bremnes RM, Richardsen E. Infiltration of CD8þ lymphocytes is an independent prognostic factor of biochemical failure-free survival in prostate cancer. Prostate. 2014;74:1452–1461. doi: 10.1002/pros.22862. [DOI] [PubMed] [Google Scholar]
  • 102.Yang Y, Attwood K, Bshara W, Mohler JL, Guru K, Xu B, Kalinski P, Chatta G. High intratumoral CD8(+) T-cell infiltration is associated with improved survival in prostate cancer patients undergoing radical prostatectomy. Prostate. 2021;81:20–28. doi: 10.1002/pros.24068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Vicier C, Ravi P, Kwak L, Werner L, Huang Y, Evan C, Loda M, Hamid AA, Sweeney CJ. Association between CD8 and PD-L1 expression and outcomes after radical prostatectomy for localized prostate cancer. Prostate. 2021;81:50–57. doi: 10.1002/pros.24079. [DOI] [PubMed] [Google Scholar]
  • 104.Zarif JC, Baena-Del Valle JA, Hicks JL, Heaphy CM, Vidal I, Luo J, Lotan TL, Hooper JE, Isaacs WB, Pienta KJ, De Marzo AM. Mannose receptor-positive macrophage infiltration correlates with prostate cancer onset and metastatic castration-resistant disease. Eur Urol Oncol. 2019;2:429–436. doi: 10.1016/j.euo.2018.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Comito G, Giannoni E, Segura CP, Barcellos-de-Souza P, Raspollini MR, Baroni G, Lanciotti M, Serni S, Chiarugi P. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2014;33:2423–2431. doi: 10.1038/onc.2013.191. [DOI] [PubMed] [Google Scholar]
  • 106.Erlandsson A, Carlsson J, Lundholm M, Fält A, Andersson SO, Andrén O, Davidsson S. M2 macrophages and regulatory T cells in lethal prostate cancer. Prostate. 2019;79:363–369. doi: 10.1002/pros.23742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.JiaWei Z, ChunXia D, CunDong L, Yang L, JianKun Y, HaiFeng D, Cheng Y, ZhiPeng H, HongYi W, DeYing L, ZhiJian L, Xiao X, QiZhao Z, KangYi X, WenBing G, Ming X, JunHao Z, JiMing B, ShanChao Z, MingKun C. M2 subtype tumor associated macrophages (M2-TAMs) infiltration predicts poor response rate of immune checkpoint inhibitors treatment for prostate cancer. Ann Med. 2021;53:730–740. doi: 10.1080/07853890.2021.1924396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Martori C, Sanchez-Moral L, Paul T, Pardo JC, Font A, Ruiz de Porras V, Sarrias MR. Macrophages as a therapeutic target in metastatic prostate cancer: a way to overcome immunotherapy resistance? Cancers (Basel) 2022;14:440. doi: 10.3390/cancers14020440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Duan Z, Luo Y. Targeting macrophages in cancer immunotherapy. Signal Transduct Target Ther. 2021;6:127. doi: 10.1038/s41392-021-00506-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell. 2015;27:462–472. doi: 10.1016/j.ccell.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nonomura N, Takayama H, Nakayama M, Nakai Y, Kawashima A, Mukai M, Nagahara A, Aozasa K, Tsujimura A. Infiltration of tumour-associated macrophages in prostate biopsy specimens is predictive of disease progression after hormonal therapy for prostate cancer. BJU Int. 2011;107:1918–1922. doi: 10.1111/j.1464-410X.2010.09804.x. [DOI] [PubMed] [Google Scholar]
  • 112.Cioni B, Zaalberg A, Van Beijnum JR, Melis MHM, Van Burgsteden J, Muraro MJ, Hooijberg E, Peters D, Hofland I, Lubeck Y, De Jong J, Sanders J, Vivié J, Van der Poel HG, De Boer JP, Griffioen AW, Zwart W, Bergman AM. Androgen receptor signalling in macrophages promotes TREM-1-mediated prostate cancer cell line migration and invasion. Nat Commun. 2020;11:4498. doi: 10.1038/s41467-020-18313-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Erlandsson A, Carlsson J, Lundholm M, Falt A, Andersson SO, Andren O, Davidsson S. M2 macrophages and regulatory T cells in lethal prostate cancer. Prostate. 2019;79:363–369. doi: 10.1002/pros.23742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hu W, Qian Y, Yu F, Liu W, Wu Y, Fang X, Hao W. Alternatively activated macrophages are associated with metastasis and poor prognosis in prostate adenocarcinoma. Oncol Lett. 2015;10:1390–1396. doi: 10.3892/ol.2015.3400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Izumi K, Fang LY, Mizokami A, Namiki M, Li L, Lin WJ, Chang C. Targeting the androgen receptor with siRNA promotes prostate cancer metastasis through enhanced macrophage recruitment via CCL2/CCR2-induced STAT3 activation. EMBO Mol Med. 2013;5:1383–1401. doi: 10.1002/emmm.201202367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Maolake A, Izumi K, Shigehara K, Natsagdorj A, Iwamoto H, Kadomoto S, Takezawa Y, Machioka K, Narimoto K, Namiki M, Lin WJ, Wufuer G, Mizokami A. Tumor-associated macrophages promote prostate cancer migration through activation of the CCL22-CCR4 axis. Oncotarget. 2017;8:9739–9751. doi: 10.18632/oncotarget.14185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Huang R, Wang S, Wang N, Zheng Y, Zhou J, Yang B, Wang X, Zhang J, Guo L, Wang S, Chen Z, Wang Z, Xiang S. CCL5 derived from tumor-associated macrophages promotes prostate cancer stem cells and metastasis via activating beta-catenin/STAT3 signaling. Cell Death Dis. 2020;11:234. doi: 10.1038/s41419-020-2435-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, Ring AM, Connolly AJ, Weissman IL. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:495–499. doi: 10.1038/nature22396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Prima V, Kaliberova LN, Kaliberov S, Curiel DT, Kusmartsev S. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc Natl Acad Sci U S A. 2017;114:1117–1122. doi: 10.1073/pnas.1612920114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.McCord R, Bolen CR, Koeppen H, Kadel EE, Oestergaard MZ, Nielsen T, Sehn LH, Venstrom JM. PD-L1 and tumor-associated macrophages in de novo DLBCL. Blood Adv. 2019;3:531–540. doi: 10.1182/bloodadvances.2018020602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sun NY, Chen YL, Wu WY, Lin HW, Chiang YC, Chang CF, Tai YJ, Hsu HC, Chen CA, Sun WZ, Cheng WF. Blockade of PD-L1 Enhances cancer immunotherapy by regulating dendritic cell maturation and macrophage polarization. Cancers. 2019;11:1400. doi: 10.3390/cancers11091400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gubin MM, Esaulova E, Ward JP, Malkova ON, Runci D, Wong P, Noguchi T, Arthur CD, Meng W, Alspach E, Medrano RFV, Fronick C, Fehlings M, Newell EW, Fulton RS, Sheehan KCF, Oh ST, Schreiber RD, Artyomov MN. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell. 2018;175:1014–1030. e19. doi: 10.1016/j.cell.2018.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Guedes LB, Antonarakis ES, Schweizer MT, Mirkheshti N, Almutairi F, Park JC, Glavaris S, Hicks J, Eisenberger MA, De Marzo AM, Epstein JI, Isaacs WB, Eshleman JR, Pritchard CC, Lotan TL. MSH2 Loss in primary prostate cancer. Clin Cancer Res. 2017;23:6863–6874. doi: 10.1158/1078-0432.CCR-17-0955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sena LA, Salles DC, Engle EL, Zhu Q, Tukachinsky H, Lotan TL, Antonarakis ES. Mismatch repair-deficient prostate cancer with parenchymal brain metastases treated with immune checkpoint blockade. Cold Spring Harb Mol Case Study. 2021;7:a006094. doi: 10.1101/mcs.a006094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Investig. 2006;116:1935–1945. doi: 10.1172/JCI27745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Chen H, Liakou CI, Kamat A, Pettaway C, Ward JF, Tang DN, Sun J, Jungbluth AA, Troncoso P, Logothetis C, Sharma P. Anti-CTLA-4 therapy results in higher CD4+ICOShi T cell frequency and IFN-gamma levels in both nonmalignant and malignant prostate tissues. Proc Natl Acad Sci U S A. 2009;106:2729–2734. doi: 10.1073/pnas.0813175106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Waitz R, Solomon SB, Petre EN, Trumble AE, Fasso M, Norton L, Allison JP. Potent induction of tumor immunity by combining tumor cryoablation with anti-CTLA-4 therapy. Cancer Res. 2012;72:430–439. doi: 10.1158/0008-5472.CAN-11-1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, Peggs KS, Ravethc JV, Allison JP, Quezada SA. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–1710. doi: 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Muthuswamy R, Corman JM, Dahl K, Chatta GS, Kalinski P. Functional reprogramming of human prostate cancer to promote local attraction of effector CD8(+) T cells. Prostate. 2016;76:1095–105. doi: 10.1002/pros.23194. [DOI] [PubMed] [Google Scholar]
  • 130.Taube JM. Unleashing the immune system: PD-1 and PD-Ls in the pre-treatment tumor microenvironment and correlation with response to PD-1/PD-L1 blockade. Oncoimmunology. 2014;3:e963413. doi: 10.4161/21624011.2014.963413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Martin AM, Nirschl TR, Nirschl CJ, Francica BJ, Kochel CM, Van Bokhoven A, Meeker AK, Lucia MS, Anders RA, DeMarzo AM, Drake CG. Paucity of PD-L1 expression in prostate cancer: Innate and adaptive immune resistance. Prostate Cancer Prostatic Dis. 2015;18:325–332. doi: 10.1038/pcan.2015.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Palicelli A, Bonacini M, Croci S, Magi-Galluzzi C, Cañete-Portillo S, Chaux A, Bisagni A, Zanetti E, De Biase D, Melli B, Sanguedolce F, Ragazzi M, Bonasoni MP, Soriano A, Ascani S, Zizzo M, Castro Ruiz C, De Leo A, Giordano G, Landriscina M, Carrieri G, Cormio L, Berney DM, Athanazio D, Gandhi J, Cavazza A, Santandrea G, Tafuni A, Zanelli M. What do we have to know about PD-L1 expression in prostate cancer? A systematic literature review. Part 1: focus on immunohistochemical results with discussion of pre-analytical and interpretation variables. Cells. 2021;10:3166. doi: 10.3390/cells10113166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Hahn E, Liu SK, Vesprini D, Xu B, Downes MR. Immune infiltrates and PD-L1 expression in treatment-naïve acinar prostatic adenocarcinoma: an exploratory analysis. J Clin Pathol. 2018;71:1023–1027. doi: 10.1136/jclinpath-2018-205404. [DOI] [PubMed] [Google Scholar]
  • 134.Wang C, Hahn E, Slodkowska E, Eskander A, Enepekides D, Higgins K, Vesprini D, Liu SK, Downes MR, Xu B. Reproducibility of PD-L1 immunohistochemistry interpretation across various types of genitourinary and head/neck carcinomas, antibody clones, and tissue types. Hum Pathol. 2018;82:131–139. doi: 10.1016/j.humpath.2018.07.024. [DOI] [PubMed] [Google Scholar]
  • 135.Fankhauser CD, Schüffler PJ, Gillessen S, Omlin A, Rupp NJ, Rueschoff JH, Hermanns T, Poyet C, Sulser T, Moch H, Wild PJ. Comprehensive immunohistochemical analysis of PD-L1 shows scarce expression in castration-resistant prostate cancer. Oncotarget. 2018;9:10284–10293. doi: 10.18632/oncotarget.22888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Matveev V, Kirichek A, Safronova V, Kokosadze N, Khalmurzaev O, Kamolov B, Liubchenko L. The prognostic value of tumor PD-L1 status in patients with metastatic prostate cancer. Cancer Urol. 2019;15:57–65. [Google Scholar]
  • 137.Iacovelli R, Ciccarese C, Brunelli M, Bogina G, Munari E, Bimbatti D, Mosillo C, Fantinel E, Bria E, Martignoni G, Tortora G. PD-L1 expression in de novo metastatic castration-sensitive prostate cancer. J Immunother. 2019;42:269–273. doi: 10.1097/CJI.0000000000000287. [DOI] [PubMed] [Google Scholar]
  • 138.Xian P, Ge D, Wu VJ, Patel A, Tang WW, Wu X, Zhang K, Li L, You Z. PD-L1 instead of PD-1 status is associated with the clinical features in human primary prostate tumors. Am J Clin Exp Urol. 2019;7:159–169. [PMC free article] [PubMed] [Google Scholar]
  • 139.Li H, Wang Z, Zhang Y, Sun G, Ding B, Yan L, Liu H, Guan W, Hu Z, Wang S, Cheng F, Xu H, Zhang X, Ye Z. The immune checkpoint regulator PDL1 is an independent prognostic biomarker for biochemical recurrence in prostate cancer patients following adjuvant hormonal therapy. J Cancer. 2019;10:3102–3111. doi: 10.7150/jca.30384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Lindh C, Kis L, Delahunt B, Samaratunga H, Yaxley J, Wiklund NP, Clements M, Egevad L. PD-L1 expression and deficient mismatch repair in ductal adenocarcinoma of the prostate. APMIS Acta Pathol Microbiol Immunol Scand. 2019;127:554–560. doi: 10.1111/apm.12970. [DOI] [PubMed] [Google Scholar]
  • 141.Von Hardenberg J, Hartmann S, Nitschke K, Worst TS, Ting S, Reis H, Nuhn P, Weis CA, Erben P. Programmed Death Ligand 1 (PD-L1) status and tumor-infiltrating lymphocytes in hot spots of primary and liver metastases in prostate cancer with neuroendocrine differentiation. Clin Genitourin Cancer. 2019;17:145–153. doi: 10.1016/j.clgc.2018.12.007. [DOI] [PubMed] [Google Scholar]
  • 142.Richter I, Jirasek T, Havlickova I, Curcikova R, Samal V, Dvorak J, Bartos J. The expression of PD-L1 in patients with castrate prostate cancer treated with enzalutamide. J BUON. 2018;23:1796–1802. [PubMed] [Google Scholar]
  • 143.Haffner MC, Guner G, Taheri D, Netto GJ, Palsgrove DN, Zheng Q, Guedes LB, Kim K, Tsai H, Esopi DM, Lotan TL, Sharma S, Meeker AK, Chinnaiyan AM, Neison WG, Yegnasubramanian S, Luo J, Mehra R, Atonarakis ES, Drake CG, DeMarzo AM. Comprehensive evaluation of programmed death-ligand 1 expression in primary and metastatic prostate cancer. Am J Pathol. 2018;188:1478–1485. doi: 10.1016/j.ajpath.2018.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Ness N, Andersen S, Khanehkenari MR, Nordbakken CV, Valkov A, Paulsen EE, Nordby Y, Bremnes RM, Donnem T, Busund LT, Richardsen E. The prognostic role of immune checkpoint markers programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) in a large, multicenter prostate cancer cohort. Oncotarget. 2017;8:26789–26801. doi: 10.18632/oncotarget.15817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Baas W, Gershburg S, Dynda D, Delfino K, Robinson K, Nie D, Yearley JH, Alanee S. Immune characterization of the programmed death receptor pathway in high risk prostate cancer. Clin Genitourin Cancer. 2017;15:577–581. doi: 10.1016/j.clgc.2017.04.002. [DOI] [PubMed] [Google Scholar]
  • 146.Lin H, Liu Q, Zeng X, Yu W, Xu G. Pembrolizumab with or without enzalutamide in selected populations of men with previously untreated metastatic castration-resistant prostate cancer harbouring programmed cell death ligand-1 staining: a retrospective study. BMC Cancer. 2021;21:399. doi: 10.1186/s12885-021-08156-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Haffner MC, Guner G, Taheri D, Netto GJ, Palsgrove DN, Zheng Q, Guedes LB, Kim K, Tsai H, Esopi DM, Lotan TL, Sharma R, Meeker Ak, Chinnaiyan AM, Nelson WG, Yegnasubramanian S, Luo J, Mehra R, Atonarakis ES, Drake CG, DeMarzo AM. Comprehensive evaluation of programmed death-ligand 1 expression in primary and metastatic prostate cancer. Am J Pathol. 2018;188:1478–85. doi: 10.1016/j.ajpath.2018.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bishop JL, Sio A, Angeles A, Roberts ME, Azad AA, Chi KN, Zoubeidi A. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget. 2015;6:234–42. doi: 10.18632/oncotarget.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Zhao SG, Lehrer J, Chang SL, Das R, Erho N, Liu Y, Sjöström M, Den RB, Freedland SJ, Klein EA, Karnes RJ, Schaeffer EM, Xu M, Speers C, Nguyen PL, Ross AE, Chan JM, Cooperberg MR, Carrol PR, Davicioni E, Fong L, Spratt DE, Feng FY. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J Natl Cancer Inst. 2019;111:301–10. doi: 10.1093/jnci/djy141. [DOI] [PubMed] [Google Scholar]
  • 150.Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, Zhao H, Chen J, Chen H, Efstathiou E, Troncoso P, Allison JP, Logothetis CJ, Wistuba II, Sepulveda MA, Sun J, Wargo J, Blando J, Sharma P. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23:551–555. doi: 10.1038/nm.4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, Rathkopf D, Morris MJ, Danila DC, Slovin SF, Carbone E, Barnett ES, Hullings M, Hechtman JF, Zehir A, Shia J, Jonsson P, Stadler ZK, Srinivasan P, Laudone VP, Reuter V, Wolchok JD, Socci ND, Taylor BS, Berger MF, Kantoff PW, Sawyers CL, Schultz N, Solit DB, Copalan A, Scher HI. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 2019;5:471–478. doi: 10.1001/jamaoncol.2018.5801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad N, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Croceniz TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Beyer KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler DW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. doi: 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Antonarakis ES, Shaukat F, Isaacsson Velho P, Kaur H, Shenderov E, Pardoll DM, Lotan TL. Clinical features and therapeutic outcomes in men with advanced prostate cancer and DNA mismatch repair gene mutations. Eur Urol. 2019;75:378–382. doi: 10.1016/j.eururo.2018.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Pritchard CC, Morrissey C, Kumar A, Zhang X, Smith C, Coleman I, Salipante SJ, Milbank J, Yu M, Grady WM, Tait JF, Corey E, Vessela RL, Walsh T, Shendure J, Nelson PS. Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat Commun. 2014;5:4988. doi: 10.1038/ncomms5988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kaur HB, Vidotto T, Mendes AA, Salles DC, Isaacs WB, Antonarakis ES, Lotan TL. Association between pathogenic germline mutations in BRCA2 and ATM and tumor-infiltrating lymphocytes in primary prostate cancer. Cancer Immunol Immunother. 2022;71:943–951. doi: 10.1007/s00262-021-03050-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pietro GD, Chornokur G, Kumar NB, Davis C, Park JY. Racial differences in the diagnosis and treatment of prostate cancer. Int Neurourol J. 2016;20(Suppl 2):S112–S119. doi: 10.5213/inj.1632722.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Tsodikov A, Gulati R, de Carvalho TM, Heijnsdijk EAM, Hunter-Merrill RA, Mariotto AB, de Koning HJ, Etzioni R. Is prostate cancer different in black men? Answers from 3 natural history models. Cancer. 2017;123:2312–2319. doi: 10.1002/cncr.30687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.McKay RR, Gold T, Zarif JC, Chowdhury-Paulino IM, Friedant A, Gerke T, Grant M, Hawthorne K, Heath E, Huang FW, Jackson MD, Mahal B, Ogbeide O, Paich K, Ragin C, Rencsok EM, Simmons S, Yates C, Vinson J, Kantoff PW, George DJ, Mucci LA. Tackling diversity in prostate cancer clinical trials: a report from the diversity working group of the IRONMAN registry. JCO Glob Oncol. 2021;7:495–505. doi: 10.1200/GO.20.00571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Sartor O, Armstrong AJ, Ahaghotu C, McLeod DG, Cooperberg MR, Penson DF, Kantoff PW, Vogelzang NJ, Hussain A, Pieczonka CM, Shore ND, Quinn DI, Small EJ, Heath EI, Tutrone RF, Schellhammer PF, Harmon M, Chang NN, Sheikh NA, Brown B, Freedland SJ, Higano CS. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis. 2020;23:517–526. doi: 10.1038/s41391-020-0213-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Higano CS, Armstrong AJ, Sartor AO, Vogelzang NJ, Kantoff PW, McLeod DG, Pieczonka CM, Penson DF, Shore ND, Vacirca J, Concepcion RS, Tutrone RF, Nordquist LT, Quinn DI, Kassabian V, Scholz MC, Harmon M, Tyler RC, Chang NN, Tang H, Cooperberg MR. Real-world outcomes of sipuleucel-T treatment in PROCEED, a prospective registry of men with metastatic castration-resistant prostate cancer. Cancer. 2019;125:4172–4180. doi: 10.1002/cncr.32445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Hawley JE, Pan S, Kandadi H, Chaimowitz MG, Sheikh N, Drake CG. Analysis of circulating immune biomarkers by race in men with metastatic castration-resistant prostate cancer treated with Sipuleucel-T. J Natl Cancer Inst. 2022;114:314–317. doi: 10.1093/jnci/djab145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Rayford W, Beksac AT, Alger J, Alshalalfa M, Ahmed M, Khan I, Falagario UG, Liu Y, Davicioni E, Spratt DE, Schaeffer EM, Feng FY, Mahal B, Nguyen PL, Den RB, Greenberger MD, Bradley R, Watson JM, Beamer M, Stamatakis L, Carmen DJ, Awasthi S, Hwang J, Weil R, Merisaari H, Mohamed N, Deane LA, Chakravarty D, Yadav KK, Yamoah K, Nair SS, Tewari AK. Comparative analysis of 1152 African-American and European-American men with prostate cancer identifies distinct genomic and immunological differences. Commun Biol. 2021;4:670. doi: 10.1038/s42003-021-02140-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Awasthi S, Berglund A, Abraham-Miranda J, Rounbehler RJ, Kensler K, Serna A, Vidal A, You S, Freeman MR, Davicioni E, Liu Y, Karnes RJ, Klein EA, Den RB, Trock BJ, Campbell JD, Einstein DJ, Gupta R, Balk S, Lal P, Park JY, Cleveland JL, Rebbeck TR, Freedland SJ, Yamoah K. Comparative genomics reveals distinct immune-oncologic pathways in African American Men with prostate cancer. Clin Cancer Res. 2021;27:320–329. doi: 10.1158/1078-0432.CCR-20-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Krieger KL, Gohlke JH, Lee KJ, Piyarathna DWB, Castro PD, Jones JA, Ittmann MM, Gassman NR, Sreekumar A. Repair-assisted damage detection reveals biological disparities in prostate cancer between African Americans and European Americans. Cancers (Basel) 2022;14:1012. doi: 10.3390/cancers14041012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Sartor O, Yang S, Ledet E, Moses M, Nicolosi P. Inherited DNA-repair gene mutations in African American men with prostate cancer. Oncotarget. 2020;11:440–442. doi: 10.18632/oncotarget.27456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Isaacsson Velho P, Qazi F, Hassan S, Carducci MA, Denmeade SR, Markowski MC, Thorek DL, DeWeese TL, Song DY, Tran PT, Eisenberger MA, Antonarakis ES. Efficacy of radium-223 in bone-metastatic Castration-resistant prostate cancer with and without homologous repair gene defects. Eur Urol. 2019;76:170–176. doi: 10.1016/j.eururo.2018.09.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Reams RR, Agrawal D, Davis MB, Yoder S, Odedina FT, Kumar N, Higginbotham JM, Akinremi T, Suther S, Soliman KF. Microarray comparison of prostate tumor gene expression in African-American and Caucasian American males: a pilot project study. Infect Agent Cancer. 2009;4(Suppl 1):S3. doi: 10.1186/1750-9378-4-S1-S3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Wallace TA, Prueitt RL, Yi M, Howe TM, Gillespie JW, Yfantis HG, Stephens RM, Caporaso NE, Loffredo CA, Ambs S. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res. 2008;68:927–936. doi: 10.1158/0008-5472.CAN-07-2608. [DOI] [PubMed] [Google Scholar]
  • 169.Kinseth MA, Jia Z, Rahmatpanah F, Sawyers A, Sutton M, Wang-Rodriguez J, Mercola D, McGuire KL. Expression differences between African American and Caucasian prostate cancer tissue reveals that stroma is the site of aggressive changes. Int J Cancer. 2014;134:81–91. doi: 10.1002/ijc.28326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Weiner AB, Vidotto T, Liu Y, Mendes AA, Salles DC, Faisal FA, Murali S, McFarlane M, Imada EL, Zhao X, Li Z, Davicioni E, Marchionni L, Chinnaiyan AM, Freedland SJ, Spratt DE, Wu JD, Lotan TL, Schaeffer EM. Plasma cells are enriched in localized prostate cancer in Black men and are associated with improved outcomes. Nat Commun. 2021;12:935. doi: 10.1038/s41467-021-21245-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Tang W, Wallace TA, Yi M, Magi-Galluzzi C, Dorsey TH, Onabajo OO, Obajemu A, Jordan SV, Loffredo CA, Stephens RM, Silverman RH, Stark GR, Klein EA, Prokunina-Olsson L, Ambs S. IFNL4-ΔG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with prostate cancer. Clin Cancer Res. 2018;24:5471–5481. doi: 10.1158/1078-0432.CCR-18-1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ahaghotu C, Tyler R, Sartor O. African American participation in oncology clinical trials--focus on prostate cancer: implications, barriers, and potential solutions. Clin Genitourin Cancer. 2016;14:105–16. doi: 10.1016/j.clgc.2015.12.003. [DOI] [PubMed] [Google Scholar]
  • 173.Ajewole VB, Akindele O, Abajue U, Ndulue O, Marshall JJ, Mossi YT. Cancer disparities and black American Representation in clinical trials leading to the approval of oral chemotherapy drugs in the United States between 2009 and 2019. JCO Oncol Pract. 2021;17:e623–e628. doi: 10.1200/OP.20.01108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Cheever MA, Higano CS. PROVENGE (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin Cancer Res. 2011;17:3520–6. doi: 10.1158/1078-0432.CCR-10-3126. [DOI] [PubMed] [Google Scholar]
  • 175.Kantoff PW, Higano CS, Shore ND, Berger R, Smal EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 176.Kobiyama K, Jounai N, Aoshi T, Tozuka M, Takeshita F, Coban C, Ishiii KJ. Innate immune signaling by, and genetic adjuvants for DNA vaccination. Vaccines. 2013;1:278–292. doi: 10.3390/vaccines1030278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kyriakopoulos CE, Eickhoff JC, Ferrari AC, Schweizer MT, Wargowski E, Olson BM, McNeel DG. 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. Clin Cancer Res. 2020;26:5162–5171. doi: 10.1158/1078-0432.CCR-20-0945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Shore ND, Morrow MP, McMullan T, Kraynyak KA, Sylvester A, Bhatt K, Cheung J, Boyer JD, Liu L, Sacchetta B, Rosencranz S, Heath EI, Nordquist L, Cheng HH, Tagawa ST, Appleman LJ, Tutrone R, Garcia JA, Whang YE, Kelly WK, Weiner DB, Bagarazzi ML, Skolnik JM. CD8+ T cells impact rising PSA in biochemically relapsed cancer patients using immunotherapy targeting tumor-associated antigens. Mol Ther. 2020;28:1238–1250. doi: 10.1016/j.ymthe.2020.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gulley JL, Madan RA, Tsang KY, Jochems C, Marté JL, Farsaci B, Tucker JA, Hodge JW, Liewehr DJ, Steinberg SM, Heery CR, Schlom J. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2:133–141. doi: 10.1158/2326-6066.CIR-13-0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Kantoff PW, Gulley JL, Pico-Navarro C. Revised overall survival analysis of a phase II, randomized, double-blind, controlled study of PROSTVAC in men with metastatic castration-resistant prostate cancer. Physiol Behav. 2016;176:100–106. doi: 10.1200/JCO.2016.69.7748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gulley JL. Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. Am Soc Clin Oncol. 2019;37:1051–1061. doi: 10.1200/JCO.18.02031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Stenzl A. Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. Eur Urol. 2020;77:131–132. doi: 10.1016/j.eururo.2019.07.025. [DOI] [PubMed] [Google Scholar]
  • 183.Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: The GVAX® vaccine for prostate cancer. Urol Oncol Semin Orig Investig. 2006;24:419–424. doi: 10.1016/j.urolonc.2005.08.021. [DOI] [PubMed] [Google Scholar]
  • 184.Gamat-Huber M, Jeon D, Johnson LE, Moseman JE, Muralidhar A, Potluri HK, Rastogi I, Wargowski E, Zahm CD, McNeel DG. Treatment combinations with DNA vaccines for the treatment of metastatic castration-resistant prostate cancer (mCRPC) Cancers (Basel) 2020;12:1–23. doi: 10.3390/cancers12102831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Obradovic AZ, Dallos MC, Zahurak ML, Partin AW, Schaeffer EM, Ross AE, Allaf ME, Nirschl TR, Liu D, Chapman CG, O’Neal T, Cao H, Durham JN, Guner G, Baena-Del Valle JA, Ertunc O, De Marzo AM, Antonarakis ES, Drake CG. T-Cell infiltration and adaptive treg resistance in response to androgen deprivation with or without vaccination in localized prostate cancer. Clin Cancer Res. 2020;26:3182–3192. doi: 10.1158/1078-0432.CCR-19-3372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Bone. 2008;23:1–7. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, Rogers LJ, Gracia GJ, jones SA, Mangiameli DP, Pelletier MM, Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati A, Rosenberg SA. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma mark. J. Clin. Oncol. 2005;23:2346–2357. doi: 10.1200/JCO.2005.00.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Fu J, Shang Y, Qian Z, Hou J, Yan F, Liu G, Dehua L, Tian X. Chimeric antigen receptor-t (Car-t) cells targeting epithelial cell adhesion molecule (epcam) can inhibit tumor growth in ovarian cancer mouse model. J Vet Med Sci. 2021;83:241–247. doi: 10.1292/jvms.20-0455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Bębnowska D, Grywalska E, Niedźwiedzka-Rystwej P, Sosnowska-Pasiarska B, Smok-Kalwat J, Pasiarski M, Góźdź S, Roliński J, Polkowski W. CAR-T cell therapy-an overview of targets in gastric cancer. J Clin Med. 2020;9:1894. doi: 10.3390/jcm9061894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.He C, Zhou Y, Li Z, Farooq MA, Ajmal I, Zhang H, Zhang L, Tao L, Yao J, Du B, Liu M, Jiang W. Co-expression of il-7 improves nkg2d-based car t cell therapy on prostate cancer by enhancing the expansion and inhibiting the apoptosis and exhaustion. Cancers (Basel) 2020;12:1–17. doi: 10.3390/cancers12071969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Narayan V, Barber-Rotenberg JS, Jung IY, Lacey SF, Rech AJ, Davis MM, Hwang WT, Lal P, Carpenter EL, Maude SL, Plesa G, Vapiwala N, Chew A, Moniak M, Sebro RA, Farwell MD, Marshall A, Gilmore J, Lledo L, Dengel K, Church SE, Hether TD, Xu J, Gohil M, Buckingham TH, Yee SS, Gonzalez VE, Kulikovskaya I, Chen F, Tian L, Tien K, Gladney W, Nobles CL, Raymond HE Prostate Cancer Cellular Therapy Program Investigators. Hexner EO, Siegel DL, Bushman FD, June CH, Fraietta JA, Haas NB. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat Med. 2022;28:724–734. doi: 10.1038/s41591-022-01726-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Rosa K. OncLive https://www.onclive.com/view/p-psma-101-elicits-encouraging-responses-in-metastatic-castrationresistant-prostate-cancer (May 2022)
  • 193.Krueger TE, Thorek DLJ, Meeker AK, Isaacs JT, Brennen WN. Tumor-infiltrating mesenchymal stem cells: Drivers of the immunosuppressive tumor microenvironment in prostate cancer? Prostate. 2019;79:320–330. doi: 10.1002/pros.23738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, Sboner A, Esgueva R, Pflueger D, Sougnez C, Onofrio R, Carter SL, Park K, Habegger L, Ambrogio L, Fennell T, Parkin M, Saksena G, Voet D, Ramos AH, Pugh TJ, Wilkinson J, Fisher S, Winckler W, Mahan S, Ardlie K, Baldwin J, Simons JW, Kitabayashi N, MacDonald TY, Kantoff PW, Chin L, Gabriel SB, Gerstein MB, Golub TR, Meyerson M, Tewari A, Lander ES, Getz G, Rubin MA, Garraway LA. The genomic complexity of primary human prostate cancer. Nature. 2011;470:214–220. doi: 10.1038/nature09744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Grasso CS, Wu Y, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, Asangani IA, Ateeq B, Chun SY, Siddiqui J, Sam L, Anstett M, Mehra R, Prensner JR, Palanisamy N, Ryslik GA, Vanadin F, Raphael BJ, Kunju LP, Rhodes DR, Pienta KJ, Chinnaiyan AM, Tomlins SA. The Mutational landscape of lethal castrate resistant prostate cancer. Nature. 2012;487:239–243. doi: 10.1038/nature11125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Yunger S, Bar El A, Zeltzer L, Fridman E, Raviv G, Laufer M, Schachter J, Markel G, Besser MJ. Tumor-infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy. Oncoimmunology. 2019;8:e1672494. doi: 10.1080/2162402X.2019.1672494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Lee HW, Nam KO, Park SJ, Kwon BS. 4-1BB enhances CD8+ T cell expansion by regulating cell cycle progression through changes in expression of cyclins D and E and cyclin-dependent kinase inhibitor p27kip1. Eur J Immunol. 2003;33:2133–2141. doi: 10.1002/eji.200323996. [DOI] [PubMed] [Google Scholar]
  • 198.Milone MC, Fish JD, Carpenito C, Carrol RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, Campana D, Riley JL, Grupp SA, Jun CH. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–1464. doi: 10.1038/mt.2009.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Hummel HD, Kufer P, Grüllich C, Seggewiss-Bernhardt R, Deschler-Baier B, Chatterjee M, Goebeler ME, Miller K, de Santis M, Loidl W, Dittrich C, Buck A, Lapa C, Thurner A, Wittemer-Rump S, Koca G, Boix O, Döcke WD, Finnern R, Kusi H, Ajavon-Hartmann A, Stienen S, Sayehli CM, Polat B, Bargou RC. Pasotuxizumab, a BiTE® immune therapy for castration-resistant prostate cancer: Phase I, dose-escalation study findings. Immunotherapy. 2021;13:125–141. doi: 10.2217/imt-2020-0256. [DOI] [PubMed] [Google Scholar]
  • 200.Deegen P, Thomas O, Nolan-Stevaux O, Li S, Wahl J, Bogner P, Aeffner F, Friedrich M, Liao MZ, Matthes K, Rau D, Rattel B, Raum T, Kufer P, Coxon A, Bailis JM. The PSMA-targeting half-life extended BiTE Therapy AMG 160 has potent antitumor activity in preclinical models of metastatic castration-resistant prostate cancer. Clin Cancer Res. 2021;27:2928–2937. doi: 10.1158/1078-0432.CCR-20-3725. [DOI] [PubMed] [Google Scholar]
  • 201.Lin TY, Park JA, Long A, Guo HF, Cheung NV. Novel potent anti-STEAP1 bispecific antibody to redirect T cells for cancer immunotherapy. J Immunother Cancer. 2021;9:e003114. doi: 10.1136/jitc-2021-003114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Dorff TB, Narayan V, Forman SJ, Zang PD, Fraietta JA, June CH, Haas NB, Priceman SJ. Novel redirected T-cell immunotherapies for advanced prostate cancer. Clin Cancer Res. 2022;28:576–584. doi: 10.1158/1078-0432.CCR-21-1483. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Clinical and Experimental Urology are provided here courtesy of e-Century Publishing Corporation

RESOURCES