Next Article in Journal
Single-Stage Trans-Vestibular and Foley’s-Assisted Epispadias Repair (STAFER) for Girls with Incontinent Epispadias: A Retrospective Study from a Tertiary-Care Center
Previous Article in Journal
Effectiveness of Ginseng, Rutin and Moringa for the Treatment of Erectile Dysfunction: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Prostate Cancer Immune Microenvironment, Biomarkers and Therapeutic Intervention

by
Yangyi Zhang
1,†,
Bethany K. Campbell
1,2,†,
Stanley S. Stylli
1,3,
Niall M. Corcoran
1,2,4,5,6 and
Christopher M. Hovens
1,2,4,6,*
1
Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia
2
University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, University of Melbourne, Parkville, VIC 3050, Australia
3
Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
4
Department of Urology, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
5
Department of Urology, Western Health, Footscray, VIC 3011, Australia
6
University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Uro 2022, 2(2), 74-92; https://doi.org/10.3390/uro2020010
Submission received: 2 March 2022 / Revised: 4 April 2022 / Accepted: 7 April 2022 / Published: 10 April 2022
(This article belongs to the Topic Prostate Cancer: Symptoms, Diagnosis & Treatment)

Abstract

:

Simple Summary

Prostate cancer is the most commonly occurring internal malignancy in men. Immunotherapies are emerging as important cancer therapies, having been successfully applied to a range of solid tumour types. However, due to the highly immunosuppressive tumour microenvironment, these successes have not been replicated in prostate cancer. To aid in the selection of patients who would be responsive to immunotherapy, efforts are underway to identify biomarkers which may be indicative of a positive therapeutic response. This review provides an overview of the prostate tumour microenvironment, summarises the immunotherapy approaches being explored for use in prostate cancer, and examines the use of biomarkers for therapy selection.

Abstract

Advanced prostate cancers have a poor survival rate and a lack of effective treatment options. In order to broaden the available treatments, immunotherapies have been investigated. These include cancer vaccines, immune checkpoint inhibitors, chimeric antigen receptor T cells and bispecific antibodies. In addition, combinations of different immunotherapies and with standard therapy have been explored. Despite the success of the Sipuleucel-T vaccine in the metastatic, castrate-resistant prostate cancer setting, other immunotherapies have not shown the same efficacy in this population at large. Some individual patients, however, have shown remarkable responsiveness to these therapies. Therefore, work is underway to identify which populations will respond positively to therapy via the identification of predictive biomarkers. These include biomarkers of the immunologically active tumour microenvironment and biomarkers indicative of high neoantigen expression in the tumour. This review examines the constitution of the prostate tumour immune microenvironment, explores the effectiveness of immunotherapies, and finally investigates how therapy selection can be optimised by the use of biomarkers.

1. Introduction

Prostate cancer is the second most frequently diagnosed cancer, and the fifth leading cause of cancer death among men in 2020 [1]. For instance, in 2018 in the U.S there were 3,245,430 men living with prostate cancer. In the U.S in 2021 there will be an estimated 248,530 new diagnoses of prostate cancer, and 34,130 deaths [2]. Due to this high prevalence, prostate cancer screening, consisting of prostate specific antigen (PSA) detection and digital rectal exam, generally commences in men in their 50s. Prostate cancer is a biologically heterogeneous disease that produces variable clinical outcomes. Low- and intermediate-risk localised prostate cancer is generally treated with curative attempts with ablative therapies such as surgery and radiotherapy. Of those patients treated for primary disease, up to 30–40% will eventually fail, and the disease will manifest through biochemical recurrence (BCR) [3,4]. Of these BCR patients who are treated with hormonal therapies approximately 10–20% will develop castrate-resistant cancer within 5 years [5]. Metastatic, castrate-resistant prostate cancer (mCRPC) is a highly aggressive stage of the disease, and has a prognosis of 9–13 months’ survival [5]. Due to this poor survival rate of mCRPC, alternate avenues of treatment are being investigated.
Prostate cancer tissue is composed of tumour cells and host components such as immune cells, stromal matrix and soluble factors (e.g., cytokines) with the host components referred to as the Tumour Immune Microenvironment (TIME). Within the TIME, crosstalk occurs between the immune cells, stromal cells, the non-cellular components and the tumour cells, resulting not only in the evolution of the TIME, but also playing a role in tumour progression, tumour clearance or treatment response The TIME interacts with soluble factors secreted by the cancer cells, and in turn, also interacts with the tumour cells. Importantly, whilst providing structural support and contact with prostate cancer cells, the TIME also produce soluble factors, all of which combined can drive prostate cancer progression [6]. Traditionally, it is believed that tumour-intrinsic signalling pathways are oncogenic pathways, however, emerging evidence is showing that this signalling can also regulate the TIME and subsequently tumour immune escape [7,8]. In prostate cancer, this can include PI3K/PTEN/AKT signalling [9,10], TLR9 [11] and p53 loss of function [12], which drives the accumulation, expansion, infiltration and activation of MDSCs. However, immune cells within the prostate cancer TIME act as a double-edged sword, because across the various stages of the disease, the immune cells can also mediate their invasive capacity.
Immunotherapies have shown substantial benefit in other cancers, however, there have been challenges in overcoming the immunosuppressive tumour microenvironment of prostate cancer.

2. The Prostate Tumour Immune Microenvironment

Immune evasion is a hallmark of cancer and dysregulation of the immune microenvironment contributes to malignant progression in prostate cancer [13]. The prostate tumour microenvironment consists of three main compartments: the stroma, the tumour and the immune cells (Figure 1). Together, cellular populations, nutrients and signalling molecules generate a highly immunosuppressive tumour microenvironment.

2.1. The Stromal Compartment

The stromal compartment is inherently plastic and can rapidly respond to damage sustained by the adjacent epithelium. When responding to such damage, stromal cells are phenotypically and genotypically altered, and there is increased matrix remodelling and altered expression of repair-associated growth factors and cytokines. This state is known as reactive stroma [14,15]. In prostate cancer development, reactive stroma initiates during pre-malignant prostatic intraepithelial neoplasia, and co-evolves as the cancer develops [16]. Carcinoma-associated fibroblasts (CAFs) are the main type of cells present in reactive stroma and are central in mediating pro-survival signalling in cancer cells. In terms of the immune microenvironment, CAF proliferation has been shown to lead to the development of a fibrous stroma, which induces localized vasculature remodelling and a state of hypoxia and chronic inflammation [17]. Chronic inflammation is akin to a pre-cancerous state inducing re-modelling of the normal tissue environment, where NF-κB signalling pathways play a defining role. Here, NF-κB-controlled signalling networks modulate the expression of cascades of pro-inflammatory genes, particularly cytokines and chemokines, and also regulate inflammasome formation. In response, immunosuppressive cell populations are recruited to the microenvironment, while cytotoxic T-cell function and dendritic cell maturation are inhibited [18,19,20,21]. The transcription factor (HIF-1) which is regulated by oxygen, is also overexpressed in prostate cancer, and is correlated to the clinical stage of the disease [22,23]. Importantly, hypoxia can mediate prostatic adenocarcinoma cell plasticity with the acquisition of a mesenchymal phenotype in a process known as epithelial–mesenchymal transition (EMT). This process can also contribute to immune escape via a loss of cell–cell recognition, as observed with decreased e-cadherin modulating the T-cell synapse which is required for an efficient immune response [24]. In addition, mesenchymal cells exhibit decreased MHC1 expression levels which promotes differentiation/recruitment of T-regulatory lymphocytes (Tregs), immature DCs and ultimately tumour immunosuppression [24].

2.2. The Immune Cell Compartment

The prostate tumour microenvironment has altered levels of the various classes of immune cells compared to healthy prostate tissue. This includes tumour-associated macrophages (TAMs), T cells and neutrophils [25]. Interestingly, increased levels of inactive immune cells such as resting natural killer cells, naive B cells and resting dendritic cells are present, which may suggest that the prostate cancer microenvironment inhibits the activation of these cells [25].
The prostate cancer TIME is traditionally regarded as immunologically ‘cold’ due to its relatively low levels of infiltrating T lymphocytes [26]. This is in part due to the low tumour mutational burden observed in prostate tumours compared to tumours such as melanoma and renal cell carcinoma, thus reducing the presence of tumour neoantigens [25,27]. This, paired with the frequent loss or reduction in major histocompatibility complex (MHC) class I and II expression in antigen-presenting cells, means that the number of anti-tumour T cells being attracted to the tumour is minimal [28,29,30,31]. Further contributing to the immunosuppressive microenvironment is the lack of afferent lymphatics to the prostate and the immunosuppressive properties of seminal fluid [32].
In prostate cancer, T cells are the major class of tumour-infiltrating lymphocytes. Higher numbers of infiltrating T cells have been correlated with better clinical outcomes in a variety of solid tumours, including bladder cancer, colorectal cancer, ovarian cancer and melanoma [33,34,35,36]. In contrast, the prognostic value of tumour T-cell infiltration is controversial, with both very low and high levels being associated with worse clinical outcomes in prostatectomy specimens [37,38,39,40]. This suggests that in cases with low levels of T cells infiltrating, the immune system has failed to mount an effective anti-tumour response. On the other hand, cases with high levels of T-cell infiltration may suggest that the T cells being recruited are able to function effectively. Alternatively, there may be a defect in recruiting the appropriate T-cell subpopulations. For example, these patients might have high infiltration of Tregs. Tregs play an important role in downregulating the cytotoxic T-cell response, and high levels of FOXP3+ Treg infiltration is a negative predictor of overall survival in a range of cancers [41]. High levels of FOXP3+ Tregs have been identified in prostate cancer tumour samples and peripheral blood samples, and may in part explain the dysfunction observed in the cytotoxic CD8+ T cells [42,43].
CD8+ T cells have potent cytotoxic activity and play a key role in anti-tumour immunity. The prognostic value of CD8+ T-cell infiltration in prostate cancer is inconclusive. Several studies have suggested that high CD8+ T-cell infiltration in the tumour epithelium has been reported to be associated with improved overall survival or lower risk of disease relapse and progression in prostatectomy specimens [44,45]. Another study concluded that higher CD8+ and lower programmed death ligand 1 (PD-L1) expression was associated with lower risk of biochemical recurrence and metastasis development [46]. However, others have shown that high levels of CD8+ T cells are associated with worse prognosis [47,48]. One of these studies hypothesised that the failure of high levels of CD8+ T cells to reduce disease recurrence may be due to T-cell exhaustion, and associated expression of PD-1. Interestingly, another study demonstrated that low levels of PD-L1, an immune-suppression marker and ligand of PD-1, combined with high levels of CD8+ T cells, was associated with improved prognosis [46]. This suggests that T-cell exhaustion may play a role in this finding, given the importance of the PD-1/PD-L1 axis in T-cell activation.
Myeloid-derived suppressor cells (MDSCs) and TAMs are key inflammatory cells which also contribute to the immunosuppressive prostate cancer TIME. MDSCs are activated by Tregs and exert their immunosuppressive functions by depletion of arginine and tryptophan in the surrounding tissue. Ultimately, this leads to T-cell cell-cycle arrest and decreased expression of T-cell receptors [49,50]. TAMs can be stratified into two distinct subpopulations, however, the majority of TAMS present in prostate cancer are M2-like, which are associated with an anti-inflammatory phenotype [51]. The presence of M2-like TAMS in both epithelial and stromal compartments is associated with tumour aggressiveness and poorer patient outcomes [52,53].

2.3. Cytokines and Signalling Molecules

A plethora of cytokines are present in the TIME and play an important role in promoting tumorigenesis and regulating the immunosuppressive environment. One example is transforming growth factor-β (TGF-β). In healthy systems, TGF-β acts as a tumour suppressor, where it inhibits proliferation and induces apoptosis. However, when overexpressed in cancers, such as prostate cancer, TGF-β becomes a potent promoter of tumour invasiveness and metastasis [54,55]. One way it mediates this is through promotion of tumour immune evasion through suppression of proliferation and differentiation of lymphocytes, natural killer cells and macrophages [56,57]. On the other hand, anti-tumour cytokines, such as type I interferon (IFN), and their respective signalling pathways, are often suppressed in prostate cancer, especially in the metastatic state [58]. As IFN is important to the coordination of the immune response, the reactivation of the pathway with therapy may promote long-term anti-tumour immunity.

3. Immunotherapy Strategies in Prostate Cancer

Immunotherapy aims to enhance the adaptive immune response, either through enhancing specificity or promoting stronger activation against the tumour. This approach has found success in a range of cancers. However, the immunologically ‘cold’ nature of prostate cancer has made the development of effective immunotherapies more challenging. Clinical trials currently investigating the use of immunotherapies in prostate cancer patients are summarized in Supplementary Table S1.

3.1. Cancer Vaccines

One class of immunotherapies is cancer vaccines. Cancer vaccines prime the patient’s immune system to recognise tumour-associated antigens (Figure 2A). Numerous types of cancer vaccines have been trialled in the prostate cancer setting, including autologous and allogeneic cellular vaccines to stimulate the function of antigen presenting cells, DNA and peptide vaccines which deliver engineered nucleic acids mimicking prostate tumour antigens, and oncolytic virus vaccines which cause direct lysis of tumour cells allowing for the release of a broad range of tumour antigens [59].
Sipuleucel-T is the only FDA-approved immunotherapy for mCRPC. Sipuleucel-T is an autologous cellular cancer vaccine which targets the immune system against prostatic acid phosphatase [60]. Sipuleucel-T treatment significantly increased median overall survival in mCRCP patients [61,62]. Of note, the greatest survival benefits were observed in patients with lower PSA [63]. This highlights that treatment may be more beneficial in the early disease stages, as newly activated cytotoxic T cells have more time to function [64,65]. The importance of an activated immune response is further highlighted by the fact that patients treated with Sipuleucel-T exhibited a 3-fold increase in the presence of activated effector T cells in the tumour microenvironment [64].
Additional vaccines which exhibit signs of efficacy in prostate cancer include PROSTVAC, GVAX and DCVAC/PCa. PROSTVAC is a virus-based vaccine which targets PSA and employs a triad of co-stimulatory molecules (TRICOM; CD-80, ICAM-1, LFA-3) which aid T-cell function, ultimately eliciting a robust immune response [66]. Despite promising initial results in mCRPC, other trials have failed to demonstrate its associated benefits as a monotherapy [67,68]. Similarly, despite promising results of improved patient survival in phase I and II trials, phase III trials with the allogenic GVAX in mCRPC demonstrated poor results and were terminated early [69,70,71].

3.2. Immune Checkpoint Inhibitors

In order to prevent autoimmunity, healthy cells display proteins known as immune checkpoint molecules. When a T cell binds these, an ‘off’ signal is sent to prevent T-cell-mediated destruction of the healthy cell. In cancer, however, these immune checkpoints are often upregulated and enhance immune evasion by the tumour. Immune checkpoint inhibitor (ICIs) monoclonal antibodies work by stopping this ‘off’ signal, and therefore allow the tumour to be targeted by the T cells (Figure 3) [72,73].
Ipilimumab is an ICI that targets CTLA-4, which has been trialled in mCRPC. In mCRPC patients who had failed docetaxel therapy, treatment with ipilimumab, alone or in combination with radiation therapy, resulted in increased median progression-free survival. However, no benefit to overall survival was observed [74]. The benefit was stronger in patients with favourable prognostic factors, especially the absence of visceral metastases. However, a follow-up study in asymptomatic or minimally symptomatic mCRPC patients without visceral metastases did not demonstrate any significant benefit to overall survival [75]. Similarly, anti-PD-1 and anti-PD-L1 agents, such as pembrolizumab, avelumab and nivolumab have been trialled in heavily pre-treated mCRPC. In a phase I trial of nivolumab, an objective response was not observed, while a phase 1 trial of aveulamab reported that 39% of patients had stable disease at >24 weeks [76]. In the same clinical settings, pembrolizmab treatment demonstrated an objective response rate of 17% [77].

3.3. Chimeric Antigen Receptor T Cells

Chimeric Antigen Receptor T Cells (CAR-T) are genetically enhanced T cells modified to engage with specific patient tumour antigens [78], (Figure 2B). This approach has found success in haematological cancers but has shown limited efficacy in the treatment of solid tumours. CAR-T cells targeting PSMA have been developed for the treatment of prostate cancer. This approach has had limited success in clearing tumours, but has demonstrated tumouristatic effects in some preliminary studies [79,80]. Newer generations of CAR-T therapy are integrating elements to counteract the immunosuppressive environment. One study looking at PSMA-directed/TGF-β-insensitive CAR-T cells has reported promising initial results [81,82].

3.4. Bispecific Antibodies

Bispecific antibodies consist of two monoclonal antibodies known as bispecific T-cell engagers (BiTEs) attached by a flexible linker (Figure 2C). These antibodies conjugate simultaneously with tumour antigens and the T cell to promote cytotoxic 4T-cell trafficking and function [83]. A phase I study using the BiTE Pasotuxizumab, which engages CD3 on T cells and PSMA, in androgen deprivation therapy (ADT) and chemotherapy-refractory mCRPC reported positive results, with 88% of patients exhibiting a PSA response in a dose-dependent manner. Interestingly, this resulted in a long-term response in 12.5% of the patients [84]. Another PMSA targeting BiTE, AMG 160, has resulted in a greater than 50% decline in PSA in about one-third of patients, with two patients showing a partial response and eight patients a stable disease (NCT03792841). Trispecific approaches are also being investigated, as demonstrated by the phase I study investigating the safety profile of HPN24 in mCRPC patients who have progressed in systemic therapy (NCT03577028). This is a CD3-PSMA-targeting monoclonal antibody with an albumin-binding domain for extending the half-life of the compound.

3.5. Combination Therapy Strategies

Given the lack of success in finding a single-agent immunotherapy for prostate cancer, the focus has shifted to identifying combination therapies. This includes combining with standard therapies as well as identifying useful dual-immunotherapy approaches.
As chemotherapy is one of the main treatments for cancer, there is strong interest in combining it with immunotherapies, and this approach has yielded early promise. A study of nivolumab plus docetaxel in chemotherapy-native mCRPC patients with ongoing ADT therapy reported an objective response rate of 40% and a median overall survival of 18 months following treatment [85]. Interim analysis from a second study of pembrolizumab and docetaxel and prednisone in mCRPC previously treated with ADT reported an objective response rate of 23.1% [86]. It has been suggested that chemotherapy releases neoantigens from the tumour upon cell death and these neoantigens promote the activation of CD8+ cells, thus promoting the efficacy of immunotherapies.
Like chemotherapy, poly-ADP ribose polymerase (PARP) inhibitors promote the necrotic release of tumour neoantigens. Additionally, like chemotherapy, their effectiveness as part of a combination therapy with immunotherapies has also been investigated. Most promisingly, treatment with durbalumab and PARP inhibitor, oliparib, in mCRPC patients who had progressed with androgen deprivation therapy reported that 53% of patients had a radiographic and/or PSA response [87]. An interim analysis of a second study reports a PSA response in 9%, and a partial response in 8% of those with measurable disease in mCRPC patients previously treated with docetaxel, following treatment with pembrolizumab and oliparib [88].
Another combination of interest is treatment with androgen-deprivation therapy and immunotherapies. It has been shown that ADT results in increased trafficking of anti-tumour immune cells, and as such, ADT may enhance the effectiveness of immunotherapies [89]. Across multiple studies, treatment with pembrolizumab in patients with mCRPC who had previously been treated with ADT reported a disease control rate of between 35 and 51%, however, one study did not show any benefit of therapy [90,91,92].
The combination of anti-angiogenic therapies with those of immunotherapies has also recently attracted interest. Here, it has been shown that tumour angiogenesis-targeted agents such as Sunitinib can induce a pro-immunogenic state in the tumour by inducing miR-221 expression. This is then thought to cause an induction of an interferon-related gene signature in the prostate cancer cells supported by miR-221 upregulation [93]. These data would suggest that the treatment combination of tumour kinase inhibitors (TKIs) and immune-based approaches might be a promising avenue to explore with TKIs treatment, inducing an immune responsive environment due to boosting miR-221 expression levels.
One of the main immunotherapy combination strategies under investigation is co-treatment with anti-PD-1 and anti-CTLA-4 agents. A study on asymptomatic or minimally symptomatic mCRPC patients not previously treated with ipilimumab and nivolumab showed an objective response rate of 26% at interim reporting, while in those who had progressed with chemotherapy, the objective response rate was 10% [94,95]. Ipilimumab and nivolumab have also been trialled in a subgroup of mCRPC patients positive for AR-V7. Outcomes tended to be better in a subset of patients, specifically in those with DNA damage repair mutations, however this was only significantly different in terms of progression-free survival [96]. This highlights the importance of biomarkers to enable selection of patients who will respond to therapy.

4. Immune Therapy Biomarkers

Metastatic prostate cancer presents a range of challenges to treatment with immunotherapy. Early studies on the use of these agents in prostate cancer have been characterised by a lack of response in a high percentage of patients. However, there is evidence that some subpopulations are highly responsive to immunotherapy. Therefore, it will likely be pertinent to identify improved molecular biomarkers that will allow for the selection of those patients that are most likely to benefit.

4.1. Immune Checkpoint Molecules as Biomarkers

ICI directly targets the immune checkpoint molecules; therefore, it follows that the expression of such molecules may be an important determinant of therapy response. PD-L1 levels have been shown to have varying degrees of association with therapy, with expression correlated with ICI response in melanoma but not in squamous cell carcinoma and non-small-cell lung cancer [97,98,99]. The expression of PD-L1 and PD-1 is low in healthy prostate tissue, present in around 0.5% and 1.5% of cases, respectively, and is increased in prostate cancer cases, with one study reporting positive staining in 13.2% and 7.7%, respectively [100]. Although dynamic expression is observed, high PD-L1 expression is associated with prostate cancer aggressiveness, with 61.7% of aggressive primary prostate tumours and 50% of CRPC expressing PD-L1 [101,102]. Despite this, PD-L1 expression does not strongly predict response to ICI in prostate cancer. A study examining pembrolizumab efficacy in PD-L1-positive versus PD-L1-negative cohorts of treatment-refractory mCRPC patients reported objective response rates of 5% and 3%, respectively [103]. This suggests that other factors contribute to the effectiveness of anti-PD-L1 therapy in these patients and that simple measurement of PD-L1 levels in tissue may not be an effective biomarker of treatment response.

4.2. Genetic Variations as Biomarkers

Genetic mutations present in a tumour can be a powerful predictor of therapy response. Prostate cancer has traditionally been difficult to target with immunotherapy due to its low number of mutations and therefore presence of fewer neoantigens, especially in relation to other highly mutated cancers such as melanoma and lung cancer [27].
Patients with high microsatellite instability (MSI-H) or mismatch-repair-deficient (MMRd) tumours have a high tumour mutational burden (TMB) and increased presence of tumour neoantigens, and are therefore strong candidates for immune checkpoint inhibitor immunotherapy [104,105]. The ICI pembrolizumab is FDA-approved for use in metastatic/unresectable MSI-H/MMRd tumours, however it is only effective in a subset of prostate cancer patients [106]. A retrospective study reported durable clinical benefit in 45.5% of MSI-H/MMRd prostate tumours treated with anti-PD1 or anti-PDL1 agents, either as monotherapy or combination therapy [107]. In addition, a second study observed increased overall survival in patients with higher-than-median TMB following ICI therapy [94].
Increased neoantigen expression and higher TMB is also associated with mutations in specific genes, particularly those involved in DNA damage repair such as BRCA1/2, ATM, MSH2, POLE and CDK12 [108,109,110,111]. A comprehensive analysis of POLE/POLD1 mutation in multiple cancer types reported that 1.8% of primary prostate tumours had mutations in one or both genes, correlating with a significantly higher TMB. Patients with POLE/POLD1 mutations have been shown to have longer overall survival following ICI treatment across all tumour types, with POLE/POLD1 reported as an independent risk factor for identifying prostate cancer patients who benefited from ICI [110].
CDK12 mutations are observed in 1.2% of primary prostate tumours, rising to 6.9% of mCRPC. A unique mutation signature was observed for CDK12-deficient tumours, differentiating it distinctly from MMRd prostate tumours [105]. PSA decline following ICI treatment has been observed in patients with CDK-12 mutations in a number of studies [105,111,112,113]. Interestingly, there was no difference observed between patients with mono- versus bi-allelic mutations in PSA decline, however, PSA-progression-free survival was marginally improved for those with biallelic mutations [113]. These retrospective studies indicate that prospective randomized trials incorporating genomic screening and selection of patients with MSI-H/MMRd prostate tumours with higher-than-median TMB or CDK12 mutations would be highly informative as to the usefulness of treating these patients with ICI-based therapies. At the present time, this combined strategy would appear to be the most promising of the genomic-based approaches to enhance ICI response rates in prostate cancer.

4.3. Peripheral Immune-Based Biomarkers

Traditionally, mutational status of tumours has been assessed via biopsy, however biopsy acquisition is an invasive procedure that is not always practical for cancer patients. In contrast, blood samples are relatively easy to obtain and are being investigated as a potentially rich source of biomarkers.
One such source of biomarkers is circulating tumour DNA (ctDNA). ctDNA-based assays are becoming increasingly popular as an alternative to biopsy for the detection of actionable genetic mutations. A case series reported the detection of MSI-H status in two prostate cancer patients via ctDNA samples. Importantly, they were able to monitor the response to treatment by using the frequency of variant alleles in blood samples as a readout, since it can be repeated for real-time monitoring of tumour clones [114]. This demonstrates the utility of such technologies as biomarkers of potential treatment efficacy and in monitoring response.
Cytokines are key regulators of the immune response; therefore, several studies have explored cytokine levels at baseline and their correlation with response to immunotherapy treatment, in particular treatment with cancer vaccines. One study suggests that higher baseline circulating interleukin-10 (IL-10) levels in CRPC patients may be negatively associated with response to treatment with a DNA vaccine encoding prostatic acid phosphatase [115]. A second study demonstrated that higher baseline levels of interleukin-6 (IL-6) was associated with shorter survival in patients treated with the personalized peptide vaccine [116].
The presence of various immune cell populations, such as T cells and MDSCs, have also been investigated for their merit as predictive biomarkers for clinical response to immunotherapy. Lower baseline levels of PD-1+Tim-3NEG CD4 effector memory cells and higher baseline PD-1NEGTim-3+ CD8 and CTLA-4NEG Tregs in mCRPC predicted improved survival following treatment with PROSTVAC and ipilimumab [117]. Similarly, higher baseline levels of CD4+CTLA-4+ T cells predicted improved survival when treated with GVAX and ipilimumab. In contrast, CRPC patients with higher CD14+HLA-DR-monocytic MDSCs had worse survival following treatment [118]. There is also evidence that early changes in circulating immune cells can be used to monitor the response and adjust treatments accordingly, as a study reported that in mCRPC patients treated with DCVac and docetaxel an on-treatment decrease in MDSC independently predicted disease-specific survival [119].

5. Conclusions

As the second most commonly diagnosed cancer in males, prostate cancer is a major health concern. In particular, metastatic, castrate-resistant prostate cancer patients have poor survival rates. Immunotherapies have seen success in many other cancers; however, the treatment of prostate cancer through these methods suffers due to the immunosuppressive tumour microenvironment. Recent studies have helped elucidate many of the immune-suppressive mechanisms at play in prostate cancer. This knowledge, in combination with biomarkers, can help inform treatment selection and allow these mechanisms to be overcome through combination therapy strategies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/uro2020010/s1, Table S1: Current immunotherapy clinical trials in prostate cancer [120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159].

Author Contributions

Writing—original draft preparation, Y.Z., B.K.C.; writing—review and editing, B.K.C., S.S.S., N.M.C., C.M.H.; supervision, N.M.C., C.M.H. All authors have read and agreed to the published version of the manuscript.

Funding

Support for this work was provided through the NHMRC project grant 1162514 to C.M.H. and the PRECEPT program grant, co-funded by Movember and the Australian Federal Government to N.M.C. N.M.C. was supported by a David Bickart Clinician Researcher Fellowship from the Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, and more recently by a Movember—Distinguished Gentleman’s Ride Clinician Scientist Award through the Prostate Cancer Foundation of Australia’s Research Program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADT (Androgen-deprivation therapy); AKT (Ak strain transforming); ATM (ataxia telangiectasia mutated); BCR (biochemical recurrence); BRCA1 (Breast Cancer gene 1); BRAC2 (Breast Cancer Gene 2); CAFS (carcinoma-associated fibroblasts); CAR-T (Chimeric antigen receptor T cells); CD3 (cluster of differentiation 3); CD38 (cluster of differentiation 38); CD39 (cluster of differentiation 39); CD73 (cluster of differentiation 73); CDK12 (cyclin-dependent kinase 12); CRPC (castrate-resistant prostate cancer); CtDNA (circulating tumour DNA); (CTLA-4 cytotoxic T-lymphocyte-associated protein 4); DCs (dendritic cells); DLL3 (delta-like ligand 3); FDA (Food and Drug Administration); FOXP3 (Forkhead box P3); DNA (deoxyribonucleic acid); ICAM-1 (intercellular adhesion molecule-1); ICI (immune checkpoint inhibitor); IFN (interferon); IL-6 (interleukin 6); IL-8 (interleukin 8); IL-10 (interleukin 10); LFA-3 (Lymphocyte Function-Associated Antigen 3); mCRPC (metastatic, castrate-resistant prostate cancer); MDSCs (myeloid-derived suppressor cells); MHC (major histocompatibility complex); MMRd (mismatch repair deficient); MSH-I (high microsatellite instability); NK (natural killer cells); MSH2 (MutS homolog 2); PAP (prostatic acid phosphatase); PARP (poly-ADP ribose polymerase); (PD-1 (Programmed cell death protein 1); PD-L1 (Programmed death-ligand 1); PI3K (phosphoinositide 3-kinases); POLD1 (DNA Polymerase Delta 1); POLE (DNA polymerase epsilon); PSA (prostate-specific antigen); PSMA (Prostate Specific Membrane Antigen); PTEN (phosphatase and tensin homolog); STEAP1 (six-transmembrane epithelial antigen of prostate); TAMS (tumour-associated macrophages); TCR (T-cell receptor); TGF-β (transforming growth factor-β); TGF-β RII (transforming growth factor-β receptor II); TIME (tumour immune microenvironment); TKI (tyrosine kinase inhibitor); Tregs (regulatory T cells); Tumour mutational burden (TMB)

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Islami, F.; Ward, E.M.; Sung, H.; Cronin, K.A.; Tangka, F.K.L.; Sherman, R.L.; Zhao, J.; Anderson, R.N.; Henley, S.J.; Yabroff, K.R.; et al. Annual Report to the Nation on the Status of Cancer, Part 1: National Cancer Statistics. JNCI J. Natl. Cancer Inst. 2021, 113, 1648–1669. [Google Scholar] [CrossRef] [PubMed]
  3. Freedland, S.J.; Humphreys, E.B.; Mangold, L.A.; Eisenberger, M.; Dorey, F.J.; Walsh, P.C.; Partin, A.W. Risk of prostate cancer-specific mortality following biochemical recurrence after radical prostatectomy. J. Am. Med. Assoc. 2005, 294, 433–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Roehl, K.A.; Han, M.; Ramos, C.G.; Antenor, J.A.V.; Catalona, W.J. Cancer progression and survival rates following anatomical radical retropubic prostatectomy in 3,478 consecutive patients: Long-term results. J. Urol. 2004, 172, 910–914. [Google Scholar] [CrossRef] [Green Version]
  5. Kirby, M.; Hirst, C.; Crawford, E.D. Characterising the castration-resistant prostate cancer population: A systematic review. Int. J. Clin. Pract. 2011, 65, 1180–1192. [Google Scholar] [CrossRef]
  6. Keller, E.T.; Zhang, J.; Cooper, C.R.; Smith, P.C.; McCauley, L.K.; Pienta, K.J.; Taichman, R.S. Prostate carcinoma skeletal metastases: Cross-talk between tumor and bone. Cancer Metastasis Rev. 2001, 20, 333–349. [Google Scholar] [CrossRef] [Green Version]
  7. Altorki, N.K.; Markowitz, G.J.; Gao, D.; Port, J.L.; Saxena, A.; Stiles, B.; McGraw, T.; Mittal, V. The lung microenvironment: An important regulator of tumour growth and metastasis. Nat. Rev. Cancer 2019, 19, 9–31. [Google Scholar] [CrossRef] [Green Version]
  8. Spranger, S.; Gajewski, T.F. Tumor-intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology 2016, 5, e1086862. [Google Scholar] [CrossRef] [Green Version]
  9. Garcia, A.J.; Ruscetti, M.; Arenzana, T.L.; Tran, L.M.; Bianci-Frias, D.; Sybert, E.; Priceman, S.J.; Wu, L.; Nelson, P.S.; Smale, S.T.; et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression. Mol. Cell. Biol. 2014, 34, 2017–2028. [Google Scholar] [CrossRef] [Green Version]
  10. Wang, G.; Lu, X.; Dey, P.; Deng, P.; Wu, C.C.; Jiang, S.; Fang, Z.; Zhao, K.; Konaparthi, R.; Hua, S.; et al. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov. 2016, 6, 80–95. [Google Scholar] [CrossRef] [Green Version]
  11. Won, H.; Moreira, D.; Gao, C.; Duttagupta, P.; Zhao, X.; Manuel, E.; Diamond, D.; Yuan, Y.; Liu, Z.; Jones, J.; et al. TLR9 expression and secretion of LIF by prostate cancer cells stimulates accumulation and activity of polymorphonuclear MDSCs. J. Leukoc. Biol. 2017, 102, 423–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Guo, G.; Marrero, L.; Rodriguez, P.; Del Valle, L.; Ochoa, A.; Cui, Y. Trp53 inactivation in the tumor microenvironment promotes tumor progression by expanding the immunosuppressive lymphoid-like stromal network. Cancer Res. 2013, 73, 1668–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
  14. Rowley, D.R. What might a stromal response mean to prostate cancer progression? Cancer Metastasis Rev. 1998, 17, 411–419. [Google Scholar] [CrossRef] [PubMed]
  15. Tuxhorn, J.A.; Ayala, G.E.; Smith, M.J.; Smith, V.C.; Dang, T.D.; Rowley, D.R. Reactive stroma in human prostate cancer: Induction of myofibroblast phenotype and extracellular matrix remodeling. Clin. Cancer Res. 2002, 8, 2912–2923. [Google Scholar] [PubMed]
  16. Tuxhorn, J.A.; Ayala, G.E.; Rowley, D.R. Reactive stroma in prostate cancer progression. J. Urol. 2001, 166, 2472–2483. [Google Scholar] [CrossRef]
  17. Ammirante, M.; Shalapour, S.; Kang, Y.; Jamieson, C.A.M.; Karin, M. Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc. Natl. Acad. Sci. USA 2014, 111, 14776–14781. [Google Scholar] [CrossRef] [Green Version]
  18. Huang, Y.; Yuan, J.; Righi, E.; Kamoun, W.S.; Ancukiewicz, M.; Nezivar, J.; Santosuosso, M.; Martin, J.D.; Martin, M.R.; Vianello, F.; et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl. Acad. Sci. USA 2012, 109, 17561–17566. [Google Scholar] [CrossRef] [Green Version]
  19. Gabrilovich, D.; Ishida, T.; Oyama, T.; Ran, S.; Kravtsov, V.; Nadaf, S.; Carbone, D.P. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 1998, 92, 4150–4166. [Google Scholar] [CrossRef]
  20. Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 1996, 2, 1096–1103. [Google Scholar] [CrossRef]
  21. Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.L.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C.; et al. VEGF-A modulates expression of inhibitory checkpoints on CD8++ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef] [PubMed]
  22. Movsas, B.; Chapman, J.D.; Greenberg, R.E.; Hanlon, A.L.; Horwitz, E.M.; Pinover, W.H.; Stobbe, C.; Hanks, G.E. Increasing levels of hypoxia in prostate carcinoma correlate significantly with increasing clinical stage and patient age: An Eppendorf pO2 study. Cancer 2000, 89, 2018–2024. [Google Scholar] [CrossRef]
  23. Turaka, A.; Buyyounouski, M.K.; Hanlon, A.L.; Horwitz, E.M.; Greenberg, R.E.; Movsas, B. Hypoxic prostate/muscle Po 2 ratio predicts for outcome in patients with localized prostate cancer: Long-term results. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, e433–e439. [Google Scholar] [CrossRef] [PubMed]
  24. Terry, S.; Savagner, P.; Ortiz-Cuaran, S.; Mahjoubi, L.; Saintigny, P.; Thiery, J.P.; Chouaib, S. New insights into the role of EMT in tumor immune escape. Mol. Oncol. 2017, 11, 824–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. 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. [Google Scholar] [CrossRef]
  26. Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold tumors: A therapeutic challenge for immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef] [Green Version]
  27. Berger, M.F.; Lawrence, M.S.; Demichelis, F.; Drier, Y.; Cibulskis, K.; Sivachenko, A.Y.; Sboner, A.; Esgueva, R.; Pflueger, D.; Sougnez, C.; et al. The genomic complexity of primary human prostate cancer. Nature 2011, 470, 214–220. [Google Scholar] [CrossRef] [Green Version]
  28. Sharpe, J.C.; Abel, P.D.; Gilbertston, J.A.; Brawn, P.; Foster, C.S. Modulated expression of human leucocyte antigen class I and class II determinants in hyperplastics and malignant human prostatic epithelium. Br. J. Urol. 1994, 74, 609–616. [Google Scholar] [CrossRef]
  29. Blades, R.A.; Keating, P.J.; McWilliam, L.J.; George, N.J.R.; Stern, P.L. Loss of HLA class I expression in prostate cancer: Implications for immunotherapy. Urology 1995, 46, 681–687. [Google Scholar] [CrossRef]
  30. Blumenfeld, W.; Ye, J.Q.; Dahiya, R.; Griffiss, J.M.; Narayan, P. HLA expression by benign and malignant prostatic epithelium: Augmentation by interferon-gamma. J. Urol. 1993, 150, 1289–1292. [Google Scholar] [CrossRef]
  31. Bander, N.H.; Yao, D.; Liu, H.; Chen, Y.T.; Steiner, M.; Zuccaro, W.; Moy, P. MHC class I and II expression in prostate carcinoma and modulation by interferon-alpha and -gamma. Prostate 1997, 33, 233–239. [Google Scholar] [CrossRef]
  32. McClinton, S.; Miller, I.D.; Eremin, O. An immunohistochemical characterisation of the inflammatory cell infiltrate in benign and malignant prostatic disease. Br. J. Cancer 1990, 61, 400–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bogunovic, D.; O’Neill, D.W.; Belitskaya-Levy, I.; Vacic, V.; Yu, Y.L.; Adams, S.; Darvishian, F.; Berman, R.; Shapiro, R.; Pavlick, A.C.; et al. Immune profile and mitotic index of metastatic melanoma lesions enhance clinical staging in predicting patient survival. Proc. Natl. Acad. Sci. USA 2009, 106, 20429–20434. [Google Scholar] [CrossRef] [Green Version]
  34. Simpson, J.A.D.; Al-Attar, A.; Watson, N.F.S.; Scholefield, J.H.; Ilyas, M.; Durrant, L.G. Intratumoral T cell infiltration, MHC class I and STAT1 as biomarkers of good prognosis in colorectal cancer. Gut 2010, 59, 926–933. [Google Scholar] [CrossRef] [PubMed]
  35. Oble, D.A.; Loewe, R.; Yu, P.; Mihm, M.C. Focus on TILs: Prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun. 2009, 9, 3. [Google Scholar] [PubMed]
  36. Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T Cells, Recurrence, and Survival in Epithelial Ovarian Cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Flammiger, A.; Bayer, F.; Cirugeda-Kühnert, A.; Huland, H.; Tennstedt, P.; Simon, R.; Minner, S.; Bokemeyer, C.; Sauter, G.; Schlomm, T.; et al. Intratumoral T but not B lymphocytes are related to clinical outcome in prostate cancer. APMIS 2012, 120, 901–908. [Google Scholar] [CrossRef]
  38. Kärjä, V.; Aaltomaa, S.; Lipponen, P.; Isotalo, T.; Talja, M.; Mokka, R. Tumour-infiltrating lymphocytes: A prognostic factor of psa-free survival in patients with local prostate carcinoma treated by radical prostatectomy. Anticancer Res. 2005, 25, 4435–4438. [Google Scholar]
  39. Vesalainen, S.; Lipponen, P.; Talja, M.; Syrjänen, K. Histological grade, perineural infiltration, tumour-infiltrating lymphocytes and apoptosis as determinants of long-term prognosis in prostatic adenocarcinoma. Eur. J. Cancer 1994, 30, 1797–1803. [Google Scholar] [CrossRef]
  40. Irani, J.; Goujon, J.M.; Ragni, E.; Peyrat, L.; Hubert, J.; Saint, F.; Mottet, N. High-grade inflammation in prostate cancer as a prognostic factor for biochemical recurrence after radical prostatectomy. Urology 1999, 54, 467–472. [Google Scholar] [CrossRef]
  41. Saleh, R.; Elkord, E. FoxP3+ T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett. 2020, 490, 174–185. [Google Scholar] [CrossRef] [PubMed]
  42. Miller, A.M.; Lundberg, K.; Özenci, V.; Banham, A.H.; Hellström, M.; Egevad, L.; Pisa, P. CD4 + CD25 high T Cells Are Enriched in the Tumor and Peripheral Blood of Prostate Cancer Patients. J. Immunol. 2006, 177, 7398–7405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kiniwa, Y.; Miyahara, Y.; Wang, H.Y.; Peng, W.; Peng, G.; Wheeler, T.M.; Thompson, T.C.; Old, L.J.; Wang, R.F. CD8+ Foxp3+ regulatory T cells mediate immunosuppression in prostate cancer. Clin. Cancer Res. 2007, 13, 6947–6958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yang, Y.; Attwood, K.; Bshara, W.; Mohler, J.L.; 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. [Google Scholar] [CrossRef]
  45. Sorrentino, C.; Musiani, P.; Pompa, P.; Cipollone, G.; Di Carlo, E. Androgen deprivation boosts prostatic infiltration of cytotoxic and regulatory T lymphocytes and has no effect on disease-free survival in prostate cancer patients. Clin. Cancer Res. 2011, 17, 1571–1581. [Google Scholar] [CrossRef] [Green Version]
  46. Vicier, C.; Ravi, P.; Kwak, L.; Werner, L.; Huang, Y.; Evan, C.; Loda, M.; Hamid, A.A.; Sweeney, C.J. Association between CD8 and PD-L1 expression and outcomes after radical prostatectomy for localized prostate cancer. Prostate 2021, 81, 50–57. [Google Scholar] [CrossRef]
  47. Ness, N.; Andersen, S.; Valkov, A.; Nordby, Y.; Donnem, T.; Al-Saad, S.; Busund, L.T.; Bremnes, R.M.; Richardsen, E. Infiltration of CD8+ lymphocytes is an independent prognostic factor of biochemical failure-free survival in prostate cancer. Prostate 2014, 74, 1452–1461. [Google Scholar] [CrossRef]
  48. Zhao, S.G.; Lehrer, J.; Chang, S.L.; Das, R.; Erho, N.; Liu, Y.; Sjöström, M.; Den, R.B.; Freedland, S.J.; Klein, E.A.; et al. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J. Natl. Cancer Inst. 2019, 111, 301–310. [Google Scholar] [CrossRef]
  49. Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M.; et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [Green Version]
  50. Rodriguez, P.C.; Quiceno, D.G.; Ochoa, A.C. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 2007, 109, 1568–1573. [Google Scholar] [CrossRef] [Green Version]
  51. Lundholm, M.; Hägglöf, C.; Wikberg, M.L.; Stattin, P.; Egevad, L.; Bergh, A.; Wikström, P.; Palmqvist, R.; Edin, S. Secreted factors from colorectal and prostate cancer cells skew the immune response in opposite directions. Sci. Rep. 2015, 5, 15651. [Google Scholar] [CrossRef]
  52. Lanciotti, M.; Masieri, L.; Raspollini, M.R.; Minervini, A.; Mari, A.; Comito, G.; Giannoni, E.; Carini, M.; Chiarugi, P.; Serni, S. The Role of M1 and M2 Macrophages in Prostate Cancer in relation to Extracapsular Tumor Extension and Biochemical Recurrence after Radical Prostatectomy. BioMed Res. Int. 2014, 2014, 486798. [Google Scholar] [CrossRef] [PubMed]
  53. Erlandsson, A.; Carlsson, J.; Lundholm, M.; Fält, A.; Andersson, S.O.; Andrén, O.; Davidsson, S. M2 macrophages and regulatory T cells in lethal prostate cancer. Prostate 2019, 79, 363–369. [Google Scholar] [CrossRef]
  54. Cao, Z.; Kyprianou, N. Mechanisms navigating the TGF-β pathway in prostate cancer. Asian J. Urol. 2015, 2, 11–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Haas, C.; Krinner, E.; Brischwein, K.; Hoffmann, P.; Lutterbüse, R.; Schlereth, B.; Kufer, P.; Baeuerle, P.A. Mode of cytotoxic action of T cell-engaging BiTE antibody MT110. Immunobiology 2009, 214, 441–453. [Google Scholar] [CrossRef] [PubMed]
  56. Donkor, M.K.; Sarkar, A.; Savage, P.A.; Franklin, R.A.; Johnson, L.K.; Jungbluth, A.A.; Allison, J.P.; Li, M.O. T cell surveillance of oncogene-induced prostate cancer is impeded by T cell-derived TGF-β1 cytokine. Immunity 2011, 35, 123–134. [Google Scholar] [CrossRef] [Green Version]
  57. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.L.; Flavell, R.A. Transforming growth factor-β regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  58. Owen, K.L.; Gearing, L.J.; Zanker, D.J.; Brockwell, N.K.; Khoo, W.H.; Roden, D.L.; Cmero, M.; Mangiola, S.; Hong, M.K.; Spurling, A.J.; et al. Prostate cancer cell-intrinsic interferon signaling regulates dormancy and metastatic outgrowth in bone. EMBO Rep. 2020, 21, e50162. [Google Scholar] [CrossRef]
  59. Paston, S.J.; Brentville, V.A.; Symonds, P.; Durrant, L.G. Cancer Vaccines, Adjuvants, and Delivery Systems. Front. Immunol. 2021, 12, 627932. [Google Scholar] [CrossRef]
  60. Thara, E.; Dorff, T.B.; Pinski, J.K.; Quinn, D.I. Vaccine therapy with sipuleucel-T (Provenge) for prostate cancer. Maturitas 2011, 69, 296–303. [Google Scholar] [CrossRef]
  61. Small, E.J.; Schellhammer, P.F.; Higano, C.S.; Redfern, C.H.; Nemunaitis, J.J.; Valone, F.H.; Verjee, S.S.; Jones, L.A.; Hershberg, R.M. Placebo-controlled phase III trial of immunologic therapy with Sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J. Clin. Oncol. 2006, 24, 3089–3094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kawalec, P.; Paszulewicz, A.; Holko, P.; Pilc, A. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. A systematic review and meta-analysis. Arch. Med. Sci. 2012, 8, 767–775. [Google Scholar] [CrossRef] [PubMed]
  63. Schellhammer, P.F.; Chodak, G.; Whitmore, J.B.; Sims, R.; Frohlich, M.W.; Kantoff, P.W. Lower baseline prostate-specific antigen is associated with a greater overall survival benefit from sipuleucel-T in the immunotherapy for prostate adenocarcinoma treatment (IMPACT) trial. Urology 2013, 81, 1297–1302. [Google Scholar] [CrossRef] [PubMed]
  64. Fong, L.; Carroll, P.; Weinberg, V.; Chan, S.; Lewis, J.; Corman, J.; Amling, C.L.; Stephenson, R.A.; Simko, J.; Sheikh, N.A.; et al. Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer. J. Natl. Cancer Inst. 2014, 106, dju268. [Google Scholar] [CrossRef] [PubMed]
  65. Hagihara, K.; Chan, S.; Zhang, L.; Oh, D.Y.; Wei, X.X.; Simko, J.; Fong, L. Neoadjuvant sipuleucel-T induces both Th1 activation and immune regulation in localized prostate cancer. Oncoimmunology 2019, 8, e1486953. [Google Scholar] [CrossRef] [PubMed]
  66. Gulley, J.L.; Madan, R.A.; Tsang, K.Y.; Jochems, C.; Marté, J.L.; Farsaci, B.; Tucker, J.A.; Hodge, J.W.; Liewehr, D.J.; Steinberg, S.M.; et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol. Res. 2014, 2, 133–141. [Google Scholar] [CrossRef] [Green Version]
  67. Kantoff, P.W.; Schuetz, T.J.; Blumenstein, B.A.; Michael Glode, L.; Bilhartz, D.L.; Wyand, M.; Manson, K.; Panicali, D.L.; Laus, R.; Schlom, J.; et al. Overall survival analysis of a phase II randomized controlled trial of a poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2010, 28, 1099–1105. [Google Scholar] [CrossRef]
  68. Gulley, J.L.; Borre, M.; Vogelzang, N.J.; Ng, S.; Agarwal, N.; Parker, C.C.; Pook, D.W.; Rathenborg, P.; Flaig, T.W.; Carles, J.; et al. Phase III Trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2019, 37, 1051–1061. [Google Scholar] [CrossRef]
  69. Higano, C.S.; Corman, J.M.; Smith, D.C.; Centeno, A.S.; Steidle, C.P.; Gittleman, M.; Simons, J.W.; Sacks, N.; Aimi, J.; Small, E.J. Phase 1/2 dose-escalation study of a GM-CSF-secreting, allogeneic, cellular immunotherapy for metastatic hormone-refractory prostate cancer. Cancer 2008, 113, 975–984. [Google Scholar] [CrossRef]
  70. Small, E.J.; Sacks, N.; Nemunaitis, J.; Urba, W.J.; Dula, E.; Centeno, A.S.; Nelson, W.G.; Ando, D.; Howard, C.; Borellini, F.; et al. Granulocyte macrophage colony-stimulating factor-secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clin. Cancer Res. 2007, 13, 3883–3891. [Google Scholar] [CrossRef] [Green Version]
  71. Higano, C.; Saad, F.; Somer, B.; Curti, B.; Petrylak, D.; Drake, C.G.; Schnell, F.; Redfern, C.H.; Schrijvers, D.; Sacks, N. A phase III trial of GVAX immunotherapy for prostate cancer versus docetaxel plus prednisone in asymptomatic, castration-resistant prostate cancer (CRPC). In Proceedings of the 2009 Genitourinary Cancers Symposium, Orlando, FL, USA, 26–28 February 2009; p. LBA150. [Google Scholar]
  72. He, X.; Xu, C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020, 30, 660–669. [Google Scholar] [CrossRef] [PubMed]
  73. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; Van den Eertwegh, A.J.M.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef] [Green Version]
  75. Beer, T.M.; Kwon, E.D.; Drake, C.G.; Fizazi, K.; Logothetis, C.; Gravis, G.; Ganju, V.; Polikoff, J.; Saad, F.; Humanski, P.; et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J. Clin. Oncol. 2017, 35, 40–47. [Google Scholar] [CrossRef] [PubMed]
  76. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti–PD-1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  77. Hansen, A.R.; Massard, C.; Ott, P.A.; Haas, N.B.; Lopez, J.S.; Ejadi, S.; Wallmark, J.M.; Keam, B.; Delord, J.P.; Aggarwal, R.; et al. Pembrolizumab for advanced prostate adenocarcinoma: Findings of the KEYNOTE-028 study. Ann. Oncol. 2018, 29, 1807–1813. [Google Scholar] [CrossRef] [PubMed]
  78. Ramos, C.A.; Dotti, G. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Expert Opin. Biol. Ther. 2011, 11, 855–873. [Google Scholar] [CrossRef]
  79. Junghans, R.P.; Ma, Q.; Rathore, R.; Gomes, E.M.; Bais, A.J.; Lo, A.S.Y.; Abedi, M.; Davies, R.A.; Cabral, H.J.; Al-Homsi, A.S.; et al. Phase I Trial of Anti-PSMA Designer CAR-T Cells in Prostate Cancer: Possible Role for Interacting Interleukin 2-T Cell Pharmacodynamics as a Determinant of Clinical Response. Prostate 2016, 76, 1257–1270. [Google Scholar] [CrossRef]
  80. Slovin, S.F.; Wang, X.; Hullings, M.; Arauz, G.; Bartido, S.; Lewis, J.S.; Schöder, H.; Zanzonico, P.; Scher, H.I.; Sadelain, M.; et al. Chimeric antigen receptor (CAR + ) modified T cells targeting prostate-specific membrane antigen (PSMA) in patients (pts) with castrate metastatic prostate cancer (CMPC). J. Clin. Oncol. 2013, 31, 72. [Google Scholar] [CrossRef]
  81. Kloss, C.C.; Lee, J.; Zhang, A.; Chen, F.; Melenhorst, J.J.; Lacey, S.F.; Maus, M.V.; Fraietta, J.A.; Zhao, Y.; June, C.H. Dominant-Negative TGF-β Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation And Augments Prostate Cancer Eradication. Mol. Ther. 2018, 26, 1855–1866. [Google Scholar] [CrossRef] [Green Version]
  82. Narayan, V.; Barber-Rotenberg, J.; Fraietta, J.; Hwang, W.-T.; Lacey, S.F.; Plesa, G.; Carpenter, E.L.; Maude, S.L.; Lal, P.; Vapiwala, N.; et al. A phase I clinical trial of PSMA-directed/TGFβ-insensitive CAR-T cells in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2021, 39, 125. [Google Scholar] [CrossRef]
  83. Hummel, H.D.; Kufer, P.; Grüllich, C.; Seggewiss-Bernhardt, R.; Deschler-Baier, B.; Chatterjee, M.; Goebeler, M.E.; Miller, K.; De Santis, M.; Loidl, W.; et al. Pasotuxizumab, a BiTE®immune therapy for castration-resistant prostate cancer: Phase I, dose-escalation study findings. Immunotherapy 2021, 13, 125–141. [Google Scholar] [CrossRef] [PubMed]
  84. Hummel, H.-D.; Kufer, P.; Grüllich, C.; Deschler-Baier, B.; Chatterjee, M.; Goebeler, M.-E.; Miller, K.; De Santis, M.; Loidl, W.C.; Buck, A.; et al. Phase 1 study of pasotuxizumab (BAY 2010112), a PSMA-targeting Bispecific T cell Engager (BiTE) immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2019, 37, 5034. [Google Scholar] [CrossRef]
  85. Fizazi, K.; González Mella, P.; Castellano, D.; Minatta, J.N.; Rezazadeh, A.; Shaffer, D.R.; Vazquez Limon, J.C.; Sánchez López, H.M.; Armstrong, A.J.; Horvath, L.; et al. CheckMate 9KD Arm B final analysis: Efficacy and safety of nivolumab plus docetaxel for chemotherapy-naïve metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2021, 39, 12. [Google Scholar] [CrossRef]
  86. Appleman, L.J.; Kolinsky, M.P.; Berry, W.R.; Retz, M.; Mourey, L.; Piulats, J.M.; Romano, E.; Gravis, G.; Gurney, H.; De Bono, J.S.; et al. KEYNOTE-365 cohort B: Pembrolizumab (pembro) plus docetaxel and prednisone in abiraterone (abi) or enzalutamide (enza)–pretreated patients with metastatic castration-resistant prostate cancer (mCRPC)—New data after an additional 1 year of follow-up. J. Clin. Oncol. 2021, 39, 10. [Google Scholar] [CrossRef]
  87. Karzai, F.; Madan, R.A.; Owens, H.; Couvillon, A.; Hankin, A.; Williams, M.; Bilusic, M.; Cordes, L.M.; Trepel, J.B.; Killian, K.; et al. A phase 2 study of olaparib and durvalumab in metastatic castrate-resistant prostate cancer (mCRPC) in an unselected population. J. Clin. Oncol. 2018, 36, 163. [Google Scholar] [CrossRef]
  88. Yu, E.Y.; Piulats, J.M.; Gravis, G.; Laguerre, B.; Arranz Arija, J.A.; Oudard, S.; Fong, P.C.C.; Kolinsky, M.P.; Augustin, M.; Feyerabend, S.; et al. KEYNOTE-365 cohort A updated results: Pembrolizumab (pembro) plus olaparib in docetaxel-pretreated patients (pts) with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, 100. [Google Scholar] [CrossRef]
  89. Long, X.; Hou, H.; Wang, X.; Liu, S.; Diao, T.; Lai, S.; Hu, M.; Zhang, S.; Liu, M.; Zhang, H. Immune signature driven by ADT-induced immune microenvironment remodeling in prostate cancer is correlated with recurrence-free survival and immune infiltration. Cell Death Dis. 2020, 11, 779. [Google Scholar] [CrossRef]
  90. Graff, J.N.; Antonarakis, E.S.; Hoimes, C.J.; Tagawa, S.T.; Hwang, C.; Kilari, D.; Ten Tije, A.; Omlin, A.G.; McDermott, R.S.; Vaishampayan, U.N.; et al. Pembrolizumab (pembro) plus enzalutamide (enza) for enza-resistant metastatic castration-resistant prostate cancer (mCRPC): KEYNOTE-199 cohorts 4-5. J. Clin. Oncol. 2020, 38, 15. [Google Scholar] [CrossRef]
  91. Yu*, E.Y.; Fong, P.; Piulats, J.M.; Appleman, L.; Conter, H.; Feyerabend, S.; Shore, N.; Gravis, G.; Laguerre, B.; Gurney, H.; et al. PD16-12 pembrolizumab plus enzalutamide in abiraterone-pretreated patients with metastatic castration-resistant prostate cancer: Updated results from KEYNOTE-365 cohort C. J. Urol. 2020, 203, e368. [Google Scholar] [CrossRef]
  92. Powles, T.; Fizazi, K.; Gillessen, S.; Drake, C.G.; Rathkopf, D.E.; Narayanan, S.; Green, M.C.; Mecke, A.; Schiff, C.; Sweeney, C. A phase III trial comparing atezolizumab with enzalutamide vs enzalutamide alone in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2017, 35, TPS5090. [Google Scholar] [CrossRef]
  93. Krebs, M.; Solimando, A.G.; Kalogirou, C.; Marquardt, A.; Frank, T.; Sokolakis, I.; Hatzichristodoulou, G.; Kneitz, S.; Bargou, R.; Kübler, H.; et al. miR-221-3p Regulates VEGFR2 Expression in High-Risk Prostate Cancer and Represents an Escape Mechanism from Sunitinib In Vitro. J. Clin. Med. 2020, 9, 670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Sharma, P.; Pachynski, R.K.; Narayan, V.; Fléchon, A.; Gravis, G.; Galsky, M.D.; Mahammedi, H.; Patnaik, A.; Subudhi, S.K.; Ciprotti, M.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  95. Sharma, P.; Pachynski, R.K.; Narayan, V.; Flechon, A.; Gravis, G.; Galsky, M.D.; Mahammedi, H.; Patnaik, A.; Subudhi, S.K.; Ciprotti, M.; et al. 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] [CrossRef]
  96. Boudadi, K.; Suzman, D.L.; Anagnostou, V.; Fu, W.; Luber, B.; Wang, H.; Niknafs, N.; White, J.R.; Silberstein, J.L.; Sullivan, R.; et al. Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018, 9, 28561–28571. [Google Scholar] [CrossRef] [Green Version]
  97. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
  98. Ferris, R.L.; Blumenschein, G.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [Green Version]
  99. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [Green Version]
  100. Sharma, M.; Yang, Z.; Miyamoto, H.; Lucarelli, G. Immunohistochemistry of immune checkpoint markers PD-1 and PD-L1 in prostate cancer. Medicine 2019, 98, e17257. [Google Scholar] [CrossRef]
  101. Gevensleben, H.; Dietrich, D.; Golletz, C.; Steiner, S.; Jung, M.; Thiesler, T.; Majores, M.; Stein, J.; Uhl, B.; Müller, S.; et al. The immune checkpoint regulator PD-L1 is highly expressed in aggressive primary prostate cancer. Clin. Cancer Res. 2016, 22, 1969–1977. [Google Scholar] [CrossRef] [Green Version]
  102. Massari, F.; Ciccarese, C.; Caliò, A.; Munari, E.; Cima, L.; Porcaro, A.B.; Novella, G.; Artibani, W.; Sava, T.; Eccher, A.; et al. Magnitude of PD-1, PD-L1 and T Lymphocyte Expression on Tissue from Castration-Resistant Prostate Adenocarcinoma: An Exploratory Analysis. Target. Oncol. 2016, 11, 345–351. [Google Scholar] [CrossRef]
  103. Antonarakis, E.S.; Piulats, J.M.; Gross-Goupil, M.; Goh, J.; Ojamaa, K.; Hoimes, C.J.; Vaishampayan, U.; Berger, R.; Sezer, A.; Alanko, T.; et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: Multicohort, open-label phase II KEYNOTE-199 study. J. Clin. Oncol. 2020, 38, 395–405. [Google Scholar] [CrossRef] [PubMed]
  104. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wu, Y.M.; Cieślik, M.; Lonigro, R.J.; Vats, P.; Reimers, M.A.; Cao, X.; Ning, Y.; Wang, L.; Kunju, L.P.; de Sarkar, N.; et al. Inactivation of CDK12 Delineates a Distinct Immunogenic Class of Advanced Prostate Cancer. Cell 2018, 173, 1770–1782.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA approval summary: Pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef] [Green Version]
  107. Abida, W.; Cheng, M.L.; Armenia, J.; Middha, S.; Autio, K.A.; Vargas, H.A.; Rathkopf, D.; Morris, M.J.; Danila, D.C.; Slovin, S.F.; et al. Analysis of the Prevalence of Microsatellite Instability in Prostate Cancer and Response to Immune Checkpoint Blockade. JAMA Oncol. 2019, 5, 471–478. [Google Scholar] [CrossRef] [PubMed]
  108. Pritchard, C.C.; Morrissey, C.; Kumar, A.; Zhang, X.; Smith, C.; Coleman, I.; Salipante, S.J.; Milbank, J.; Yu, M.; Grady, W.M.; et al. Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat. Commun. 2014, 5, 4988. [Google Scholar] [CrossRef] [Green Version]
  109. Strickland, K.C.; Howitt, B.E.; Shukla, S.A.; Rodig, S.; Ritterhouse, L.L.; Liu, J.F.; Garber, J.E.; Chowdhury, D.; Wu, C.J.; D’Andrea, A.D.; et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016, 7, 13587–13598. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, F.; Zhao, Q.; Wang, Y.N.; Jin, Y.; He, M.M.; Liu, Z.X.; Xu, R.H. Evaluation of POLE and POLD1 Mutations as Biomarkers for Immunotherapy Outcomes Across Multiple Cancer Types. JAMA Oncol. 2019, 5, 1504–1506. [Google Scholar] [CrossRef] [Green Version]
  111. Antonarakis, E.S. Cyclin-Dependent Kinase 12, Immunity, and Prostate Cancer. N. Engl. J. Med. 2018, 379, 1087–1089. [Google Scholar] [CrossRef]
  112. Antonarakis, E.S.; Isaacsson Velho, P.; Fu, W.; Wang, H.; Agarwal, N.; Santos, V.S.; Maughan, B.L.; Pili, R.; Adra, N.; Sternberg, C.N.; et al. CDK12 -Altered Prostate Cancer: Clinical Features and Therapeutic Outcomes to Standard Systemic Therapies, Poly (ADP-Ribose) Polymerase Inhibitors, and PD-1 Inhibitors. JCO Precis. Oncol. 2020, 4, 370–381. [Google Scholar] [CrossRef] [PubMed]
  113. Schweizer, M.T.; Ha, G.; Gulati, R.; Brown, L.C.; McKay, R.R.; Dorff, T.; Hoge, A.C.H.; Reichel, J.; Vats, P.; Kilari, D.; et al. CDK12 -Mutated Prostate Cancer: Clinical Outcomes With Standard Therapies and Immune Checkpoint Blockade. JCO Precis. Oncol. 2020, 4, 382–392. [Google Scholar] [CrossRef] [PubMed]
  114. Ravindranathan, D.; Russler, G.A.; Yantorni, L.; Drusbosky, L.M.; Bilen, M.A. Detection of Microsatellite Instability via Circulating Tumor DNA and Response to Immunotherapy in Metastatic Castration-Resistant Prostate Cancer: A Case Series. Case Rep. Oncol. 2021, 14, 190–196. [Google Scholar] [CrossRef] [PubMed]
  115. Johnson, L.E.; Olson, B.M.; McNeel, D.G. Pretreatment antigen-specific immunity and regulation—association with subsequent immune response to anti-tumor DNA vaccination. J. Immunother. Cancer 2017, 5, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Komatsu, N.; Matsueda, S.; Tashiro, K.; Ioji, T.; Shichijo, S.; Noguchi, M.; Yamada, A.; Doi, A.; Suekane, S.; Moriya, F.; et al. Gene expression profiles in peripheral blood as a biomarker in cancer patients receiving peptide vaccination. Cancer 2012, 118, 3208–3221. [Google Scholar] [CrossRef] [Green Version]
  117. Jochems, C.; Tucker, J.A.; Tsang, K.Y.; Madan, R.A.; Dahut, W.L.; Liewehr, D.J.; Steinberg, S.M.; Gulley, J.L.; Schlom, J. A combination trial of vaccine plus ipilimumab in metastatic castration-resistant prostate cancer patients: Immune correlates. Cancer Immunol. Immunother. 2014, 63, 407–418. [Google Scholar] [CrossRef]
  118. Santegoets, S.J.A.M.; Stam, A.G.M.; Lougheed, S.M.; Gall, H.; Scholten, P.E.T.; Reijm, M.; Jooss, K.; Sacks, N.; Hege, K.; Lowy, I.; et al. T cell profiling reveals high CD4+CTLA-4+ T cell frequency as dominant predictor for survival after Prostate GVAX/ipilimumab treatment. Cancer Immunol. Immunother. 2013, 62, 245–256. [Google Scholar] [CrossRef] [PubMed]
  119. Kongsted, P.; Borch, T.H.; Ellebaek, E.; Iversen, T.Z.; Andersen, R.; Met, Ö.; Hansen, M.; Lindberg, H.; Sengeløv, L.; Svane, I.M. Dendritic cell vaccination in combination with docetaxel for patients with metastatic castration-resistant prostate cancer: A randomized phase II study. Cytotherapy 2017, 19, 500–513. [Google Scholar] [CrossRef]
  120. Ross, A.; Armstrong, A.J.; Pieczonka, C.M.; Bailen, J.L.; Tutrone, R.F.; Cooperberg, M.R.; Pavlovich, C.P.; Renzulli, J.F.; Haynes, H.; Sheikh, N.A.; et al. A comparison of sipuleucel-T (sip-T) product parameters from two phase III studies: PROVENT in active surveillance prostate cancer and IMPACT in metastatic castrate-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, 321. [Google Scholar] [CrossRef]
  121. Madan, R.A.; Slovin, S.; Harshman, L.C.; Wei, X.X.; Bilusic, M.; Karzai, F.H.; Donahue, R.N.; Toney, N.J.; Strauss, J.; Cordes, L.; et al. 681P Clinical and immune responses to immunotherapy in biochemically recurrent (non-metastatic castration sensitive) prostate cancer (BCRpc). Ann. Oncol. 2020, 31, S542. [Google Scholar] [CrossRef]
  122. Tran, B.; Horvath, L.; Dorff, T.B.; Greil, R.; Machiels, J.-P.H.; Roncolato, F.; Autio, K.A.; Rettig, M.; Fizazi, K.; Lolkema, M.P.; et al. Phase I study of AMG 160, a half-life extended bispecific T-cell engager (HLE BiTE) immune therapy targeting prostate-specific membrane antigen (PSMA), in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, TPS261. [Google Scholar] [CrossRef]
  123. Kelly, W.K.; Pook, D.W.; Appleman, L.J.; Waterhouse, D.M.; Horvath, L.; Edenfield, W.J.; Matsubara, N.; Danila, D.C.; Aggarwal, R.R.; Petrylak, D.P.; et al. Phase I study of AMG 509, a STEAP1 x CD3 T-cell recruiting XmAb 2+1 immune therapy, in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2021, 39, TPS183. [Google Scholar] [CrossRef]
  124. Markowski, M.C.; Kilari, D.; Eisenberger, M.A.; McKay, R.R.; Dreicer, R.; Trikha, M.; Heath, E.I.; Li, J.; Garzone, P.D.; Young, T.S. Phase I study of CCW702, a bispecific small molecule-antibody conjugate targeting PSMA and CD3 in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2021, 39, TPS5094. [Google Scholar] [CrossRef]
  125. Chang Lee, S.; Ma, J.S.Y.; Kim, M.S.; Laborda, E.; Choi, S.H.; Hampton, E.N.; Yun, H.; Nunez, V.; Muldong, M.T.; Wu, C.N.; et al. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. Sci. Adv. 2021, 7, eabi8193. [Google Scholar] [CrossRef]
  126. Aggarwal, R.R.; Aparicio, A.; Heidenreich, A.; Sandhu, S.K.; Zhang, Y.; Salvati, M.; Shetty, A.; Hashemi Sadraei, N. Phase 1b study of AMG 757, a half-life extended bispecific T-cell engager (HLE BiTEimmune-oncology therapy) targeting DLL3, in de novo or treatment emergent neuroendocrine prostate cancer (NEPC). J. Clin. Oncol. 2021, 39, TPS5100. [Google Scholar] [CrossRef]
  127. Tsimberidou, A.M.; Drakaki, A.; Khalil, D.; Kummar, S.; Hodi, F.S.; Oh, D.Y.; Cabanski, C.R.; Tezlaff, M.; LaVallee, T.; Spasic, M.; et al. An exploratory study of nivolumab (nivo) with or without ipilimumab (ipi) according to the percentage of tumoral CD8 cells in advanced metastatic cancer. J. Clin. Oncol. 2021, 39, 2573. [Google Scholar] [CrossRef]
  128. Reimers, M.A.; Abida, W.; Chou, J.; George, D.J.; Heath, E.I.; McKay, R.R.; Pachynski, R.K.; Zhang, J.; Choi, J.E.; Feng, F.Y.; et al. IMPACT: Immunotherapy in patients with metastatic cancers and CDK12 mutations. J. Clin. Oncol. 2019, 37, TPS5091. [Google Scholar] [CrossRef]
  129. Wong, Y.N.S.; Sankey, P.; Josephs, D.H.; Jones, R.J.; Crabb, S.J.; Beare, S.; Duggan, M.; White, L.; Charlaftis, N.; Wheeler, G.; et al. Nivolumab and ipilimumab treatment in prostate cancer with an immunogenic signature (NEPTUNES). J. Clin. Oncol. 2019, 37, TPS5090. [Google Scholar] [CrossRef]
  130. Mehra, N.; Kloots, I.; Slootbeek, P.; den Brok, M.; Adema, G.; Kerkmeijer, L.; Smeenk, R.J.; Westdorp, H.; Bloemendal, H.; Schalken, J.; et al. 642TiP Phase II CA184-585 (INSPIRE) trial of ipilimumab with nivolumab for molecular-selected patients with castration-resistant prostate cancer. Ann. Oncol. 2021, 32, S671. [Google Scholar] [CrossRef]
  131. Bansal, D.; Beck, R.; Arora, V.; Knoche, E.M.; Picus, J.; Reimers, M.A.; Roth, B.J.; Gulley, J.L.; Schreiber, R.; Pachynski, R.K. A pilot trial of neoantigen DNA vaccine in combination with nivolumab/ipilimumab and prostvac in metastatic hormone-sensitive prostate cancer (mHSPC). J. Clin. Oncol. 2021, 39, TPS192. [Google Scholar] [CrossRef]
  132. Redman, J.M.; Steinberg, S.M.; Gulley, J.L. Quick efficacy seeking trial (QuEST1): A novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer 11 Medical and Health Sciences 1107 Immunology. J. Immunother. Cancer 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Autio, K.A.; Eastham, J.A.; Danila, D.C.; Slovin, S.F.; Morris, M.J.; Abida, W.; Laudone, V.P.; Touijer, K.A.; Gopalan, A.; Wong, P.; et al. A phase II study combining ipilimumab and degarelix with or without radical prostatectomy (RP) in men with newly diagnosed metastatic noncastration prostate cancer (mNCPC) or biochemically recurrent (BR) NCPC. J. Clin. Oncol. 2017, 35, 203. [Google Scholar] [CrossRef]
  134. Gratzke, C.; Burgents, J.E.; Niu, C.; Poehlein, C.H.; Drake, C.G. Phase III study of pembrolizumab (pembro) plus enzalutamide (enza) and androgen deprivation therapy (ADT) for patients (pts) with metastatic hormone-sensitive prostate cancer (mHSPC): KEYNOTE-991. J. Clin. Oncol. 2020, 38, TPS5595. [Google Scholar] [CrossRef]
  135. Fizazi, K.; González Mella, P.; Castellano, D.; Minatta, J.N.; Rezazadeh Kalebasty, A.; Shaffer, D.; Vázquez Limón, J.C.; Sánchez López, H.M.; Armstrong, A.J.; Horvath, L.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  136. Drake, C.G.; Saad, F.; Clark, W.R.; Ciprotti, M.; Sharkey, B.; Subudhi, S.K.; Fizazi, K. 690TiP A phase III, randomized, double-blind trial of nivolumab or placebo combined with docetaxel for metastatic castration-resistant prostate cancer (mCRPC.; CheckMate 7DX). Ann. Oncol. 2020, 31, S546. [Google Scholar] [CrossRef]
  137. Graff, J.N.; Liang, L.W.; Kim, J.; Stenzl, A. KEYNOTE-641: A Phase III study of pembrolizumab plus enzalutamide for metastatic castration-resistant prostate cancer. Futur. Oncol. 2021, 17, 3017–3026. [Google Scholar] [CrossRef] [PubMed]
  138. Piulats, J.; Ferrario, C.; Linch, M.; Stoeckle, M.; Laguerre, B.; Arranz, J.; Todenhoefer, T.; Fong, P.; Berry, W.; Emmenegger, U.; et al. 351 KEYNOTE-365 cohort D: Pembrolizumab plus abiraterone acetate and prednisone in patients with chemotherapy-naive metastatic castration-resistant prostate cancer (mCRPC). J. Immunother. Cancer 2021, 9, A378. [Google Scholar] [CrossRef]
  139. Berry, W.R.; Fong, P.C.C.; Piulats, J.M.; Appleman, L.J.; Conter, H.J.; Feyerabend, S.; Shore, N.D.; Gravis, G.; Laguerre, B.; Gurney, H.; et al. KEYNOTE-365 cohort C updated results: Pembrolizumab (pembro) plus enzalutamide (enza) in abiraterone (abi)-pretreated patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, 102. [Google Scholar] [CrossRef]
  140. Kolinsky, M.P.; Gravis, G.; Mourey, L.; Piulats, J.M.; Sridhar, S.S.; Romano, E.; Berry, W.R.; Gurney, H.; Retz, M.; Appleman, L.J.; et al. KEYNOTE-365 cohort B updated results: Pembrolizumab (pembro) plus docetaxel and prednisone in abiraterone (abi) or enzalutamide (enza)-pretreated patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, 103. [Google Scholar] [CrossRef]
  141. Danila, D.C.; Kuzel, T.; Cetnar, J.P.; Rathkopf, D.E.; Morris, M.J.; Alumkal, J.J.; Butler, A.; Curley, T.; Hullings, M.; Buddle, J.R.; et al. A phase 1/2 study combining ipilimumab with abiraterone acetate plus prednisone in chemotherapy- and immunotherapy-naïve patients with progressive metastatic castration resistant prostate cancer (mCRPC). J. Clin. Oncol. 2016, 34, e16507. [Google Scholar] [CrossRef]
  142. Gandhy, S.U.; Karzai, F.; Marte, J.L.; Bilusic, M.; McMahon, S.; Strauss, J.; Couvillon, A.; Williams, M.; Hankin, A.; Steinberg, S.M.; et al. PSA progression compared to radiographic or clinical progression in metastatic castration-resistant prostate cancer patients treated with enzalutamide. J. Clin. Oncol. 2020, 38, 105. [Google Scholar] [CrossRef]
  143. Gandhy, S.; Gonzales, E.M.; Karzai, F.; Marte, J.; Bilusic, M.; McMahon, S.; Strauss, J.; Steinberg, S.; Gill, A.; Tubbs, A.; et al. 643P Evaluating biomarkers in metastatic castration resistant prostate cancer (mCRPC) patients (Pts) treated with enzalutamide (Enza): PSA, circulating tumor cell (CTC) counts, AR-V7 status, PET imaging vs. CT & Tc99 scans. Ann. Oncol. 2020, 31, S527–S528. [Google Scholar] [CrossRef]
  144. Petrylak, D.P.; Ratta, R.; Gafanov, R.; Facchini, G.; Piulats, J.M.; Kramer, G.; Flaig, T.W.; Chandana, S.R.; Li, B.; Burgents, J.; et al. KEYNOTE-921: Phase III study of pembrolizumab plus docetaxel for metastatic castration-resistant prostate cancer. Futur. Oncol. 2021, 17, 3291–3299. [Google Scholar] [CrossRef] [PubMed]
  145. Agarwal, N.; Azad, A.; Carles, J.; Chowdhury, S.; McGregor, B.A.; Merseburger, A.S.; Oudard, S.; Saad, F.; Soares, A.; Panneerselvam, A.; et al. A phase III, randomized, open-label, study (CONTACT-02) of cabozantinib plus atezolizumab versus second novel hormone therapy (NHT) in patients (pts) with metastatic, castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2021, 39, TPS190. [Google Scholar] [CrossRef]
  146. Atiq, M.O.; Gandhy, S.; Karzai, F.; Bilusic, M.; Cordes, L.M.; Owens, H.; Couvillon, A.; Hankin, A.; Williams, M.; Figg, W.D.; et al. Patients with undetectable PSA 2 years after docetaxel for metastatic castration sensitive prostate cancer (mCSPC). J. Clin. Oncol. 2021, 39, e17044. [Google Scholar] [CrossRef]
  147. Chandran, E.B.A.; Atiq, M.O.; Donahue, R.N.; Karzai, F.; Bilusic, M.; Marte, J.L.; Arlen, P.M.; Cordes, L.M.; Owens, H.; Hankin, A.; et al. Evaluating the optimal sequence of immunotherapy and docetaxel in men with metastatic castration-sensitive prostate cancer. J. Clin. Oncol. 2022, 40, 130. [Google Scholar] [CrossRef]
  148. Sokolova, A.; Gulati, R.; Cheng, H.H.; Beer, T.M.; Graff, J.N.; Amador, M.; Toulouse, A.; Taylor, K.; Bailey, S.; Smith, S.; et al. Trial in progress: Durvalumab and olaparib for the treatment of prostate cancer in men predicted to have a high neoantigen load. J. Clin. Oncol. 2022, 40, TPS202. [Google Scholar] [CrossRef]
  149. Pachynski, R.K.; Retz, M.; Goh, J.C.; Burotto, M.; Gravis, G.; Castellano, D.; Flechon, A.; Zschaebitz, S.; Shaffer, D.R.; Vazquez Limon, J.C.; et al. CheckMate 9KD cohort A1 final analysis: Nivolumab (NIVO) + rucaparib for post-chemotherapy (CT) metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2021, 39, 5044. [Google Scholar] [CrossRef]
  150. Petrylak, D.P.; Perez-Gracia, J.L.; Lacombe, L.; Bastos, D.A.; Mahammedi, H.; Kwan, E.M.; Zschäbitz, S.; Armstrong, A.J.; Pachynski, R.K.; Goh, J.C.; et al. 579MO CheckMate 9KD cohort A2 final analysis: Nivolumab (NIVO) + rucaparib for chemotherapy (CT)-naïve metastatic castration-resistant prostate cancer (mCRPC). Ann. Oncol. 2021, 32, S629–S630. [Google Scholar] [CrossRef]
  151. Subudhi, S.K.; Aparicio, A.; Zurita, A.J.; Doger, B.; Kelly, W.K.; Peer, A.; Rathkopf, D.E.; Karsh, L.I.; Tryon, J.J.; Kothari, N.; et al. A phase Ib/II study of niraparib combination therapies for the treatment of metastatic castration-resistant prostate cancer (NCT03431350). J. Clin. Oncol. 2019, 37, TPS5087. [Google Scholar] [CrossRef]
  152. Yu, E.Y.; Park, S.H.; Huang, Y.-H.; Bennamoun, M.; Xu, L.; Kim, J.; Antonarakis, E.S. Phase III study of pembrolizumab (pembro) plus olaparib versus enzalutamide (enza) or abiraterone acetate (abi) in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) who progressed on chemotherapy: KEYLYNK-010. J. Clin. Oncol. 2020, 38, TPS256. [Google Scholar] [CrossRef]
  153. Supiot, S.; Libois, V.; Guimas, V.; Rio, E.; Rolland, F.; Bompas, E.; Vansteene, D.; Tigreat, M.; Lisbona, A.; Colliaux, J.; et al. Prostate cancer with oligometastatic relapse: Combining stereotactic ablative radiotherapy and durvalumab, a randomized phase II trial (POSTCARD—GETUG-P13). J. Clin. Oncol. 2019, 37, TPS5088. [Google Scholar] [CrossRef]
  154. Aggarwal, R.R.; Luch Sam, S.; Koshkin, V.S.; Small, E.J.; Feng, F.Y.; de Kouchkovsky, I.; Kwon, D.H.; Friedlander, T.W.; Borno, H.; Bose, R.; et al. Immunogenic priming with 177 Lu-PSMA-617 plus pembrolizumab in metastatic castration resistant prostate cancer (mCRPC): A phase 1b study. J. Clin. Oncol. 2021, 39, 5053. [Google Scholar] [CrossRef]
  155. Sandhu, S.K.; Joshua, A.M.; Emmett, L.; Spain, L.; Horvath, L.G.; Crumbaker, M.; Anton, A.; Wallace, R.; Pasam, A.; Bressel, M.; et al. 577O PRINCE: Interim analysis of the phase Ib study of 177Lu-PSMA-617 in combination with pembrolizumab for metastatic castration resistant prostate cancer (mCRPC). Ann. Oncol. 2021, 32, S626. [Google Scholar] [CrossRef]
  156. Dohopolski, M.; Watumull, L.; Mathews, D.; Gao, A.; Garant, A.; Choy, H.; Ahn, C.; Timmerman, R.D.; Courtney, K.; Hannan, R. Phase II Trial of Sipuleucel-T and Stereotactic Ablative Radiation therapy (SAbR) for Patients with Metastatic Castrate-Resistant Prostate Cancer (mCRPC). Int. J. Radiat. Oncol. 2020, 108, e879. [Google Scholar] [CrossRef]
  157. Reimers, M.A.; Visconti, J.L.; Cittolin Santos, G.F.; Pachynski, R.K. A phase 1b clinical trial of cabozantinib (CABO) and abiraterone (ABI) with checkpoint inhibitor immunotherapy (CPI) in metastatic hormone-sensitive prostate cancer (mHSPC) (CABIOS Trial). J. Clin. Oncol. 2022, 40, TPS214. [Google Scholar] [CrossRef]
  158. Yuan, Z.; Fernandez, D.; Dhillon, J.; Abraham-Miranda, J.; Awasthi, S.; Kim, Y.; Zhang, J.; Jain, R.; Serna, A.; Pow-Sang, J.M.; et al. Proof-of-principle Phase I results of combining nivolumab with brachytherapy and external beam radiation therapy for Grade Group 5 prostate cancer: Safety, feasibility, and exploratory analysis. Prostate Cancer Prostatic Dis. 2021, 24, 140–149. [Google Scholar] [CrossRef]
  159. Tagawa, S.T.; Osborne, J.; Dallos, M.; Nauseef, J.; Sternberg, C.N.; Gregos, P.; Patel, A.; Tan, A.; Singh, S.; Bissassar, M.; et al. Phase I/II trial of pembrolizumab and AR signaling inhibitor +/- 225Ac-J591 for chemo-naive metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2022, 40, TPS216. [Google Scholar] [CrossRef]
Figure 1. The tumour immune microenvironment of prostate cancer. The tumour microenvironment is composed of stroma, tumour cells and a variety of immune cells. The stromal components and tumour cells interact and promote a hypoxic and pro-tumour environment through cytokine production and pro-inflammatory signalling. As the immune cells infiltrate into the tumour microenvironment, immunosuppressive cells, such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) and M2 macrophages, suppress the anti-tumour activity of dendritic cells (DCs), cytotoxic T cells, natural killer (NK) and B cells, which together promote an immunosuppressive environment.
Figure 1. The tumour immune microenvironment of prostate cancer. The tumour microenvironment is composed of stroma, tumour cells and a variety of immune cells. The stromal components and tumour cells interact and promote a hypoxic and pro-tumour environment through cytokine production and pro-inflammatory signalling. As the immune cells infiltrate into the tumour microenvironment, immunosuppressive cells, such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) and M2 macrophages, suppress the anti-tumour activity of dendritic cells (DCs), cytotoxic T cells, natural killer (NK) and B cells, which together promote an immunosuppressive environment.
Uro 02 00010 g001
Figure 2. Immunotherapy Strategies in Prostate Cancer. Immunotherapy aims to enhance the adaptive immune response by enhancing specificity or promoting stronger activation against the tumour. (A) Cancer vaccines involve vaccination of the patient with tumour peptides, allogeneic whole tumour cells or autologous DCs as vehicles for delivery of the tumour antigen, priming the patient’s immune system to recognize the tumour-associated antigens. (B) Chimeric Antigen Receptor T Cells (CAR-T) are genetically modified T cells which express a patient antigen-specific chimeric receptor combining both antibody specificity and T-cell effector and regulatory functions. (C) Bispecific antibodies (BiTEs) are designed to target the CD3 protein through an effector arm and a tumour antigen via a target arm, which promotes the interaction between tumour cells and CD8+ T cells, resulting in tumour cell death.
Figure 2. Immunotherapy Strategies in Prostate Cancer. Immunotherapy aims to enhance the adaptive immune response by enhancing specificity or promoting stronger activation against the tumour. (A) Cancer vaccines involve vaccination of the patient with tumour peptides, allogeneic whole tumour cells or autologous DCs as vehicles for delivery of the tumour antigen, priming the patient’s immune system to recognize the tumour-associated antigens. (B) Chimeric Antigen Receptor T Cells (CAR-T) are genetically modified T cells which express a patient antigen-specific chimeric receptor combining both antibody specificity and T-cell effector and regulatory functions. (C) Bispecific antibodies (BiTEs) are designed to target the CD3 protein through an effector arm and a tumour antigen via a target arm, which promotes the interaction between tumour cells and CD8+ T cells, resulting in tumour cell death.
Uro 02 00010 g002
Figure 3. The PD-1 and PD-L1 interaction and its inhibition by immunotherapy. (A) PD-1 is expressed on cytotoxic T cells and PD-L1 is expressed on tumour cells. The T-cell receptor (TCR) binds to the tumour antigen (Ag) presented on the tumour cell surface by major histocompatibility complex class I (MHC I). When PD-L1 binds to PD-1, the activity of the cytotoxic T cell will be suppressed and the interaction between TCR and MHC I is blocked. The subsequent immune checkpoint activation can cause apoptosis of cytotoxic T cells. (B) Inhibition of PD-1 and PD-L1 activation by anti-PD-1/PD-L1 immunotherapy. Immune checkpoint inhibitors are antibodies that can specifically bind to the immune checkpoint molecules, such as PD-1 and its ligand PD-L1. This binding blocks the interaction between PD-1 and PD-L1 and promotes the anti-tumour activity of the cytotoxic T cell.
Figure 3. The PD-1 and PD-L1 interaction and its inhibition by immunotherapy. (A) PD-1 is expressed on cytotoxic T cells and PD-L1 is expressed on tumour cells. The T-cell receptor (TCR) binds to the tumour antigen (Ag) presented on the tumour cell surface by major histocompatibility complex class I (MHC I). When PD-L1 binds to PD-1, the activity of the cytotoxic T cell will be suppressed and the interaction between TCR and MHC I is blocked. The subsequent immune checkpoint activation can cause apoptosis of cytotoxic T cells. (B) Inhibition of PD-1 and PD-L1 activation by anti-PD-1/PD-L1 immunotherapy. Immune checkpoint inhibitors are antibodies that can specifically bind to the immune checkpoint molecules, such as PD-1 and its ligand PD-L1. This binding blocks the interaction between PD-1 and PD-L1 and promotes the anti-tumour activity of the cytotoxic T cell.
Uro 02 00010 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, Y.; Campbell, B.K.; Stylli, S.S.; Corcoran, N.M.; Hovens, C.M. The Prostate Cancer Immune Microenvironment, Biomarkers and Therapeutic Intervention. Uro 2022, 2, 74-92. https://doi.org/10.3390/uro2020010

AMA Style

Zhang Y, Campbell BK, Stylli SS, Corcoran NM, Hovens CM. The Prostate Cancer Immune Microenvironment, Biomarkers and Therapeutic Intervention. Uro. 2022; 2(2):74-92. https://doi.org/10.3390/uro2020010

Chicago/Turabian Style

Zhang, Yangyi, Bethany K. Campbell, Stanley S. Stylli, Niall M. Corcoran, and Christopher M. Hovens. 2022. "The Prostate Cancer Immune Microenvironment, Biomarkers and Therapeutic Intervention" Uro 2, no. 2: 74-92. https://doi.org/10.3390/uro2020010

Article Metrics

Back to TopTop