Next Article in Journal
RRx-001 Increases Erythrocyte Preferential Adhesion to the Tumor Vasculature
Next Article in Special Issue
Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
Previous Article in Journal
Development of an Antigen-Antibody Co-Display System for Detecting Interaction of G-Protein-Coupled Receptors and Single-Chain Variable Fragments
Previous Article in Special Issue
Hyperprogressive Disease: Main Features and Key Controversies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoint Inhibitors: A Promising Treatment Option for Metastatic Castration-Resistant Prostate Cancer?

by
Vicenç Ruiz de Porras
1,2,*,†,
Juan Carlos Pardo
2,3,†,
Lucia Notario
2,3,
Olatz Etxaniz
2,3 and
Albert Font
2,3,*
1
Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
2
Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
3
Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(9), 4712; https://doi.org/10.3390/ijms22094712
Submission received: 16 April 2021 / Revised: 27 April 2021 / Accepted: 28 April 2021 / Published: 29 April 2021
(This article belongs to the Special Issue Immune Responses in Cancer Immunology and Immunotherapy)

Abstract

:
Since 2010, several treatment options have been available for men with metastatic castration-resistant prostate cancer (mCRPC), including immunotherapeutic agents, although the clinical benefit of these agents remains inconclusive in unselected mCRPC patients. In recent years, however, immunotherapy has re-emerged as a promising therapeutic option to stimulate antitumor immunity, particularly with the use of immune checkpoint inhibitors (ICIs), such as PD-1/PD-L1 and CTLA-4 inhibitors. There is increasing evidence that ICIs may be especially beneficial in specific subgroups of patients with high PD-L1 tumor expression, high tumor mutational burden, or tumors with high microsatellite instability/mismatch repair deficiency. If we are to improve the efficacy of ICIs, it is crucial to have a better understanding of the mechanisms of resistance to ICIs and to identify predictive biomarkers to determine which patients are most likely to benefit. This review focuses on the current status of ICIs for the treatment of mCRPC (either as monotherapy or in combination with other drugs), mechanisms of resistance, potential predictive biomarkers, and future challenges in the management of mCRPC.

1. Current Treatment Options for Metastatic Castration-Resistant Prostate Cancer (mCRPC)

Prostate cancer (PC) is the most common malignancy in men and a major cause of cancer death [1]. For patients with PC who experience disease relapse after local therapy or for those with metastatic disease, androgen deprivation therapy (ADT) is the backbone of systemic therapy. However, despite significant initial responses to ADT, almost all metastatic patients progress to an incurable metastatic castration-resistant prostate cancer (mCRPC), defined as radiographic progression and/or a rise in prostate-specific antigen (PSA) despite having a castrate level of testosterone [2,3]. Sadly, mCRPC has a poor prognosis with a median overall survival (OS) of approximately 3 years, making it a significant therapeutic challenge. Importantly, the majority of castration-resistant prostate tumors retain their dependence on the androgen receptor (AR), mainly through genomic amplification or gain-of-function mutations of the AR gene, AR mRNA splice variants, or alterations in androgen synthesis [4]. Thus, potent next-generation AR pathway inhibitors (ARPIs), such as abiraterone, an irreversible inhibitor of CYP17A1, and enzalutamide, an AR antagonist, are also commonly used in asymptomatic and mildly symptomatic mCRPC patients [5,6,7,8]. The AR antagonists enzalutamide, apalutamide and darolutamide have also demonstrated improved outcomes in men with non-metastatic CRPC [9,10,11].
Additionally, docetaxel, the standard first-line chemotherapeutic treatment for symptomatic mCRPC patients, has shown a benefit in terms of pain relief, serum PSA levels, quality of life (QoL) and OS [12,13,14]. More recently, docetaxel was also evaluated in high-volume disease metastatic castration-sensitive prostate cancer patients in combination with ADT and positive results were reported in two different clinical trials [15,16]. Nevertheless, as is often the case with chemotherapy, the efficacy of docetaxel is limited by the development of tumor resistance. However, since 2010, men who are resistant to docetaxel-based chemotherapy can receive cabazitaxel, a second-generation taxane, which has shown activity in docetaxel-resistant cell lines [17] and demonstrated clinical efficacy in the pivotal phase III TROPIC [18] and CARD [19] trials.
Unfortunately, very few therapeutic options are available to patients progressing to taxanes, although platinum-based treatments have demonstrated a limited benefit in some patients. Modest response rates have been observed with single-drug platinum-based regimens, but more encouraging results have been obtained with platinum/taxane combinations [20,21]. Of note, Corn and colleagues suggested that cabazitaxel plus carboplatin is well tolerated and confers superior progression-free survival (PFS) and response rate (RR) than cabazitaxel alone in docetaxel-resistant mCRPC patients [22]. Interestingly, patients with aggressive variant prostate cancer (AVPC) [23] were found to benefit most from this combination [22]. AVPC is less dependent on AR signaling and harbors defects in tumor suppressor genes such as TP53, RB1, and PTEN [24,25]. In this context, our group recently demonstrated that the CXCR2/BCL-2 antiapoptotic axis is markedly reduced in taxane-exposed mCRPC tumors, representing an emerging vulnerability to genotoxic platinum-based treatments. Taken together, these findings indicate that taxane-resistant tumors are sensitized to platinum treatment, and taxane/platinum combinations have antitumor efficacy in aggressive mCRPC [26].
Alterations in DNA damage repair (DDR) genes, such as those involving the homologous recombination (HR) pathway genes, such as BRCA2, BRCA1, ATM, CDK12, and PALB2, occur in approximately 20–25% of mCRPC cases at either the somatic or the germline level [27,28]. In this regard, two Poly (ADP-ribose) polymerase inhibitors (PARPi), rucaparib and olaparib, have been recently approved by the FDA for the treatment of mCRPC [29]. While both agents are approved for tumors with BRCA1/2 alterations, the approval for olaparib also includes patients with 12 other HR gene alterations, including ATM and PALB2 [30,31]. In fact, the association between alterations in DDR genes and positive response to PARPi is the first example of precision medicine in mCRPC.
In the last years, immunotherapy—particularly immune checkpoint inhibitors (ICIs)—is re-emerging as a viable option for PC, and especially for mCRPC, to stimulate anti-tumor immunity [32,33,34]. Here we review and discuss the rationale, translational background, and clinical trials of ICIs, either as monotherapy or in combination with other drugs currently used in mCRPC.

2. ICIs in mCRPC: An Overview

A growing body of evidence has demonstrated that tumors can suppress the antitumor immune response by several mechanisms, including cytokine release, recruitment of immunosuppressive cells, and upregulation of co-inhibitory receptors, known as immune checkpoints [35]. Although immunotherapy has emerged as a promising treatment option for many solid tumors, such as melanoma [36,37], non-small-cell lung cancer (NSCLC) [38], renal cell carcinoma [39], urothelial cancer [40], and head and neck cancer [41], progress in PC has been relatively modest, even though PC was one of the first diseases where an immunotherapeutic agent, sipuleucel-T, was approved. In fact, sipuleucel-T, an autologous antigen presenting cell (APC)-based immunotherapy, has shown a benefit in OS, but not in PFS, in clinical trials [42,43,44] and is currently the only FDA-approved immunotherapy option for mCRPC.
ICIs are monoclonal antibodies (mAbs) that can inhibit immune checkpoint receptors and therefore prevent the inactivation of T-cell function. The clinically most important immune checkpoint receptors include programmed death 1 (PD-1), programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) [32]. Interestingly, mismatch repair deficiency (dMMR) or microsatellite instability-high (MSI-H) tumors are sensitive to standard and novel hormonal therapies and to PD-1 inhibitors [45,46]. In a study by Abida et al., 32 of 1033 (3%) screened CRPC patients had dMMR. Eleven patients were treated with anti-PD-1/PD-L1 therapy; six had a PSA response >50% and four had a radiographic response. Long-lasting disease control was observed in five of six responders [47]. Genomic events leading to dMMR and MSI-H mostly affect the MSH2 and MSH6 genes [46,48] and are associated with more aggressive clinical and pathological features and a higher tumor mutational burden (TMB) [49]. Since a high TMB is commonly associated with better clinical outcome to immunotherapy [50,51,52,53], it is important to take into account that PC generally has a lower TMB. Only 3–8.3% of advanced PC tumors have a high TMB, which represents a significant obstacle for immunotherapy efficacy in these patients [54,55]. Here we summarize and discuss the clinical trials evaluating the blockade of the PD-1/PD-L1 and CTLA-4 pathways in mCRPC.

3. PD-1/PD-L1 Inhibitors in mCRPC

PD-1 is a transmembrane protein expressed on T cells that interacts with PD-L1, which is expressed on both normal and cancer cells. This interaction is an important checkpoint for CD8+ T-cell inhibition. PD-1 binding to PD-L1 on tumor cells results in an inhibition of apoptosis, T-lymphocyte tolerance, and an increase in tumor cell survival [56]. Currently, ICIs that act as inhibitors of PD-1 include nivolumab and pembrolizumab, whereas anti-PD-L1 mAbs include atezolizumab, avelumab, and durvalumab [56]. Although antibody-mediated blockade of the PD-1/PD-L1 interaction is an effective clinical strategy in several tumor types, data from trials in mCRPC patients suggest that PD-1/PD-L1 blockade is less effective in this disease, with no observable objective responses after nivolumab treatment [57]. In the same line, in a phase Ia study with atezolizumab in 35 mCRPC patients who progressed to sipuleucel-T or enzalutamide, PSA response was only 8.6%, only one patient had an objective partial response, and median OS was 14.7 months [58]. In the KEYNOTE-199 trial, mCRPC patients previously treated with docetaxel and at least one ARPI were treated with pembrolizumab. Three cohorts of patients were included: PD-L1-positive tumors (cohort 1: 133 patients), PD-L1-negative tumors (cohort 2: 66 patients) and bone-predominant disease (cohort 3: 59 patients). The activity of pembrolizumab was poor in all three cohorts, with <10% PSA response and <5% overall response rate (ORR), while radiographic PFS (rPFS) was 2.1, 2.1, and 3.7 months, respectively, in the three cohorts [59].
The value of PD-L1 expression as a prognostic and predictive biomarker of response to ICIs is currently a question for debate due to its variable predictive value among different solid tumor types. Several studies have suggested that the relatively low expression levels of PD-L1 in mCRPC tumors can represent an important obstacle for anti-PD-1/PD-L1 efficacy as monotherapy [60,61]. On that basis, the KEYNOTE-028 trial of pembrolizumab in advanced prostate adenocarcinoma only included pre-treated patients with PD-L1 expression in >1% of tumor or stroma cells. Results showed an ORR of 17.4%, with 34.8% of patients exhibiting stable disease and a median duration of response of 13.5 months after pembrolizumab administration [62].
Interestingly, progression to enzalutamide in mCRPC has been associated with an upregulation of tumor PD-L1, suggesting that PD-L1 might be a dynamic biomarker that is involved not only in immune evasion but also in resistance to enzalutamide [63]. Based on this hypothesis, a promising therapeutic strategy could be the combination of an anti-PD-L1 with enzalutamide. In fact, in a recent study, 28 CRPC patients who had progressed to enzalutamide were treated with pembrolizumab plus enzalutamide. A significant PSA response rate of >50% was observed in 5 (18%) patients; 3 of 12 patients (25%) had a partial response; OS was 21.9 months. Importantly, in two cases, response was maintained after 37 and 39 months [64]. Along the same lines, in cohort C of the KEYNOTE-365 trial, 102 mCRPC patients who had progressed on abiraterone were treated with pembrolizumab plus enzalutamide. PSA response was 22%, ORR was 12%, rPFS was 6.1 months, and OS was 20.4 months [65]. Based on these results, a randomized phase III trial of pembrolizumab plus enzalutamide versus enzalutamide alone is currently ongoing (KEYNOTE-641; NCT03834493). In contrast, in the IMbassador250 phase III trial, the combination of atezolizumab plus enzalutamide did not demonstrate an improvement in OS compared to enzalutamide alone [66].
Taken together, these results suggest that anti-PD1/PD-L1 has a limited activity in mCRPC as monotherapy and combinatory approaches warrant investigation. Several ongoing trials are exploring the promising strategy of PD-1/PD-L1 blockade in combination with other immunotherapies, including vaccines (NCT02933255, NCT03600350, NCT02499835 [67]), kinase inhibitors like ipatasertib (NCT03673787 [68]), ARPIs (NCT02312557, NCT03016312), radium-223 (NCT03093428, NCT02814669 [69]), sipuleucel-T (NCT03024216 [70]), PARPi (NCT03572478; NCT03330405 [71]), navarixin (a CXCR1/CXCR2 antagonist; NCT03473925), and other ICIs, such as the CTLA-4 checkpoint inhibitor ipilimumab (Table 1 and Table 2).

4. CTLA-4 Inhibitors in mCRPC

T lymphocytes can express two related transmembrane receptors, CD28 and CTLA-4, on their cell surface, and both bind to the same ligands, CD80 and CD86, on APC. Whereas ligand engagement of CD28 activates T cells, interaction between CTLA-4 and these ligands inhibits T-cell stimulation, thereby promoting a negative feedback loop that prevents T cells from killing cancer cells [56]. Blockade of CTLA-4 is thus an important mechanism for increasing T-cell immunity and, potentially, T-cell antitumor responses. Ipilimumab, a first-in-class fully human mAb that binds to CTLA-4 [81], has been widely studied in several phase I, II, and III clinical trials for mCRPC patients, both as monotherapy and in combination therapy. An initial phase I trial demonstrated that a single dose of ipilimumab given to mCRPC patients was safe and led to a PSA decline of >50% in 14% of patients without inducing significant clinical autoimmunity [82]. On the basis of these results, two phase II trials evaluated the efficacy of ipilimumab in combination with ADT or radiotherapy [77,83]. Both trials demonstrated a greater antitumor activity for ipilimumab plus ADT [83] or radiotherapy [77] as compared to either treatment as monotherapy, with manageable toxicities. Subsequently, preliminary results from two phase III trials demonstrated antitumor activity for ipilimumab, with longer PFS and higher PSA response than placebo, but without significant improvement in OS [74,76]. However, the long-term analysis of the phase III CA184-043 trial showed improved OS with ipilimumab plus radiotherapy compared to placebo plus radiotherapy for post-docetaxel mCRPC patients. Two-, three-, and five-year OS were higher in patients treated with ipilimumab (25% vs 16%, 15% vs 8%, and 7.9% vs 2.7%, respectively) [75,84]. In a recent study of ipilimumab in 30 metastatic PC patients who had an incomplete biochemical response to ADT alone, 30% of patients had a >50% reduction in PSA, with one patient achieving an exceptional >90% reduction. However, this study was interrupted at the interim analysis because the primary endpoint was not achieved [85]. In summary, although these studies suggest that ipilimumab treatment may confer a PFS benefit in mCRPC patients, to date, there is insufficient evidence to justify its use in routine clinical practice.
Previous studies demonstrate that although treatment with ipilimumab increases tumor-infiltrating T cells in PC patients, it also induces compensatory immune-inhibitory pathways, including PD-1/PD-L1, likely suppressing T-cell responses [86]. In fact, due to its poor efficacy as monotherapy, ipilimumab is being tested in several combination trials for mCRPC patients. One promising strategy is combining two different ICIs, as was done in the CheckMate-650 trial, which combined the PD-1 inhibitor nivolumab with ipilimumab in the second- and third-line settings. Results showed a 25% ORR in the pre-chemotherapy cohort 1 and a 10% ORR in the post-chemotherapy cohort 2; median rPFS was 5.5 and 3.8 months, and median OS was 19.0 and 15.2 months, respectively [72]. Interestingly, the biomarker analysis demonstrated improved efficacy in patients with high TMB, HR deficiency, DDR gene alterations, and PD-L1 expression. However, the high rates of toxicity, treatment-related discontinuation, and deaths in the CheckMate-650 trial highlight the need for improving the tolerability of the combination of nivolumab plus ipilimumab [87]. In fact, this study will expand inclusion with 405 additional patients in four arms to assess the role of the combination of ipilimumab and nivolumab at different schedules and in two additional arms with ipilimumab as monotherapy and in combination with cabazitaxel. Similarly, other trials have evaluated the efficacy of ipilimumab in combination with PD-1/PD-L1 inhibitors, especially in patients with AR-V7-positive disease or alterations in DDR genes [73,88].
Finally, ipilimumab has been also tested in combination with ARPIs (NCT01688492), sipuleucel-T [78], vaccines [79], and evofosfamide, an investigational hypoxia-activated prodrug that can potentially reduce hypoxia in the tumor microenvironment (TME) and improve the efficacy of ICIs (NCT03098160) (Table 1 and Table 2).

5. Mechanisms of Resistance and Potential Predictive Biomarkers of ICI Response

The real efficacy of ICIs in mCRPC remains elusive and investigational. A better understanding of the mechanisms of sensitivity and resistance to ICIs could help improve the efficacy of these therapies as well as identify new molecular and histological predictive biomarkers [34]. Several potential mechanisms of PC resistance to immunotherapy have been proposed. The slow disease progression of PC has been suggested as a potential explanation for resistance and tolerance to immunotherapy [89]. However, one of the most important factors in tumor resistance to ICIs is the TME. Patients with metastatic PC have dysfunctional cellular immunity and an increased immunosuppressive TME, making the prostate TME unsuitable for tumor-infiltrating immune cells with antitumor activities, such as CD8+ T cells and natural killer (NK) cells, leading to an immunosuppressive environment [90] (Figure 1).
Histocompatibility complex (MHC) Class I proteins are normally expressed on nucleated cells, such as dendritic cells (DC), and present cytosolic peptides to T lymphocytes, triggering an immunostimulatory signal cascade resulting in T cell proliferation and activation [91]. Consequently, loss of MHC Class I expression is a common immune evasion mechanism employed by a variety of cancer types, including PC [91,92]. Interestingly, MHC class I chain-related molecules (MICs), such as MICA and MICB, are proteins expressed in the membrane of tumor cells that bind to the C-type lectin-like stimulatory immune receptor NKG2D in NK cells and CD8+ T cells, activating its cytotoxic effects [93]. Therefore, it has been suggested that the MIC-NKG2D axis participates in epithelial tumor immune surveillance [94]. Of note, it has been shown that aggressive tumors, such as metastatic PC, cleave MICA/B from the membrane of NK and CD8+ T cells, release the soluble form (sMIC) into the plasma, and downregulate the NKG2D receptor from the immune cells, thereby promoting immune suppression and tumor escape [95,96,97]. Several studies in PC have found that the loss of MICA/B expression from the cell surface and the release of sMIC are associated with a more aggressive tumor phenotype [98]. Importantly, high levels of circulating soluble NKG2D ligands have been associated with poor clinical outcome to PD1/PD-L1 blockade [99], while targeting sMIC improves the response of sMIC+ tumors to PD1/PD-L1 inhibition by enhancing antigen-specific CD8+ T cell enrichment and function [100].
Furthermore, another factor involved in the importance of the TME in tumor resistance to ICIs is the role of cancer-associated fibroblasts (CAFs), the predominant cell type in the TME. CAFs promote PC carcinogenesis, metastatic progression and therapy resistance by promoting a reactive stroma [101]. It is well described that PC cells establish a crosstalk with CAFs and inflammatory cells to coordinate immune cell recruitment and activation through several signalling factors, including cytokines, chemokines, and growth factors [101,102]. Interestingly, CAFs can recruit monocytes by releasing monocyte chemotactic protein-1 (MCP-1) and stromal cell-derived factor-1 (SDF-1) cytokines, thereby causing them to differentiate into M2-like macrophages, which are capable of exerting their immunosuppressive role via the PD-1 axis. Moreover, it has been suggested that CAFs are able to induce the trans-differentiation of M1 macrophages to tumor-associated M2 macrophages (TAMs) in breast cancer models [103]. In the same line, Ting and colleagues demonstrated that the inhibition of CAF-mediated MCP-1 secretion reduces PC tumor growth and in turn is associated with a reduction in immune cell recruitment [104]. Taken together, these studies suggest that targeting CAFs may represent a new attractive therapeutic strategy to use in combination with ICIs. CAFs have a high expression of fibroblast activation protein-alpha (FAPα), which has been shown to be involved in resistance to immunotherapy [105]. However, although FAP depletion synergizes with anti-PD-L1 immunotherapy in preclinical models of pancreatic ductal carcinoma [106], sadly, FAP-targeted approaches have shown a lack of efficacy in clinical trials [107]. However, novel and potent antibody–drug conjugates like OMTX705 have shown preclinical antitumor activity as single agents and, in combination with chemotherapy in tumors resistant to PD-1 inhibitors, suggest that they may represent an important therapeutic target in combination with ICIs in mCRPC [108].
On the other hand, mCRPC patients present an increased number of immunosuppressive cells including myeloid-derived suppressor cells (MSDC) and regulatory T (Treg) cells in the TME and in peripheral blood [109,110]. Treg cells, one of the key components in tumor immune tolerance and evasion, act by suppressing the pro-inflammatory type 1 CD4+ helper T (Th1) and CD8+ T cells [111]. Cytokines, such as vascular endothelial growth factor (VEGF), transforming growth factor β (TGF-β), interleukin-10 (IL-10), and prostaglandin E2, in the TME are responsible for the recruitment of Tregs and the inhibition of proliferation, activation, and infiltration of cytotoxic lymphocytes [112]. Therefore, depletion of Treg cells is being studied as a possible cancer therapy that could improve the therapeutic effect of PD-1/PD-L1 and CTLA-4 inhibitors [113]. Interestingly, Jiao and colleagues demonstrated that mCRPC bone metastases promote osteoclast-mediated bone resorption that releases TGF-β, which restrains Th1 lineage development. Therefore, blocking TGF-β along with ICIs increases Th1 subsets, promotes clonal expansion of CD8+ T cells and subsequent regression of bone metastases, and improves survival [114]. In addition to the presence of immunosuppressive Treg cells, several reports have demonstrated that high infiltration of M2 TAMs in the PC TME is pro-tumorigenic. In fact, these macrophages secrete high levels of M2-associated immunosuppressive cytokines and chemokines, with TGF-β2 being the most highly expressed [115]. Given the established role of TGF-β in immune evasion, this may be an important factor contributing to poor infiltration of cytotoxic lymphocytes in PC. These findings reinforce the critical immunosuppressive role of TGF-β in the context of current ICIs, where targeting TGF-β prior to ICI treatment has been suggested as an approach to improve response [116]. Interestingly, in preclinical models, PTEN-null prostate tumors are strongly infiltrated by TAMs expressing CXCR2, and activation of this receptor through CXCL2 polarizes macrophages toward an anti-inflammatory phenotype. Pharmacological blockade of the CXCR2 receptor by a selective antagonist promoted the re-education of TAMs toward a pro-inflammatory phenotype [117]. In fact, a phase II trial combining pembrolizumab with the CXCR1/CXCR2 antagonist navarixin in mCRPC patients is currently ongoing (NCT03473925). Additionally, Wise and colleagues described Dickkopf-1 (DKK1) gene expression as a novel contributor to the immunosuppressive TME of mCRPCs with low AR and without neuroendocrine signaling [118]. These data provided the rationale for an ongoing clinical trial targeting DKK1 in mCRPC (NCT03837353). In conclusion, the development of mechanism-driven immunotherapies that can restore NK and CD8+ T-cell function, as well as overcome the immunosuppressive prostate TME, are essential to improving the efficacy of ICIs in mCRPC.
Although this remains a controversial issue, compelling evidence indicates that tumors with an elevated TMB, such as melanoma or NSCLC, promote the expression of mutation-associated neoantigens that might be recognized by T cells, which then attack tumor cells [52,53]. PC has what is commonly referred to as a “cold” TME, with a lower level of tumor-associated antigens and neoantigens, which is an important mechanism of resistance to ICIs [55,119]. Interestingly, defects in HR pathway genes may also be associated with an increase in genomic instability, neoantigen load, PD-L1 expression, and tumor-infiltrating lymphocytes [120]. Alterations in HR pathway genes, including BRCA2, BRCA1, ATM, CDK12, and PALB2, occur in approximately 20–25% of mCRPC cases at either the somatic or the germline level [27,28], making these patients potentially more responsive to ICIs and suggesting that HR pathway genes may be potential predictive biomarkers of response to ICIs. Indeed, several studies, including the KEYNOTE-199 and CheckMate-650 trials, have shown a trend towards a higher ORR to pembrolizumab in HR-deficient mCRPC patients [59,72]. Particularly, mCRPC tumors with CDK12 biallelic mutations (approximately 4–7% of all mCRPC tumors) have a unique immune signature and a large number of gene fusions, with an increase in neoantigens and subsequent T-cell infiltration, indicating that these CDK12 mutations may be a useful predictive biomarker to identify mCRPC patients likely to respond to PD-1 inhibitors [121,122]. Clinical trials to prospectively assess the efficacy of ICIs in patients with CDK12 alterations are ongoing (NCT03810105, NCT03570619). Interestingly, preliminary evidence also suggests that AR-V7-expressing metastatic PC, an aggressive tumor phenotype with poor PFS and OS, may be enriched for DNA-repair defects, rendering them more sensitive to immune-checkpoint blockade [123] (NCT03061539, NCT02601014; Table 1 and Table 2). However, a recent phase II non-randomized trial showed only modest activity for nivolumab plus ipilimumab in patients with AR-V7-expressing PC [73].
The role of alterations in DDR genes in PARPi sensitivity is widely known [30,31] and several trials have evaluated the combination of ICIs with PARPi. The efficacy of pembrolizumab plus olaparib was evaluated in the cohort A of the KEYNOTE-365 trial, with promising results. Seven patients (9%) achieved a PSA response of >50%, eight (10%) had an ORR, and median OS was 14 months [80]. In addition, another trial combining the PARPi rucaparib with nivolumab is currently ongoing (NCT03572478; Table 2).
Another potential biomarker of sensitivity to ICIs may be PD-L1 expression. In fact, PD-L1 is downregulated in many advanced PC cases, which might partly explain the negative results observed in trials with PD-1/PD-L1 inhibitors [60,61,124]. It has been suggested that PD-L1 expression varies in the different stages of PC progression and may depend on therapies received prior to disease progression [125]. Although the KEYNOTE-028 trial suggested that PD-L1 expression could predict response to ICIs [62], the larger KEYNOTE-199 trial found that the ORR in the PD-L1-negative and the PD-L1-positive cohorts was 3% and 5%, respectively [59]. These results illustrate the low accuracy of PD-L1 as a biomarker of response to ICIs in mCRPC patients. Moreover, it is also important to take into account that PD-L1 expression—in PC and in other cancers—may vary depending on the assay technique and antibody clone as well as on the cut-off used to define positivity/negativity. The fact that the method used for assessing PD-L1 expression is not yet standardized is an important barrier to its clinical use as a predictive biomarker.
In conclusion, although recent years have witnessed some progress, further research is warranted to elucidate the biological and pathological mechanisms underlying resistance to ICIs in order to find effective predictive biomarkers of response.

6. Conclusions and Future Challenges

Taken together, all these data suggest that while it is true that one of the most promising approaches to activate therapeutic antitumor immunity is the blockade of immune checkpoints such as CTLA-4 or PD-1/PD-L1, the overall activity of immune checkpoint mAbs in unselected patients with advanced PC has so far been limited. However, it is noteworthy that some subgroups of PC patients may obtain a long-term benefit from immunotherapy; therefore, it is important to focus our efforts on elucidating the biological and pathological mechanisms underlying resistance to ICIs to identify those patients who are likely to benefit. We must be able to extrapolate and adapt the knowledge acquired in other tumors, where immunotherapy is more established, and apply it in PC. To date, there are several biomarkers being studied that may help to predict the benefit of CTLA-4 and PD-1/PD-L1 inhibition, including PD-L1 expression in tumor and immune cells and high TMB in patients with dMMR, MSI-H or HR deficiency. However, we must be aware that it is unlikely that a single marker will be able to predict response to immunotherapy and a panel of biomarkers will probably be needed to define which patients may benefit from these treatments.
In fact, there are currently over 100 ongoing immunotherapy-based clinical trials in PC, many of which are testing combination therapies or new ICIs. For instance, B7-H3, a member of the B7 ligands superfamily, is a newly investigated immune checkpoint target. Although its receptor is still unknown, B7-H3 is expressed by prostate tumor cells [126,127] and its overexpression inhibits T-cell function, thereby contributing to immune evasion [128,129]. For this reason, B7-H3 inhibitors, such as enoblituzumab, are under clinical investigation in several phase I and II trials in metastatic PC, with promising preliminary results [130] (NCT02628535, NCT02923180, NCT01391143). In addition, other immune checkpoint targets are in various stages of clinical development, including LAG-3 [131], OX40 [132], and 4-1BBL [133].
In summary, although there are still many obstacles to overcome, the scenario of immunotherapy in PC is encouraging. Current advances in the combination of immunotherapy with other treatments, the identification of predictive biomarkers, and the determination of new immune checkpoint targets suggest that immunotherapy will be a promising strategy in mCRPC in the coming years.

Author Contributions

Conceptualization: V.R.d.P. and A.F. Writing—Original draft preparation: V.R.d.P.; J.C.P. and A.F. Writing—Review and Editing: V.R.d.P., J.C.P., L.N., O.E. and A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors thank Renée Grupp for assistance in drafting the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Davies, A.; Conteduca, V.; Zoubeidi, A.; Beltran, H. Biological Evolution of Castration-resistant Prostate Cancer. Eur. Urol. Focus 2019, 5, 147–154. [Google Scholar] [CrossRef] [PubMed]
  3. Sartor, O.; de Bono, J.S. Metastatic Prostate Cancer. N. Engl. J. Med. 2018, 378, 645–657. [Google Scholar] [CrossRef] [PubMed]
  4. Watson, P.A.; Arora, V.K.; Sawyers, C.L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer 2015, 15, 701–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ryan, C.J.; Smith, M.R.; de Bono, J.S.; Molina, A.; Logothetis, C.J.; de Souza, P.; Fizazi, K.; Mainwaring, P.; Piulats, J.M.; Ng, S.; et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 2013, 368, 138–148. [Google Scholar] [CrossRef] [Green Version]
  6. De Bono, J.S.; Logothetis, C.J.; Molina, A.; Fizazi, K.; North, S.; Chu, L.; Chi, K.N.; Jones, R.J.; Goodman, O.B., Jr.; Saad, F.; et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 2011, 364, 1995–2005. [Google Scholar] [CrossRef]
  7. Beer, T.M.; Armstrong, A.J.; Rathkopf, D.E.; Loriot, Y.; Sternberg, C.N.; Higano, C.S.; Iversen, P.; Bhattacharya, S.; Carles, J.; Chowdhury, S.; et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 2014, 371, 424–433. [Google Scholar] [CrossRef] [Green Version]
  8. Scher, H.I.; Fizazi, K.; Saad, F.; Taplin, M.E.; Sternberg, C.N.; Miller, K.; de Wit, R.; Mulders, P.; Chi, K.N.; Shore, N.D.; et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 2012, 367, 1187–1197. [Google Scholar] [CrossRef] [Green Version]
  9. Smith, M.R.; Saad, F.; Chowdhury, S.; Oudard, S.; Hadaschik, B.A.; Graff, J.N.; Olmos, D.; Mainwaring, P.N.; Lee, J.Y.; Uemura, H.; et al. Apalutamide Treatment and Metastasis-free Survival in Prostate Cancer. N. Engl. J. Med. 2018, 378, 1408–1418. [Google Scholar] [CrossRef]
  10. Hussain, M.; Fizazi, K.; Saad, F.; Rathenborg, P.; Shore, N.; Ferreira, U.; Ivashchenko, P.; Demirhan, E.; Modelska, K.; Phung, B.S.; et al. Enzalutamide in Men with Nonmetastatic, Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2018, 378, 2465–2474. [Google Scholar] [CrossRef]
  11. Fizazi, K.; Shore, N.; Tammela, T.L.; Ulys, A.; Vjaters, E.; Polyakov, S.; Jievaltas, M.; Luz, M.; Alekseev, B.; Kuss, I.; et al. Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2019, 380, 1235–1246. [Google Scholar] [CrossRef]
  12. Berthold, D.R.; Pond, G.R.; Soban, F.; de Wit, R.; Eisenberger, M.; Tannock, I.F. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: Updated survival in the TAX 327 study. J. Clin. Oncol. 2008, 26, 242–245. [Google Scholar] [CrossRef]
  13. Petrylak, D.P.; Tangen, C.M.; Hussain, M.H.; Lara, P.N., Jr.; Jones, J.A.; Taplin, M.E.; Burch, P.A.; Berry, D.; Moinpour, C.; Kohli, M.; et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N. Engl. J. Med. 2004, 351, 1513–1520. [Google Scholar] [CrossRef] [Green Version]
  14. Tannock, I.F.; de Wit, R.; Berry, W.R.; Horti, J.; Pluzanska, A.; Chi, K.N.; Oudard, S.; Theodore, C.; James, N.D.; Turesson, I.; et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 2004, 351, 1502–1512. [Google Scholar] [CrossRef] [Green Version]
  15. James, N.D.; Sydes, M.R.; Clarke, N.W.; Mason, M.D.; Dearnaley, D.P.; Spears, M.R.; Ritchie, A.W.; Parker, C.C.; Russell, J.M.; Attard, G.; et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): Survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 2016, 387, 1163–1177. [Google Scholar] [CrossRef] [Green Version]
  16. Sweeney, C.J.; Chen, Y.H.; Carducci, M.; Liu, G.; Jarrard, D.F.; Eisenberger, M.; Wong, Y.N.; Hahn, N.; Kohli, M.; Cooney, M.M.; et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N. Engl. J. Med. 2015, 373, 737–746. [Google Scholar] [CrossRef] [PubMed]
  17. Vrignaud, P.; Semiond, D.; Lejeune, P.; Bouchard, H.; Calvet, L.; Combeau, C.; Riou, J.F.; Commercon, A.; Lavelle, F.; Bissery, M.C. Preclinical antitumor activity of cabazitaxel, a semisynthetic taxane active in taxane-resistant tumors. Clin. Cancer Res. 2013, 19, 2973–2983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. De Bono, J.S.; Oudard, S.; Ozguroglu, M.; Hansen, S.; Machiels, J.P.; Kocak, I.; Gravis, G.; Bodrogi, I.; Mackenzie, M.J.; Shen, L.; et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: A randomised open-label trial. Lancet 2010, 376, 1147–1154. [Google Scholar] [CrossRef]
  19. de Wit, R.; de Bono, J.; Sternberg, C.N.; Fizazi, K.; Tombal, B.; Wulfing, C.; Kramer, G.; Eymard, J.C.; Bamias, A.; Carles, J.; et al. Cabazitaxel versus Abiraterone or Enzalutamide in Metastatic Prostate Cancer. N. Engl. J. Med. 2019, 381, 2506–2518. [Google Scholar] [CrossRef] [PubMed]
  20. Hager, S.; Ackermann, C.J.; Joerger, M.; Gillessen, S.; Omlin, A. Anti-tumour activity of platinum compounds in advanced prostate cancer-a systematic literature review. Ann. Oncol. 2016, 27, 975–984. [Google Scholar] [CrossRef]
  21. Leal, F.; Garcia-Perdomo, H.A. Effectiveness of Platinum-Based Chemotherapy in Patients With Metastatic Prostate Cancer: Systematic Review and Meta-analysis. Clin. Genitourin. Cancer 2019, 17, e627–e644. [Google Scholar] [CrossRef]
  22. Corn, P.G.; Heath, E.I.; Zurita, A.; Ramesh, N.; Xiao, L.; Sei, E.; Li-Ning-Tapia, E.; Tu, S.M.; Subudhi, S.K.; Wang, J.; et al. Cabazitaxel plus carboplatin for the treatment of men with metastatic castration-resistant prostate cancers: A randomised, open-label, phase 1–2 trial. Lancet Oncol. 2019, 20, 1432–1443. [Google Scholar] [CrossRef]
  23. Aparicio, A.M.; Harzstark, A.L.; Corn, P.G.; Wen, S.; Araujo, J.C.; Tu, S.M.; Pagliaro, L.C.; Kim, J.; Millikan, R.E.; Ryan, C.; et al. Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin. Cancer Res. 2013, 19, 3621–3630. [Google Scholar] [CrossRef] [Green Version]
  24. Beltran, H.; Prandi, D.; Mosquera, J.M.; Benelli, M.; Puca, L.; Cyrta, J.; Marotz, C.; Giannopoulou, E.; Chakravarthi, B.V.; Varambally, S.; et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat. Med. 2016, 22, 298–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Aparicio, A.M.; Shen, L.; Tapia, E.L.; Lu, J.F.; Chen, H.C.; Zhang, J.; Wu, G.; Wang, X.; Troncoso, P.; Corn, P.; et al. Combined Tumor Suppressor Defects Characterize Clinically Defined Aggressive Variant Prostate Cancers. Clin. Cancer Res. 2016, 22, 1520–1530. [Google Scholar] [CrossRef] [Green Version]
  26. Ruiz de Porras, V.; Wang, X.C.; Palomero, L.; Marin-Aguilera, M.; Sole-Blanch, C.; Indacochea, A.; Jimenez, N.; Bystrup, S.; Bakht, M.; Conteduca, V.; et al. Taxane-induced Attenuation of the CXCR2/BCL-2 Axis Sensitizes Prostate Cancer to Platinum-based Treatment. Eur. Urol. 2020. [Google Scholar] [CrossRef]
  27. Lozano, R.; Castro, E.; Aragon, I.M.; Cendon, Y.; Cattrini, C.; Lopez-Casas, P.P.; Olmos, D. Genetic aberrations in DNA repair pathways: A cornerstone of precision oncology in prostate cancer. Br. J. Cancer 2021, 124, 552–563. [Google Scholar] [CrossRef] [PubMed]
  28. Nombela, P.; Lozano, R.; Aytes, A.; Mateo, J.; Olmos, D.; Castro, E. BRCA2 and Other DDR Genes in Prostate Cancer. Cancers 2019, 11, 352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Nizialek, E.; Antonarakis, E.S. PARP Inhibitors in Metastatic Prostate Cancer: Evidence to Date. Cancer Manag. Res. 2020, 12, 8105–8114. [Google Scholar] [CrossRef] [PubMed]
  30. Mateo, J.; Carreira, S.; Sandhu, S.; Miranda, S.; Mossop, H.; Perez-Lopez, R.; Nava Rodrigues, D.; Robinson, D.; Omlin, A.; Tunariu, N.; et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N. Engl. J. Med. 2015, 373, 1697–1708. [Google Scholar] [CrossRef]
  31. De Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef] [PubMed]
  32. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bilusic, M.; Madan, R.A.; Gulley, J.L. Immunotherapy of Prostate Cancer: Facts and Hopes. Clin. Cancer Res. 2017, 23, 6764–6770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rizzo, A.; Mollica, V.; Cimadamore, A.; Santoni, M.; Scarpelli, M.; Giunchi, F.; Cheng, L.; Lopez-Beltran, A.; Fiorentino, M.; Montironi, R.; et al. Is There a Role for Immunotherapy in Prostate Cancer? Cells 2020, 9, 2051. [Google Scholar] [CrossRef]
  35. Ribas, A. Releasing the Brakes on Cancer Immunotherapy. N. Engl. J. Med. 2015, 373, 1490–1492. [Google Scholar] [CrossRef]
  36. Wolchok, J.D.; Kluger, H.; Callahan, M.K.; Postow, M.A.; Rizvi, N.A.; Lesokhin, A.M.; Segal, N.H.; Ariyan, C.E.; Gordon, R.A.; Reed, K.; et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2013, 369, 122–133. [Google Scholar] [CrossRef] [Green Version]
  37. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [Green Version]
  38. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.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]
  39. Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Procopio, G.; Plimack, E.R.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1803–1813. [Google Scholar] [CrossRef]
  40. Sharma, P.; Callahan, M.K.; Bono, P.; Kim, J.; Spiliopoulou, P.; Calvo, E.; Pillai, R.N.; Ott, P.A.; de Braud, F.; Morse, M.; et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): A multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 2016, 17, 1590–1598. [Google Scholar] [CrossRef] [Green Version]
  41. Ferris, R.L.; Blumenschein, G., Jr.; 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] [PubMed]
  42. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. 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]
  44. Higano, C.S.; Schellhammer, P.F.; Small, E.J.; Burch, P.A.; Nemunaitis, J.; Yuh, L.; Provost, N.; Frohlich, M.W. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer 2009, 115, 3670–3679. [Google Scholar] [CrossRef] [PubMed]
  45. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Antonarakis, E.S.; Shaukat, F.; Isaacsson Velho, P.; Kaur, H.; Shenderov, E.; Pardoll, D.M.; Lotan, T.L. Clinical Features and Therapeutic Outcomes in Men with Advanced Prostate Cancer and DNA Mismatch Repair Gene Mutations. Eur. Urol. 2019, 75, 378–382. [Google Scholar] [CrossRef] [PubMed]
  47. 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]
  48. 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]
  49. Raymond, V.M.; Mukherjee, B.; Wang, F.; Huang, S.C.; Stoffel, E.M.; Kastrinos, F.; Syngal, S.; Cooney, K.A.; Gruber, S.B. Elevated risk of prostate cancer among men with Lynch syndrome. J. Clin. Oncol. 2013, 31, 1713–1718. [Google Scholar] [CrossRef] [Green Version]
  50. Blankenstein, T.; Coulie, P.G.; Gilboa, E.; Jaffee, E.M. The determinants of tumour immunogenicity. Nat. Rev. Cancer 2012, 12, 307–313. [Google Scholar] [CrossRef]
  51. Miao, D.; Van Allen, E.M. Genomic determinants of cancer immunotherapy. Curr. Opin. Immunol. 2016, 41, 32–38. [Google Scholar] [CrossRef]
  52. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
  54. Maleki Vareki, S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 157. [Google Scholar] [CrossRef]
  55. Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.L.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013, 499, 214–218. [Google Scholar] [CrossRef] [PubMed]
  56. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  57. 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] [PubMed]
  58. Petrylak, D.P.; Loriot, Y.; Shaffer, D.; Braiteh, F.; Powderly, J.; Harshman, L.C.; Conkling, P.; Delord, J.P.; Gordon, M.; Kim, J.W.; et al. Safety and Clinical Activity of Atezolizumab in Patients With Metastatic Castration-Resistant Prostate Cancer: A Phase I Study. Clin. Cancer Res. 2021. [Google Scholar] [CrossRef]
  59. 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]
  60. Martin, A.M.; Nirschl, T.R.; Nirschl, C.J.; Francica, B.J.; Kochel, C.M.; van Bokhoven, A.; Meeker, A.K.; Lucia, M.S.; Anders, R.A.; DeMarzo, A.M.; et al. Paucity of PD-L1 expression in prostate cancer: Innate and adaptive immune resistance. Prostate Cancer Prostatic Dis. 2015, 18, 325–332. [Google Scholar] [CrossRef] [Green Version]
  61. Baas, W.; Gershburg, S.; Dynda, D.; Delfino, K.; Robinson, K.; Nie, D.; Yearley, J.H.; Alanee, S. Immune Characterization of the Programmed Death Receptor Pathway in High Risk Prostate Cancer. Clin. Genitourin Cancer 2017, 15, 577–581. [Google Scholar] [CrossRef] [PubMed]
  62. 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]
  63. Bishop, J.L.; Sio, A.; Angeles, A.; Roberts, M.E.; Azad, A.A.; Chi, K.N.; Zoubeidi, A. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget 2015, 6, 234–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Graff, J.N.; Beer, T.M.; Alumkal, J.J.; Slottke, R.E.; Redmond, W.L.; Thomas, G.V.; Thompson, R.F.; Wood, M.A.; Koguchi, Y.; Chen, Y.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. Sweeney, C.J.; Gillessen, S.; Rathkopf, D.; Matsubara, N.; Drake, C.; Fizazi, K.; Piulats, J.M.; Wysocki, P.J.; Buchschacher, G.L.; Doss, J.; et al. Abstract CT014: IMbassador250: A phase III trial comparing atezolizumab with enzalutamide vs enzalutamide alone in patients with metastatic castration-resistant prostate cancer (mCRPC). Cancer Res. 2020, 80, CT014. [Google Scholar]
  67. McNeel, D.G.; Eickhoff, J.C.; Wargowski, E.; Zahm, C.; Staab, M.J.; Straus, J.; Liu, G. Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget 2018, 9, 25586–25596. [Google Scholar] [CrossRef] [Green Version]
  68. Lopez, J.S.; Biondo, A.; Tiu, C.; Scaranti, M.; Ameratunga, M.; Zachariou, A.; Turner, A.; Tunariu, N.; Prout, T.; Parmar, M.; et al. Abstract CT140: Proof-of-concept evidence of immune modulation by blockade of the phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway in the phase I dose escalation study of Ipatasertib (Ipa) in combination with atezolizumab (A) in patients (pts) with advanced solid tumors (Ice-CAP). Cancer Res. 2020, 80, CT140. [Google Scholar]
  69. Morris, M.J.; Fong, L.; Petrylak, D.P.; Sartor, A.O.; Higano, C.S.; Pagliaro, L.C.; Alva, A.S.; Appleman, L.J.; Tan, W.; Vaishampayan, U.N.; et al. Safety and clinical activity of atezolizumab (atezo) + radium-223 dichloride (r-223) in 2L metastatic castration-resistant prostate cancer (mCRPC): Results from a phase Ib clinical trial. J. Clin. Oncol. 2020, 38, 5565. [Google Scholar] [CrossRef]
  70. Rosser, C.J.; Hirasawa, Y.; Acoba, J.D.; Tamura, D.J.; Pal, S.K.; Huang, J.; Scholz, M.C.; Dorff, T.B. Phase Ib study assessing different sequencing regimens of atezolizumab (anti-PD-L1) and sipuleucel-T (SipT)in patients who have asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer. J. Clin. Oncol. 2020, 38, e17564. [Google Scholar] [CrossRef]
  71. Yap, T.A.; Konstantinopoulos, P.; Telli, M.L.; Saraykar, S.; Beck, J.T.; Galsky, M.D.; Abraham, J.; Wise, D.R.; Khasraw, M.; Rubovszky, G.; et al. Abstract P1–19-03: JAVELIN PARP Medley, a phase 1b/2 study of avelumab plus talazoparib: Results from advanced breast cancer cohorts. Cancer Res. 2020, 80, P1-19-03. [Google Scholar]
  72. 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. 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]
  73. Shenderov, E.; Boudadi, K.; Fu, W.; Wang, H.; Sullivan, R.; Jordan, A.; Dowling, D.; Harb, R.; Schonhoft, J.; Jendrisak, A.; et al. Nivolumab plus ipilimumab, with or without enzalutamide, in AR-V7-expressing metastatic castration-resistant prostate cancer: A phase-2 nonrandomized clinical trial. Prostate 2021, 81, 326–338. [Google Scholar] [CrossRef] [PubMed]
  74. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; van den Eertwegh, A.J.; 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. Fizazi, K.; Drake, C.G.; Beer, T.M.; Kwon, E.D.; Scher, H.I.; Gerritsen, W.R.; Bossi, A.; den Eertwegh, A.; Krainer, M.; Houede, N.; et al. 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. [Google Scholar] [CrossRef]
  76. 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]
  77. Slovin, S.F.; Higano, C.S.; Hamid, O.; Tejwani, S.; Harzstark, A.; Alumkal, J.J.; Scher, H.I.; Chin, K.; Gagnier, P.; McHenry, M.B.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  78. Scholz, M.; Yep, S.; Chancey, M.; Kelly, C.; Chau, K.; Turner, J.; Lam, R.; Drake, C.G. Phase I clinical trial of sipuleucel-T combined with escalating doses of ipilimumab in progressive metastatic castrate-resistant prostate cancer. Immunotargets Ther. 2017, 6, 11–16. [Google Scholar] [CrossRef] [Green Version]
  79. Van den Eertwegh, A.J.; Versluis, J.; van den Berg, H.P.; Santegoets, S.J.; van Moorselaar, R.J.; van der Sluis, T.M.; Gall, H.E.; Harding, T.C.; Jooss, K.; Lowy, I.; et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 509–517. [Google Scholar] [CrossRef]
  80. Yu, E.Y.; Piulats, J.M.; Gravis, G.; Laguerre, B.; Arija, J.A.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]
  81. Tang, F.; Du, X.; Liu, M.; Zheng, P.; Liu, Y. Anti-CTLA-4 antibodies in cancer immunotherapy: Selective depletion of intratumoral regulatory T cells or checkpoint blockade? Cell Biosci. 2018, 8, 30. [Google Scholar] [CrossRef] [Green Version]
  82. Small, E.J.; Tchekmedyian, N.S.; Rini, B.I.; Fong, L.; Lowy, I.; Allison, J.P. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin. Cancer Res. 2007, 13, 1810–1815. [Google Scholar] [CrossRef] [Green Version]
  83. Tollefson, M.; Karnes, R.J.; Thompson, R.H.; Granberg, C.; Hillman, D.; Breau, R.; Allison, J.; Kwon, E.; Blute, M. 668 a randomized phase ii study of ipilimumab with androgen ablation compared with androgen ablation alone in patients with advanced prostate cancer. J. Urol. 2010, 183, e261. [Google Scholar] [CrossRef]
  84. Pachynski, R.K. A “Tail” of Immunotherapy in Metastatic Prostate Cancer. Eur. Urol. 2020, 78, 831–833. [Google Scholar] [CrossRef] [PubMed]
  85. Graff, J.N.; Stein, M.N.; Surana, R.; Al Rabadi, L.; Liu, E.; Fong, L.; Bailey, S.; Latour, E.; Newby, T.A.; Moran, A.E.; et al. Phase II Study of Ipilimumab in Men With Metastatic Prostate Cancer With an Incomplete Response to Androgen Deprivation Therapy. Front. Oncol. 2020, 10, 1381. [Google Scholar] [CrossRef] [PubMed]
  86. Gao, J.; Ward, J.F.; Pettaway, C.A.; Shi, L.Z.; Subudhi, S.K.; Vence, L.M.; Zhao, H.; Chen, J.; Chen, H.; Efstathiou, E.; et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat. Med. 2017, 23, 551–555. [Google Scholar] [CrossRef] [Green Version]
  87. Wilson, B.E.; Hansen, A.R. Dual Checkpoint Blockade in Metastatic Castration-Resistant Prostate Cancer: Just a Gambit or Real CheckMate? Cancer Cell. 2020, 38, 438–440. [Google Scholar] [CrossRef]
  88. 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]
  89. Zlotta, A.R.; Egawa, S.; Pushkar, D.; Govorov, A.; Kimura, T.; Kido, M.; Takahashi, H.; Kuk, C.; Kovylina, M.; Aldaoud, N.; et al. Prevalence of prostate cancer on autopsy: Cross-sectional study on unscreened Caucasian and Asian men. J. Natl. Cancer Inst. 2013, 105, 1050–1058. [Google Scholar] [CrossRef] [Green Version]
  90. Pal, S.K.; Moreira, D.; Won, H.; White, S.W.; Duttagupta, P.; Lucia, M.; Jones, J.; Hsu, J.; Kortylewski, M. Reduced T-cell Numbers and Elevated Levels of Immunomodulatory Cytokines in Metastatic Prostate Cancer Patients De Novo Resistant to Abiraterone and/or Enzalutamide Therapy. Int. J. Mol. Sci. 2019, 20, 1831. [Google Scholar] [CrossRef] [Green Version]
  91. Garcia-Lora, A.; Algarra, I.; Garrido, F. MHC class I antigens, immune surveillance, and tumor immune escape. J. Cell Physiol. 2003, 195, 346–355. [Google Scholar] [CrossRef]
  92. 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]
  93. Blery, M.; Vivier, E. NKG2D-MICA Interaction: A Paradigm Shift in Innate Recognition. J. Immunol. 2018, 200, 2229–2230. [Google Scholar] [CrossRef]
  94. Vivier, E.; Tomasello, E.; Paul, P. Lymphocyte activation via NKG2D: Towards a new paradigm in immune recognition? Curr. Opin. Immunol. 2002, 14, 306–311. [Google Scholar] [CrossRef]
  95. Wu, J.D.; Higgins, L.M.; Steinle, A.; Cosman, D.; Haugk, K.; Plymate, S.R. Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J. Clin. Investig. 2004, 114, 560–568. [Google Scholar] [CrossRef] [PubMed]
  96. Lundholm, M.; Schroder, M.; Nagaeva, O.; Baranov, V.; Widmark, A.; Mincheva-Nilsson, L.; Wikstrom, P. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: Mechanism of immune evasion. PLoS ONE 2014, 9, e108925. [Google Scholar] [CrossRef]
  97. Salih, H.R.; Rammensee, H.G.; Steinle, A. Cutting edge: Down-regulation of MICA on human tumors by proteolytic shedding. J. Immunol. 2002, 169, 4098–4102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Sakiyama, M.J.; Espinoza, I.; Reddy, A.; de Carlo, F.; Kumar, A.; Levenson, A.S.; Bae, S.; Zhou, X.; Claudio, P.P.; Lewin, J.; et al. Race-associated expression of MHC class I polypeptide-related sequence A (MICA) in prostate cancer. Exp. Mol. Pathol. 2019, 108, 173–182. [Google Scholar] [CrossRef] [PubMed]
  99. Lopez-Soto, A.; Gonzalez, S.; Galluzzi, L. Soluble NKG2D ligands limit the efficacy of immune checkpoint blockade. Oncoimmunology 2017, 6, e1346766. [Google Scholar] [CrossRef]
  100. Zhang, J.; Larrocha, P.S.; Zhang, B.; Wainwright, D.; Dhar, P.; Wu, J.D. Antibody targeting tumor-derived soluble NKG2D ligand sMIC provides dual co-stimulation of CD8 T cells and enables sMIC(+) tumors respond to PD1/PD-L1 blockade therapy. J. Immunother. Cancer 2019, 7, 223. [Google Scholar] [CrossRef] [Green Version]
  101. Bonollo, F.; Thalmann, G.N.; Kruithof-de Julio, M.; Karkampouna, S. The Role of Cancer-Associated Fibroblasts in Prostate Cancer Tumorigenesis. Cancers 2020, 12, 1887. [Google Scholar] [CrossRef]
  102. Vickman, R.E.; Broman, M.M.; Lanman, N.A.; Franco, O.E.; Sudyanti, P.A.G.; Ni, Y.; Ji, Y.; Helfand, B.T.; Petkewicz, J.; Paterakos, M.C.; et al. Heterogeneity of human prostate carcinoma-associated fibroblasts implicates a role for subpopulations in myeloid cell recruitment. Prostate 2020, 80, 173–185. [Google Scholar] [CrossRef] [PubMed]
  103. Gok Yavuz, B.; Gunaydin, G.; Gedik, M.E.; Kosemehmetoglu, K.; Karakoc, D.; Ozgur, F.; Guc, D. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1(+) TAMs. Sci. Rep. 2019, 9, 3172. [Google Scholar] [CrossRef]
  104. Ting, H.; Deep, G.; Kumar, S.; Jain, A.K.; Agarwal, C.; Agarwal, R. Beneficial effects of the naturally occurring flavonoid silibinin on the prostate cancer microenvironment: Role of monocyte chemotactic protein-1 and immune cell recruitment. Carcinogenesis 2016, 37, 589–599. [Google Scholar] [CrossRef] [Green Version]
  105. Kraman, M.; Bambrough, P.J.; Arnold, J.N.; Roberts, E.W.; Magiera, L.; Jones, J.O.; Gopinathan, A.; Tuveson, D.A.; Fearon, D.T. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010, 330, 827–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Scott, A.M.; Wiseman, G.; Welt, S.; Adjei, A.; Lee, F.T.; Hopkins, W.; Divgi, C.R.; Hanson, L.H.; Mitchell, P.; Gansen, D.N.; et al. A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin. Cancer Res. 2003, 9, 1639–1647. [Google Scholar]
  108. Fabre, M.; Ferrer, C.; Dominguez-Hormaetxe, S.; Bockorny, B.; Murias, L.; Seifert, O.; Eisler, S.A.; Kontermann, R.E.; Pfizenmaier, K.; Lee, S.Y.; et al. OMTX705, a Novel FAP-Targeting ADC Demonstrates Activity in Chemotherapy and Pembrolizumab-Resistant Solid Tumor Models. Clin. Cancer Res. 2020, 26, 3420–3430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Idorn, M.; Kollgaard, T.; Kongsted, P.; Sengelov, L.; Thor Straten, P. Correlation between frequencies of blood monocytic myeloid-derived suppressor cells, regulatory T cells and negative prognostic markers in patients with castration-resistant metastatic prostate cancer. Cancer Immunol. Immunother. 2014, 63, 1177–1187. [Google Scholar] [CrossRef]
  110. Lopez-Bujanda, Z.; Drake, C.G. Myeloid-derived cells in prostate cancer progression: Phenotype and prospective therapies. J. Leukoc. Biol. 2017, 102, 393–406. [Google Scholar] [CrossRef] [Green Version]
  111. Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [Green Version]
  112. Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat. Rev. Clin. Oncol. 2018, 15, 325–340. [Google Scholar] [CrossRef]
  113. Hoos, A. Development of immuno-oncology drugs—from CTLA4 to PD1 to the next generations. Nat. Rev. Drug Discov. 2016, 15, 235–247. [Google Scholar] [CrossRef]
  114. Jiao, S.; Subudhi, S.K.; Aparicio, A.; Ge, Z.; Guan, B.; Miura, Y.; Sharma, P. Differences in Tumor Microenvironment Dictate T Helper Lineage Polarization and Response to Immune Checkpoint Therapy. Cell 2019, 179, 1177–1190.e13. [Google Scholar] [CrossRef]
  115. Lundholm, M.; Hagglof, C.; Wikberg, M.L.; Stattin, P.; Egevad, L.; Bergh, A.; Wikstrom, 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] [PubMed]
  116. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  117. Di Mitri, D.; Mirenda, M.; Vasilevska, J.; Calcinotto, A.; Delaleu, N.; Revandkar, A.; Gil, V.; Boysen, G.; Losa, M.; Mosole, S.; et al. Re-education of Tumor-Associated Macrophages by CXCR2 Blockade Drives Senescence and Tumor Inhibition in Advanced Prostate Cancer. Cell Rep. 2019, 28, 2156–2168.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Wise, D.R.; Schneider, J.A.; Armenia, J.; Febles, V.A.; McLaughlin, B.; Brennan, R.; Thoren, K.L.; Abida, W.; Sfanos, K.S.; De Marzo, A.M.; et al. Dickkopf-1 Can Lead to Immune Evasion in Metastatic Castration-Resistant Prostate Cancer. JCO Precis. Oncol. 2020, 4, 1167–1179. [Google Scholar] [CrossRef] [PubMed]
  119. Vitkin, N.; Nersesian, S.; Siemens, D.R.; Koti, M. The Tumor Immune Contexture of Prostate Cancer. Front. Immunol. 2019, 10, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. 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] [PubMed] [Green Version]
  121. Wu, Y.M.; Cieslik, 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] [Green Version]
  122. Antonarakis, E.S. Cyclin-Dependent Kinase 12, Immunity, and Prostate Cancer. N. Engl. J. Med. 2018, 379, 1087–1089. [Google Scholar] [CrossRef]
  123. 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]
  124. Isaacsson Velho, P.; Antonarakis, E.S. PD-1/PD-L1 pathway inhibitors in advanced prostate cancer. Expert Rev. Clin. Pharmacol. 2018, 11, 475–486. [Google Scholar] [CrossRef]
  125. Haffner, M.C.; Guner, G.; Taheri, D.; Netto, G.J.; Palsgrove, D.N.; Zheng, Q.; Guedes, L.B.; Kim, K.; Tsai, H.; Esopi, D.M.; et al. Comprehensive Evaluation of Programmed Death-Ligand 1 Expression in Primary and Metastatic Prostate Cancer. Am. J. Pathol. 2018, 188, 1478–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chapoval, A.I.; Ni, J.; Lau, J.S.; Wilcox, R.A.; Flies, D.B.; Liu, D.; Dong, H.; Sica, G.L.; Zhu, G.; Tamada, K.; et al. B7-H3: A costimulatory molecule for T cell activation and IFN-gamma production. Nat. Immunol. 2001, 2, 269–274. [Google Scholar] [CrossRef] [PubMed]
  127. Benzon, B.; Zhao, S.G.; Haffner, M.C.; Takhar, M.; Erho, N.; Yousefi, K.; Hurley, P.; Bishop, J.L.; Tosoian, J.; Ghabili, K.; et al. Correlation of B7-H3 with androgen receptor, immune pathways and poor outcome in prostate cancer: An expression-based analysis. Prostate Cancer Prostatic Dis. 2017, 20, 28–35. [Google Scholar] [CrossRef]
  128. Castellanos, J.R.; Purvis, I.J.; Labak, C.M.; Guda, M.R.; Tsung, A.J.; Velpula, K.K.; Asuthkar, S. B7-H3 role in the immune landscape of cancer. Am. J. Clin. Exp. Immunol. 2017, 6, 66–75. [Google Scholar]
  129. Picarda, E.; Ohaegbulam, K.C.; Zang, X. Molecular Pathways: Targeting B7-H3 (CD276) for Human Cancer Immunotherapy. Clin. Cancer Res. 2016, 22, 3425–3431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Shenderov, E.; Demarzo, A.; Boudadi, K.; Allaf, M.; Wang, H.; Chapman, C.; Pavlovich, C.; Bivalacqua, T.; O’Neal, T.S.; Harb, R.; et al. Phase II neoadjuvant and immunologic study of B7-H3 targeting with enoblituzumab in localized intermediate- and high-risk prostate cancer. J. Clin. Oncol. 2018, 36, 15. [Google Scholar] [CrossRef]
  131. Grosso, J.F.; Kelleher, C.C.; Harris, T.J.; Maris, C.H.; Hipkiss, E.L.; De Marzo, A.; Anders, R.; Netto, G.; Getnet, D.; Bruno, T.C.; et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J. Clin. Investig. 2007, 117, 3383–3392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Redmond, W.L.; Gough, M.J.; Charbonneau, B.; Ratliff, T.L.; Weinberg, A.D. Defects in the acquisition of CD8 T cell effector function after priming with tumor or soluble antigen can be overcome by the addition of an OX40 agonist. J. Immunol. 2007, 179, 7244–7253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Youlin, K.; Li, Z.; Xiaodong, W.; Xiuheng, L.; Hengchen, Z. Combination immunotherapy with 4–1BBL and CTLA-4 blockade for the treatment of prostate cancer. Clin. Dev. Immunol. 2012, 2012, 439235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. An overview of immune cell types involved in the PC TME and the molecular mechanisms of PD-1 and CTLA-4 attenuation of CD8+ T cell activation. APC: antigen-presenting cell; CAFs: cancer-associated fibroblasts; CD8 + CTL: CD8+ cytotoxic T lymphocyte; CD4 + Th1: type 1 CD4+ helper T; CTLA-4: cytotoxic T lymphocyte-associated protein 4; DC: dendritic cell; MDSC: myeloid-derived suppressor cell; MHC: histocompatibility complex; PC: prostate cancer; PD-L1: programmed death ligand 1; PD-1: programmed death 1; Treg: regulatory T cells; TAMs: tumor-associated macrophages; TCR: T cell receptor. Created with BioRender.com (accessed on 29 April 2021).
Figure 1. An overview of immune cell types involved in the PC TME and the molecular mechanisms of PD-1 and CTLA-4 attenuation of CD8+ T cell activation. APC: antigen-presenting cell; CAFs: cancer-associated fibroblasts; CD8 + CTL: CD8+ cytotoxic T lymphocyte; CD4 + Th1: type 1 CD4+ helper T; CTLA-4: cytotoxic T lymphocyte-associated protein 4; DC: dendritic cell; MDSC: myeloid-derived suppressor cell; MHC: histocompatibility complex; PC: prostate cancer; PD-L1: programmed death ligand 1; PD-1: programmed death 1; Treg: regulatory T cells; TAMs: tumor-associated macrophages; TCR: T cell receptor. Created with BioRender.com (accessed on 29 April 2021).
Ijms 22 04712 g001
Table 1. Summary of completed clinical trials evaluating the efficacy of ICIs in monotherapy and in combination with other treatments in mCRPC. AE: adverse events; MTD: maximum tolerated dose; ORR: overall response rate; OS: overall survival; PFS: progression-free survival; rPFS: radiographic progression-free survival; RP: radiographic progression.
Table 1. Summary of completed clinical trials evaluating the efficacy of ICIs in monotherapy and in combination with other treatments in mCRPC. AE: adverse events; MTD: maximum tolerated dose; ORR: overall response rate; OS: overall survival; PFS: progression-free survival; rPFS: radiographic progression-free survival; RP: radiographic progression.
Trial IDTreatmentPatientsPrimary End PointPatientsTrial PhaseResults
NCT02787005
(KEYNOTE-199)
[59]
PembrolizumabChemotherapy-resistant mCRPCORR370IISubstantial antitumor activity with an acceptable safety profile
NCT02054806
(KEYNOTE-028) [62]
PembrolizumabmCRPC with PD-L1 expression in
>1% of tumor or stromal cells
ORR23IbORR: 13%
NCT02312557 [64]Pembrolizumab
+
Enzalutamide
Enzalutamide resistant mCRPCPSA response20IIPSA response: 30%
NCT02499835 [67]Pembrolizumab
+
pTVG-HP(DNA vaccine)
Hormone-resistant
mCRPC
AE32I/IIAcceptable safety profile
PFS
RP
ORR
PSA response
NCT00730639
(MDX-1106)[57]
NivolumabmCRPCAE395IbNo favorable ORR
NCT02985957
(CheckMate-650) [72]
Nivolumab
+
Ipilimumab
mCRPCORRr
PFS
497IISuperior ORR (26%) in chemotherapy-naïve patients
NCT02601014
(STARVE-PC)[73]
Nivolumab
+
Ipilimumab
mCRPC with detectable AR-V7PSA Response
Safety
15IIFavorable outcomes in patients with AR-V7 + PC with DDR
NCT00861614
(CA184-043)
[74,75]
IpilimumabmCRPC following docetaxel therapyOS988IIINo significant improvement in OS
Increased PFS and PSA response
Long-term analysis:
OS improvement in Ipilimumab arm
NCT01057810
(CA184-095)
[76]
IpilimumabChemotherapy-naïve mCRPCOS837IIINo significant improvement in OS
Increased PFS and PSA response
NCT00323882 [77]Ipilimumab
+
Radiotherapy
mCRPCAE
PSA response Tumor response
75I/IIManageable AEs and PSA responses suggestive of clinical activity
NCT01832870
(SIPIPI)
[78]
Ipilimumab
+
Sipuleucel T
Progressive mCRPCAntigen-specific memory T-cell response9IAcceptable safety profile
NCT01510288 [79]Ipilimumab
+
GVAX
mCRPCAE28IAcceptable safety profile
NCT03016312
(IMbassador250) [66]
Atezolizumab
+
Enzalutamide
vs.
Enzalutamide
mCRPCOS730IIIAtezolizumab + enzalutamide do not show improvement in OS over enzalutamide alone
Table 2. Summary of ongoing clinical trials evaluating the efficacy of ICIs in monotherapy and in combination with other treatments in mCRPC. AE: adverse events; DLT: dose limiting toxicities; MTD: maximum tolerated dose; ORR: overall response rate; OS: overall survival; PFS: progression-free survival; rPFS: radiographic progression-free survival; RP: radiographic progression.
Table 2. Summary of ongoing clinical trials evaluating the efficacy of ICIs in monotherapy and in combination with other treatments in mCRPC. AE: adverse events; DLT: dose limiting toxicities; MTD: maximum tolerated dose; ORR: overall response rate; OS: overall survival; PFS: progression-free survival; rPFS: radiographic progression-free survival; RP: radiographic progression.
Trial IDTreatmentIndicationPrimary End PointPatientsTrial PhasePreliminary Results
NCT02861573
(KEYNOTE-365)
[65,80]
Pembrolizumab
+
Coh. A:
Olaparib
Coh. B: Docteaxel
Coh. C: Enzalutamide
Abiraterone resistant mCRPCORR
PSA Response
Safety
210Ib/IICohort A:
ORR: 8%
PSA Resp: 9%
Cohort B:
ORR: 23%
PSA Resp: 34%
Cohort C:
ORR: 12%
PSA Resp: 22%
NCT03834493
(KEYNOTE-641)
Pembrolizumab
+
Enzalutamide
mCRPCOS
rPFS
1200IIIN/A
NCT03093428Pembrolizumab
+
Radium-223
mCRPCExtent Of Immune Cell Infiltration45IIN/A
NCT03473925Pembrolizumab
+
Navarixin
mCRPCORR120IIN/A
NCT03040791
(ImmunoProst)
NivolumabmCRPC with DNA repair defectsPSA
Response
45IIN/A
NCT03572478Nivolumab
+
Rucaparib (PARPi)
mCRPCDLT
T Cell Inflammation
12I/IIN/A
NCT03061539
(NEPTUNES)
Nivolumab
+
Ipilimumab
mCRPC with specific immunogenic signaturesRR
PSA response
CTCs
175IIN/A
NCT03098160Ipilimumab
+
Evofosfamide
Metastatic PCMTD69IN/A
NCT03024216
[70]
Atezolizumab
+
Sipuleucel-T
Asymptomatic or minimally symptomatic mCRPCAE37IbManageable safety profile
NCT03673787
[68]
Atezolizumab
+
Ipatasertib
mCRPC with PTEN loss MTD
AE
51I/IIWell-tolerated
NCT02814669
[69]
Atezolizumab
+
Radium-223
ARPI-resistant
mCRPC
Safety
ORR
45INo dose-limiting toxicities, safety signals, or changes in serum biomarkers
NCT03330405
(JAVELIN PARP Medley)
[71]
Avelumab
+
Talazoparib
mCRPCDLT
ORR
216Ib/IIPreliminary antitumor activity and manageable safety profile
NCT03204812Durvalumab
+
Tremelimumab
Chemotherapy naïve CRPCAE27IIN/A
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ruiz de Porras, V.; Pardo, J.C.; Notario, L.; Etxaniz, O.; Font, A. Immune Checkpoint Inhibitors: A Promising Treatment Option for Metastatic Castration-Resistant Prostate Cancer? Int. J. Mol. Sci. 2021, 22, 4712. https://doi.org/10.3390/ijms22094712

AMA Style

Ruiz de Porras V, Pardo JC, Notario L, Etxaniz O, Font A. Immune Checkpoint Inhibitors: A Promising Treatment Option for Metastatic Castration-Resistant Prostate Cancer? International Journal of Molecular Sciences. 2021; 22(9):4712. https://doi.org/10.3390/ijms22094712

Chicago/Turabian Style

Ruiz de Porras, Vicenç, Juan Carlos Pardo, Lucia Notario, Olatz Etxaniz, and Albert Font. 2021. "Immune Checkpoint Inhibitors: A Promising Treatment Option for Metastatic Castration-Resistant Prostate Cancer?" International Journal of Molecular Sciences 22, no. 9: 4712. https://doi.org/10.3390/ijms22094712

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop