J Urol Oncol > Volume 21(1); 2023 > Article
Kim, Lee, and Kim: Current Update on Prostate Cancer Immunotherapy
This article has been corrected. See "Declaration of Conflict of Interest for Editorial Board Members’ Articles" in Volume 22 on page 95.

Abstract

Immunotherapy encompasses a wide array of treatment modalities that utilize our natural defense system to fight against cancer and in recent years has contributed to significant improvement in survival and quality of life in patients. However, its use in prostate cancer has been limited due to low efficacy and lack of effective biomarkers. Prostate cancer's unfavorable tumor microenvironment characterized by T-cell exclusion and expansion of T-reg cells, along with the possible inhibitory effect of androgen deprivation therapy (ADT) on the immune system further explains this poor response to immunotherapy. Here, we review current immunotherapies and ongoing clinical trials as well as potential biomarkers being investigated to predict treatment responses to immunotherapy. Finally, we discuss the conflicting results on the best approach to sequencing immunotherapy in relation to ADT.

INTRODUCTION

Prostate cancer is the most commonly diagnosed cancer in men and the second most diagnosed disease for men in the United States [1]. Although prostate cancer incidence in South Korea is significantly lower than that in Western countries, the malignancy displays minimal symptoms in the early stages and 33.8% of South Korean patients are diagnosed with at least stage III disease [2]. In contrast to the excellent 5-year survival rate of 100% in localized disease, metastatic prostate cancer has a significantly lower 5-year survival rate of 45.9% [3]. The current mainstay of therapy for advanced and metastatic prostate cancer is androgen deprivation therapy (ADT) but cancer cells eventually escape the androgen dependence and become castration resistant. Immuno-oncological therapy has transformed cancer care by drastically improving survival outcomes and quality of life [4]. Most notably immune checkpoint inhibitors (ICIs) have changed the treatment paradigm in malignancies such as melanoma and lung cancers [5,6]. Despite its promising results and recent advancements in some solid cancers, immunotherapy's utility in advanced prostate cancer has been limited [7-10]. Pembrolizumab, an ICI with a tissue-agonostic indication, and sipuleucel-T, an autologous cellular vaccine, remain the only immunotherapeutics that have received approval from the U.S. Food and Drug Administration (FDA) [11,12]. However, the clinical impact of these agents still remains limited by the high cost and modest improvement in survival.
Herein, we review the conventional and experimental immunotherapeutics in prostate cancer. In addition, cellular mechanism and potential factors that limit immunotherapy in prostate cancer are discussed. Finally, since ADT is the cornerstone of prostate cancer systemic therapy, we cover conflicting results on how ADT may affect the immune system and question the current standard treatment sequence regarding immunotherapy and ADT.

IMMUNE CHECKPOINT INHIBITORS

Tumor cells evade the body's natural immune response via activation of certain immune checkpoint pathways that typically induce T-cell anergy. ICIs interfere with the T-cell coinhibitory signaling pathways to enhance immune-mediated tumoricidal effect [13]. ICIs have become key players in the treatment of many solid tumors, but their clinical benefits in prostate cancer have been disappointing [14].
  • (1) Pembrolizumab is an IgG4 monoclonal antibody against programmed cell death protein 1 (PD-1) on lymphocytes and it remains the only ICI with FDA approval for the treatment of prostate cancer. It prevents PD-1-induced self-tolerance and inactivation of lymphocytes. Current National Comprehensive Cancer Network guidelines dictate the usage of pembrolizumab specifically for unresectable or metastatic microsatellite instability-high (MSI-H), deficient mismatch repair (dMMR) gene, or tumor mutational burden (TMB) equal or higher than 10 mutations per megabase (mut/Mb) prostate cancers [15,16]. Table 1 delineates currently open phase III prostate cancer clinical trials on pembrolizumab— however, 3 out of the 6 trials were discontinued. The KEYLYNK-010 trial combining pembrolizumab to poly ADP-ribose polymerase (PARP) inhibitor olaparib was discontinued due to a higher incidence of serious adverse events without observed benefit in overall survival (OS) or radiographic progression-free survival (PFS) [17]. Likewise, KEYNOTE-921 testing pembrolizumab in combination with chemotherapy failed to show statistically significant improvements [18]. KEYNOTE-991 which compared pembrolizumab with enzalutamide to enzalutamide with placebo in metastatic castration-sensitive prostate cancer will be discontinued after the therapy failed to demonstrate improvements at a planned interim analysis [19]. With these latest updates, KEYNOTE-641 along with KEYNOTE-921, 991 China extension trials are the only phase III trials left, while there is still phase II trial such as KEYNOTE-365 [20,21].

  • (2) Ipilimumab is an FDA-approved ICI that targets CTLA-4 but has not been approved for prostate cancer. In 2 recent phase III clinical trials, it failed to improve OS in patients with metastatic castration-resistant prostate cancer (mCRPC) [7,8]. Currently, there are no phase III trials testing ipilimumab as a monotherapy. However, the NCT03879122 trial is investigating it in combination with nivolumab, as anti-CTLA-4 therapy increases infiltrating T cells and induces interferon-γ which stimulates programmed death ligand-1 (PD-L1) expression [22].

  • (3) Nivolumab is another anti-PD1 antibody that is currently being explored, but has not received FDA approval. The STARVE-PC phase II trial demonstrated that nivolumab and ipilimumab immunotherapy for patients with mCRPC disease that expresses the constitutively active androgen receptor (AR) variant AR-V7, had a modest improvement in OS rate, but the effect size was not sufficient for pursuing a phase III trial [23]. A phase II trial CheckMate-650 investi-gating nivolumab and ipilimumab combination in men with mCRPC disease demonstrated high overall response rates but dosing and scheduling modifications are needed to optimize safety [24]. The ongoing phase III nivolumab trials in prostate cancer are detailed in Table 2. Results from the previous phase II study CheckMate 9KD supported further investigation in the phase III CheckMate-7DX trial which is currently recruiting [25].

  • (4) Atezolizumab is a PD-L1 inhibitor. Table 3 shows ongoing phase III atezolizumab trials in prostate cancer. CONTACT-02 is still recruiting. IMbassador250 trial results are being reviewed, but the trial failed to show the statistical superiority of atezolizumab over the control. However, in a preplanned subgroup analysis, superior PFS was shown in tumors with high levels of PD-L1 expression or high levels of intratumoral CD8-positive T-cell infiltration [9].

Table 1.
Open phase III pembrolizumab trials in prostate cancer
Identifier Title Patients Arms Enrolled Estimated completion (month-year)
NCT03834506* KENOTE-921 NHA-pretreated patients with mCRPC Experimental: pembrolizumab+docetaxel
Comparator: placebo+docetaxel
1,090 Oct-23
NCT03834493 KEYNOTE-641 Patients with mCRPC Experimental: pembrolizumab+enzalutamide
Comparator: placebo+enzalutamide
1,240 Feb-25
NCT04191096* KEYNOTE-991 mHSPC Experimental: pembrolizumab+enzalutamide+ADT
Comparator: placebo+enzalutamide+ADT
1,232 Sep-26
NCT04934722 KEYNOTE-991
China extension
mHSPC Experimental: pembrolizumab+enzalutamide+ADT
Comparator: placebo+enzalutamide+ADT
186 Jan-28
NCT04907227 KEYNOTE-921
China extension
Enzalutamide or abiraterone-pretreated patients with mCRPC Experimental: pembrolizumab+docetaxel
Comparator: placebo+docetaxel
81 Feb-25
NCT03834519* KEYLINK-010 Metastatic CRPC Experimental: pembrolizumab+olaparib
Comparator: abiraterone+prednisone or enzalutamide
793 Sep-23

NHA, next generation hormonal agent; mCRPC, metastatic castration-resistant prostate cancer; mHSPC, metastatic hormone-sensitive prostate cancer; ADT, androge deprivation therapy.

* Discontinued trials.

Table 2.
Open phase III nivolumab trials in prostate cancer
Identifier Title Patients Arms Enrolled Estimated completion
NCT04100018 CheckMate 7DX: a Phase 3, randomized, double-blind study of nivolumab or placebo in combination with docetaxel, in men with mCRPC mCRPC Experimental: nivolumab+docetaxel+prednisone
Comparator: placebo+docetaxel+prednisone
984 Aug-27
NCT03879122 A trial of immunotherapy strategies in metastatic hormone-sensitive prostate cancer mHSPC Experimental A: ADT+docetaxel+nivolumab
Experimental B: ADT+ipilimumab/docetaxel+nivolumab
Comparator: ADT+docetaxel
135 Dec-24

mCRPC, metastatic castration-resistant prostate cancer; ADT, androgen deprivation therapy; mHSPC, metastatic hormone-sensitive prostate cancer.

Table 3.
Open phase III atezolizumab trials
Identifier Title Patients Arms Enrolled Estimated completion
NCT04446117 CONTACT-02: a phase 3, randomized, open-label, controlled study of cabozatinib in combination with atezolizumab vs. second NHT in subjects with mCRPC mCRPC Experimental: cabozatinib+atezolizumab
Comparator: either abiraterone+prednisone or enzalutamide
580 Aug-24
NCT03016312 IMbassador250: a phase 3 study of atezolizumab in combination with enzalutamide in men with mCRPC mCRPC Experimental: atezolizumab+enzalutamide
Comparator: enzalutamide
771 Feb-23

NHT, ovel hormonal therapy; mCRPC, metastatic castration-resistant prostate cancer.

VACCINE-BASED TREATMENTS

Cancer vaccines are a form of active immunotherapy that aims to facilitate the presentation of tumor antigens to the immune system. The proposed prostate cancer's immunosuppressive environment is due in part to the dysfunction of dendritic cells (DCs). Accordingly, DC vaccine therapy has the potential to aid in overcoming the immunosuppressive tumor microenvironment. Currently, sipuleucel-T remains the only vaccine-based therapy with FDA approval for the treatment of prostate cancer. It targets prostatic acid phosphatase and the phase III clinical trial IMPACT (Integrated Model for Patient Care and Clinical Trials) demonstrated that sipuleucel-T had a 22% reduction of relative mortality risk [11]. Since its approval, multiple trials investigating combined immunotherapy have been disappointing [26]. NCT01420965 explored combining pidilizumab, but was terminated early, while NCT01832870 investigated ipilimumab and was terminated without phase I reports. Currently, there is an ongoing phase III trial comparing sipuleucel-T to active surveillance for newly diagnosed localized prostatic cancer (NCT03686683).
No other vaccine treatments have been approved for prostate cancer. VITAL-1 and VITAL-2 were clinical trials with promising results investigating GVAX, a synthesized vaccine using prostate cancer cell lines LNCaP and PC3 secreting granulocyte-macrophage colony-stimulating factor [27]. However, phase III trials were terminated early due to a lack of improved OS and increased mortality.
PROSTVAC is a viral vector containing the PSA gene and several T-cell costimulatory molecules that were used in several clinical trials. Contrary to the promising results in phase II trials, PROSTVAC demonstrated little to no improvement in OS in patients with mCRPC in phase III clinical trials [28-30]. In addition, a phase II trial tested the benefit of using PROSTVAC earlier in the disease compared to active surveillance (NCT02326805) but failed to elicit favorable results [31].

1. Adoptive Cell Therapy

This immunotherapy isolates and manipulates immune cells to increase their capacity to target cancer cells. Chimeric antigen receptor T (CAR T) cell therapy is a classic example and is extremely effective in the treatments of hematologic malignancies due to improved specificity and targeting [32]. CAR T remains a promising treatment option in mCRPC and in other solid tumors, but the safety profile of the treatment remains to be determined [33].

2. Oncolytic Virus Therapy

Oncolytic virus therapy involves utilizing viral vectors to target oncolytic cells and can potentially aid in counteracting tumor-associated immunosuppression and evasion. Conceptually, it is a relatively new mode of therapy as the virus can replicate in situ to have a direct cytocidal effect [34]. Additionally, because the prostate gland is a nonessential organ, oncolytic viral therapy can be used to completely ablate the gland [34]. Oncolytic viral therapy has largely been explored in combination with ICIs [35]. There are several clinical trials exploring this immunotherapy approach and results have been promising but further investigations to clarify the therapeutic potential in prostate cancer are needed.

BARRIERS TO IMMUNOTHERAPY IN PROSTATE CANCER

Despite these diverse immunotherapy modalities, immunotherapy has not been effective in prostate cancer due to its “cold” immune environment. For a robust antitumor immune response, 3 major steps are required: (1) generation of tumor-reactive T cells, (2) physical interaction between target and effector cells, and (3) a microenvironment permissive to immune effector functions [36]. Prostate cancer often lacks distinct characteristics that prevent a clinically meaningful immune response.
Primarily, the TMB of prostate cancer is typically low, resulting in the inefficient generation of tumor-reactive T cells. TMB is increasingly being proposed as a possible predictive marker of response to immunotherapy in various cancers and is associated with high expression levels of neoantigens which correlates with increased immunogenicity [37]. Prostate cancer cells, compared to other cancers successfully treated with immunotherapy such as melanoma, are typically characterized as having low TMB [38,39].
Next, immune effector cells may be hampered from directly contacting cancer cells, as prostate cancer is characterized by T-cell exclusion. Furthermore, T cells in the adjacent stroma and benign areas often demonstrate anergic and immunosuppressive phenotype. This in part may be explained by the chronic progressive nature of prostate cancer and its inflammatory microenvironment [36].
Additional cellular mechanisms proposed for how prostate cancer maintains an immunosuppressive environment include fibroblasts, T-reg cells, tumor-associated macro-phages, mesenchymal stem cells, and myeloid-derived suppressor cells in the tumor stroma. Collectively, these cells secrete mediators that suppress immune activity [40]. Interestingly, as much as 50% of castration-resistant prostate tumors demonstrate phosphatase with tensin homolog (PTEN) inactivation, a tumor suppressor gene. Loss of PTEN function does not affect tumor growth through the loss of its tumor suppressor function but PTEN loss itself can act as an immunosuppressive event, impairing innate and adaptive immunity [41,42]. Additionally, prostate cancer often produces high levels of transforming growth factor-β, which has a profound inhibitory effect on immune cells including natural killer (NK) cells resulting in low NK cell infiltration [43,44]. Finally, tissue aspirate from prostate cancer revealed an increased number of T-reg cells [45]. Taken together, the unfavorable prostate cancer microenvironment is likely a key factor in limiting the efficacy of immunotherapy.
It should be noted that not only the perturbation of immune effector cells and the unfavorable tumor microenvironment pose formidable challenges to developing effective immunotherapeutics in prostate cancer, but factors such as race and previous use of other therapies also add another layer of complexity in prostate cancer immunotherapy. Asian/Pacific islanders have the highest 5-year survival rate in metastatic prostate cancer compared with other races [46], and African Americans respond better to most systemic therapies than Caucasians [47]. ADT can induce T-cell infiltration into the prostate tumor microenvironment [48]. In addition, enzalutamide-resistant prostate cancer expresses higher levels of PD-1 and PD-L1/2 [49]. Therefore, factors such as ethnicity and previous therapies should be considered in developing a rational strategy for immunotherapy in prostate cancer.

PREDICTIVE MARKERS IN IMMUNOTHERAPY

Because immunotherapy is not effective in unselected metastatic prostate cancer patients, predictive biomarkers are needed to identify patients who will likely benefit from immunotherapy.

1. Programmed Death Ligand-1

PD-L1 is used in other tumors as a biomarker for immunotherapy. In prostate cancers, one study reported that the objective response rate to ICI in PD-L1-positive tumors was 5% and in PD-L1-negative tumors, the rate was 3% [10]. Thus, PD-L1 is not considered a viable biomarker of immunotherapy in metastatic prostate cancer.

2. Deficient Mismatch Repair

Deficient MMR genes may result in the overexpression of a variety of immune transcripts including those associated with T cells such as PD-L1 [50]. Underlying dMMR can cause high variation in microsatellite length (MSI-H). Mutations in these genes code for mutant proteins which potentially act as neoantigens that can be recognized by CD8-positive T cells. dMMR/MSI-H tumors showed high response rates and impressive efficacy with ICI treatment [51,52]. However, dMMR/MSI-H is relatively uncommon in prostate cancer and is reported to be present in 2.2%-12% [50,51,53,54].

3. High TMB

High TMB showed a favorable response to ICI compared with taxanes alone in metastatic prostate cancer. Patients with TMB of 10 mt/Mb or greater had a significantly longer time to the next treatment and OS [55].

4. DNA Homologous Recombination Repair Gene

Homologous recombination repair gene (HRR) mutation, especially the CDK12 mutation showed a favorable response to ICI. CDK12-mutated prostate cancer is linked to poor prognosis and resistance to PARP inhibition, but increased neoantigen load for intratumoral lymphocyte infiltration opens the door to PD-1 targeted therapy [56]. However, CDK12 mutations occur in 5%-7% of patients with mCRPC [57]. There is currently an ongoing phase 2 clinical trial of nivolumab and ipilimumab combination therapy for prostate cancer patients with CDK12 mutation (NCT03570619). Collectively, effective prostate cancer immunotherapy will require a panel of biomarkers and genomic determinants to identify patients who will likely respond.

IMMUNOTHERAPY IN RELATION TO ANDROGEN DEPRIVATION THERAPY

ADT forms the basis of systemic therapy for advanced prostate cancer and when we consider integrating immunotherapy for prostate cancer treatment, we must consider the interaction between androgens and the immune system. The immunosuppressive effect of androgens has been observed in both rodent and human studies. Women produce more IgM [58] and there is a strong correlation between androgens and genes involved in lipid metabolism that correlate with low virus-neutralizing antibody in men [59]. Although variable in ratio, autoimmune diseases are more prevalent in women compared to men and men show increased susceptibility to nonreproductive cancers [60].
Accordingly, castration may restore some of the suppressed immune function. In animal studies, castrated male mice which resulted in the change of sex hormone levels induced a change in immune responsiveness [61]. Androgen deprivation showed enhanced T-cell function and resulted in complete regeneration of male mouse thymus with the restoration of peripheral T-cell function [62]. Castration may also enhance CD4-mediated immune responses [63] and castration temporarily leads to prostate Th1-type T-cell infiltration [64]. Moreover, androgen blockade could mitigate T-cell recognition tolerance and induce prostate-specific T-cell proliferation [65]. Similar findings have been found in human studies where an increase in circulating T cells were found in elderly males undergoing sex steroid ablation therapy for prostate cancer [62]. A separate study showed that patients developed expansion of naïve T-cell compartment after androgen deprivation, along with an increase in effector-cell response to stimulation and prostate tissue-associated IgG responses [66]. Based on these observations, immunotherapy should be combined with or sequenced following ADT.
Conversely, there is a body of data suggesting that ADT may suppress the immune response. For example, Jiang et al. [67] investigated the correlation between hepatocellular carcinoma and sex and reported that androgens down-regulate PD-L1. Interestingly, the same authors did not detect the same negative correlation between AR and PD-L1 in prostate cancer. A more provocative result was reported by Pu et al. [68] in 2016. The team reported that chemical castration, but not surgical castration, suppressed T-cell response in prostate cancer. Similarly, our group has observed that androgens stimulate the antitumorigenic activity of macrophages [69]. Specifically, androgen was shown to induce M1 polarization while in an isolated system, removal of macrophages following orchiectomy partially reversed castration resistance. Taken together, these observations suggest that the clinical efficacy of immunotherapy may need to be assessed in the context of ADT. Indeed, we have proposed that the optimal treatment sequence for immunotherapy in prostate cancer may be prior to the initiation of ADT. Clinical trials are being developed to assess this concept.

CONCLUSION

Despite the progress in understanding tumor immunology over the last decade, immunotherapy has not shown a meaningful clinical effect in prostate cancer. Additional studies are necessary to clarify the mechanisms underlying the “immune-coldness” of prostate cancer. Since ADT is the foundation of prostate cancer systemic treatment and androgens affect the immune system, future immunotherapy trials in prostate cancer should include a rational strategy to assess the interaction between ADT and immunotherapy.

NOTES

Conflicts of Interest

The authors have nothing to disclose.

Funding/Support

This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author Contribution

Conceptualization: IYK; Data curation: JK, KL; Formal analysis: IYK, JK, KL; Methodology: IYK; Project administration: IYK; Visualization: IYK; Writing - original draft: JK, KL; Writing - review & editing: IYK, JK, KL.

REFERENCES

1.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7-33.
crossref pmid pdf
2.Ock M, Choi WJ, Jo MW. Trend analysis of major cancer statistics according to sex and severity levels in Korea. PLoS One 2018;13:e0203110.
crossref pmid pmc
3.National Cancer Center. Cancer registration statistics and 2014-2018 cancer incidence statistics by region. Goyang (Korea): National Cancer Center; 2020.
4.Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N, Miller WH Jr. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol 2020;27(Suppl 2):S87-97.
crossref pmid pmc pdf
5.Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol 2017;35:3924-33.
crossref pmid pmc
6.Larkin J, Hodi FS, Wolchok JD. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015;373:1270-1.
crossref
7.Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, et al. Ipilimumab versus placebo after ra-diotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700-12.
pmid pmc
8.Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol 2017;35:40-7.
pmid
9.Powles T, Yuen KC, Gillessen S, Kadel EE 3rd, Rathkopf D, Matsubara N, et al. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat Med 2022;28:144-53.
crossref pmid pmc pdf
10.Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Oja-maa K, Hoimes CJ, 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.
crossref pmid pmc
11.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22.
crossref pmid
12.Mohler JL, Antonarakis ES. NCCN guidelines updates: management of prostate cancer. J Natl Compr Canc Netw 2019;17:583-6.
pmid
13.Chakravarty D, Huang L, Kahn M, Tewari AK. immunotherapy for metastatic prostate cancer: current and emerging treatment options. Urol Clin North Am 2020;47:487-510.
crossref pmid
14.Venkatachalam S, McFarland TR, Agarwal N, Swami U. Immune checkpoint inhibitors in prostate cancer. Cancers (Basel) 2021;13:2187.
crossref pmid pmc
15.Tucker MD, Zhu J, Marin D, Gupta RT, Gupta S, Berry WR, et al. Pembrolizumab in men with heavily treated metastatic castrate-resistant prostate cancer. Cancer Med 2019;8:4644-55.
crossref pmid pmc pdf
16.National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) Prostate Cancer. Plymouth Meeting (PA): National Comprehensive Cancer Network; 2022.
17.Merck announces KEYLYNK-010 trial evaluating KEYTRUDA® (pembrolizumab) in combination with LYNPARZA® (olaparib) in patients with metastatic castration-resistant prostate cancer to stop for futility [Internet]. Rahway (NJ): Merck; 2022;Available from: https://www.merck.com/news/merck-announces-keylynk-010-trial-evaluating-keytruda-pembrolizumab-in-combination-with-lynparza-olaparib-in-patients-with-metastatic-castration-resistant-prostate-cancer-to-stop-for-f/.
18.Merck provides update on phase 3 KEYNOTE-921 trial evaluating KEYTRUDA® (pembrolizumab) plus chemotherapy in patients with metastatic castration-resistant prostate cancer [Internet]. Rahway (NJ): Merck; 2022;Available from: https://www.merck.com/news/merck-provides-update-on-phase-3-keynote-921-trial-evaluating-keytruda-pembrolizumab-plus-chemotherapy-in-patients-with-metastatic-castration-resistant-prostate-cancer/.
19.Merck announces KEYNOTE-991 trial evaluating KEYTRUDA® (pembrolizumab) plus enzalutamide and androgen deprivation therapy in patients with metastatic hor-mone-sensitive prostate cancer to stop for futility [Internet]. Rahway (NJ): Merck; 2022;Available from: https://www.merck.com/news/merck-announces-keynote-991-trial-eval-uating-keytruda-pembrolizumab-plus-enzalutamide-and-androgen-deprivation-therapy-in-patients-with-metastatic-hormone-sensitive-prostate-cancer-to-stop-for/.
20.Yu EY, Kolinsky MP, Berry WR, Retz M, Mourey L, Piulats JM, et al. Pembrolizumab plus docetaxel and prednisone in patients with metastatic castration-resistant prostate cancer: long-term results from the phase 1b/2 KEYNOTE-365 co-hort B study. Eur Urol 2022;82:22-30.
crossref pmid
21.Yu EY, Piulats JM, Gravis G, Fong PCC, Todenhofer T, Laguerre B, et al. Pembrolizumab plus olaparib in patients with metastatic castration-resistant prostate cancer: long-term results from the phase 1b/2 KEYNOTE-365 cohort A study. Eur Urol 2023;83:15-26.
crossref pmid
22.Ren R, Koti M, Hamilton T, Graham CH, Nayak JG, Singh J, et al. A primer on tumour immunology and prostate cancer immunotherapy. Can Urol Assoc J 2016;10:60-5.
crossref pmid pmc pdf
23.Shenderov E, Boudadi K, Fu W, Wang H, Sullivan R, Jor-dan 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;81:326-38.
crossref pmid pmc pdf
24.Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell 2020;38:489-99.e3.
crossref pmid
25.Fizazi K, Gonzalez Mella P, Castellano D, Minatta JN, Rezazadeh Kalebasty A, Shaffer D, et al. Nivolumab plus docetaxel in patients with chemotherapy-naive metastatic castration-resistant prostate cancer: results from the phase II CheckMate 9KD trial. Eur J Cancer 2022;160:61-71.
pmid
26.Cha HR, Lee JH, Ponnazhagan S. Revisiting immunotherapy: a focus on prostate cancer. Cancer Res 2020;80:1615-23.
crossref pmid pmc pdf
27.Arlen PM, Mohebtash M, Madan RA, Gulley JL. Promising novel immunotherapies and combinations for prostate cancer. Future Oncol 2009;5:187-96.
crossref pmid pmc
28.Wang I, Song L, Wang BY, Rezazadeh Kalebasty A, Uchio E, Zi X. Prostate cancer immunotherapy: a review of recent advancements with novel treatment methods and efficacy. Am J Clin Exp Urol 2022;10:210-33.
pmid pmc
29.Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bil-hartz DL, Wyand M, 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;28:1099-105.
pmid pmc
30.Gulley JL, Borre M, Vogelzang NJ, Ng S, Agarwal N, Parker CC, et al. Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J Clin Oncol 2019;37:1051-61.
pmid pmc
31.Parsons JK, Pinto PA, Pavlovich CP, Uchio E, Nguyen MN, Kim HL, et al. A phase 2, double-blind, randomized controlled trial of PROSTVAC in prostate cancer patients on active surveillance. Eur Urol Focus 2022 Dec;12:S2405-4569(22)00286-3.doi: 10.1016/j.euf.2022.12.002 [Epub].
crossref pmid pmc
32.Yu H, Pan J, Guo Z, Yang C, Mao L. CART cell therapy for prostate cancer: status and promise. Onco Targets Ther 2019;12:391-5.
crossref pmid pmc pdf
33.Perera MPJ, Thomas PB, Risbridger GP, Taylor R, Azad A, Hofman MS, et al. Chimeric antigen receptor T-cell therapy in metastatic castrate-resistant prostate cancer. Cancers (Basel) 2022;14:503.
crossref pmid pmc
34.Fukuhara H, Homma Y, Todo T. Oncolytic virus therapy for prostate cancer. Int J Urol 2010;17:20-30.
crossref pmid
35.Lee P, Gujar S. Potentiating prostate cancer immunotherapy with oncolytic viruses. Nat Rev Urol 2018;15:235-50.
crossref pmid pdf
36.Krueger TE, Thorek DLJ, Meeker AK, Isaacs JT, Brennen WN. Tumor-infiltrating mesenchymal stem cells: Drivers of the immunosuppressive tumor microenvironment in prostate cancer? Prostate 2019;79:320-30.
crossref pmid pmc pdf
37.Wang L, Pan S, Zhu B, Yu Z, Wang W. Comprehensive analysis of tumour mutational burden and its clinical sig-nificance in prostate cancer. BMC Urol 2021;21:29.
crossref pmid pmc pdf
38.Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, et al. The genomic complexity of primary human prostate cancer. Nature 2011;470:214-20.
crossref pmid pmc pdf
39.Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239-43.
crossref pmid pmc pdf
40.Stultz J, Fong L. How to turn up the heat on the cold immune microenvironment of metastatic prostate cancer. Prostate Cancer Prostatic Dis 2021;24:697-717.
crossref pmid pmc pdf
41.Chen L, Guo D. The functions of tumor suppressor PTEN in innate and adaptive immunity. Cell Mol Immunol 2017;14:581-9.
crossref pmid pmc pdf
42.Jamaspishvili T, Berman DM, Ross AE, Scher HI, De Marzo AM, Squire JA, et al. Clinical implications of PTEN loss in prostate cancer. Nat Rev Urol 2018;15:222-34.
crossref pmid pmc pdf
43.Pasero C, Gravis G, Guerin M, Granjeaud S, Thomassin-Piana J, Rocchi P, et al. Inherent and tumor-driven immune tolerance in the prostate microenvironment impairs natural killer cell antitumor activity. Cancer Res 2016;76:2153-65.
crossref pmid pdf
44.Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limon P. The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol 2010;10:554-67.
crossref pmid pmc pdf
45.Sfanos KS, Bruno TC, Maris CH, Xu L, Thoburn CJ, De-Marzo AM, et al. Phenotypic analysis of prostate-infiltrating lymphocytes reveals TH17 and Treg skewing. Clin Cancer Res 2008;14:3254-61.
crossref pmid pmc pdf
46.Siegel DA, O'Neil ME, Richards TB, Dowling NF, Weir HK. Prostate cancer incidence and survival, by stage and race/ethnicity - United States, 2001-2017. MMWR Morb Mortal Wkly Rep 2020;69:1473-80.
crossref pmid pmc
47.Bitting RL, Goodman M, George DJ. Racial disparity in response to prostate cancer systemic therapies. Curr Oncol Rep 2020;22:96.
crossref pmid pdf
48.Obradovic AZ, Dallos MC, Zahurak ML, Partin AW, Schaef-fer EM, Ross AE, et al. T-cell infiltration and adaptive treg resistance in response to androgen deprivation with or without vaccination in localized prostate cancer. Clin Cancer Res 2020;26:3182-92.
crossref pmid pmc pdf
49.Bishop JL, Sio A, Angeles A, Roberts ME, Azad AA, Chi KN, et al. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget 2015;6:234-42.
crossref pmid pmc
50.Rodrigues DN, Rescigno P, Liu D, Yuan W, Carreira S, Lambros MB, et al. Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer. J Clin Invest 2018;128:5185.
crossref pmid pmc
51.Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol 2019;5:471-8.
crossref pmid pmc
52.Graham LS, Montgomery B, Cheng HH, Yu EY, Nelson PS, Pritchard C, et al. Mismatch repair deficiency in metastatic prostate cancer: response to PD-1 blockade and standard therapies. PLoS One 2020;15:e0233260.
crossref pmid pmc
53.Pritchard CC, Morrissey C, Kumar A, Zhang X, Smith C, Coleman I, et al. Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat Commun 2014;5:4988.
crossref pmid pmc pdf
54.Antonarakis ES, Shaukat F, Isaacsson Velho P, Kaur H, Shenderov E, Pardoll DM, et al. Clinical features and therapeutic outcomes in men with advanced prostate cancer and DNA mismatch repair gene mutations. Eur Urol 2019;75:378-82.
crossref pmid pmc
55.Graf RP, Fisher V, Weberpals J, Gjoerup O, Tierno MB, Huang RSP, et al. Comparative effectiveness of immune checkpoint inhibitors vs chemotherapy by tumor mutational burden in metastatic castration-resistant prostate cancer. JAMA Netw Open 2022;5:e225394.
crossref pmid pmc
56.Antonarakis ES, Isaacsson Velho P, Fu W, Wang H, Agarwal N, Sacristan Santos V, 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;4:370-81.
crossref pmid pmc
57.Sokol ES, Pavlick D, Frampton GM, Ross JS, Miller VA, Ali SM, et al. Pan-cancer analysis of CDK12 loss-of-function alterations and their association with the focal tandem-duplicator phenotype. Oncologist 2019;24:1526-33.
crossref pmid pmc pdf
58.Butterworth M, McClellan B, Allansmith M. Influence of sex in immunoglobulin levels. Nature 1967;214:1224-5.
crossref pmid pdf
59.Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiebaut R, et al. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc Natl Acad Sci U S A 2014;111:869-74.
crossref pmid pmc
60.Whitacre CC. Sex differences in autoimmune disease. Nat Immunol 2001;2:777-80.
crossref pmid pdf
61.Eidinger D, Garrett TJ. Studies of the regulatory effects of the sex hormones on antibody formation and stem cell differentiation. J Exp Med 1972;136:1098-116.
crossref pmid pmc pdf
62.Sutherland JS, Goldberg GL, Hammett MV, Uldrich AP, Berzins SP, Heng TS, et al. Activation of thymic regeneration in mice and humans following androgen blockade. J Immunol 2005;175:2741-53.
crossref pmid pdf
63.Kissick HT, Sanda MG, Dunn LK, Pellegrini KL, On ST, Noel JK, et al. Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation. Proc Natl Acad Sci U S A 2014;111:9887-92.
crossref pmid pmc
64.Thoma C. Prostate cancer: towards effective combination of ADT and immunotherapy. Nat Rev Urol 2016;13:300.
crossref pdf
65.Drake CG, Doody AD, Mihalyo MA, Huang CT, Kelleher E, Ravi S, et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer Cell 2005;7:239-49.
crossref pmid pmc
66.Morse MD, McNeel DG. Prostate cancer patients on androgen deprivation therapy develop persistent changes in adaptive immune responses. Hum Immunol 2010;71:496-504.
crossref pmid pmc
67.Jiang G, Shi L, Zheng X, Zhang X, Wu K, Liu B, et al. Androgen receptor affects the response to immune checkpoint therapy by suppressing PD-L1 in hepatocellular carcinoma. Aging (Albany NY) 2020;12:11466-84.
crossref pmid pmc
68.Pu Y, Xu M, Liang Y, Yang K, Guo Y, Yang X, et al. androgen receptor antagonists compromise T cell response against prostate cancer leading to early tumor relapse. Sci Transl Med 2016;8:333ra47.
crossref pmid
69.Lee GT, Kim JH, Kwon SJ, Stein MN, Hong JH, Nagaya N, et al. Dihydrotestosterone increases cytotoxic activity of macrophages on prostate cancer cells via TRAIL. Endocrinology 2019;160:2049-60.
crossref pmid pmc pdf


Editorial Office
Department of Urology, Seoul National University Hospital, Seoul National University College of Medicine,
101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
TEL: +82-2-2072-0817,   FAX: +82-2-742-4665   Email: journal@e-juo.org
Korean Urological Oncology Society
50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
Tel: +82-2-704-8574, E-mail: kspe.editor@gmail.com

Copyright © The Korean Urological Oncology Society.

Developed in M2PI