Next Article in Journal
Clinical Rationale of Using Steerable Technologies for Radiofrequency Ablation Followed by Cavity Creation and Cement Augmentation in the Treatment of Painful Spinal Metastases
Next Article in Special Issue
Immune-Checkpoint Induced Skin Toxicity Masked as Squamous Cell Carcinoma: Case Report on Mimickers of Dermatological Toxicity with PD-1 Inhibition
Previous Article in Journal
How We Manage Patients with Indolent B-Cell Malignancies on Bruton’s Tyrosine Kinase Inhibitors: Practical Considerations for Nurses and Pharmacists
Previous Article in Special Issue
Pancreatic Ductal Adenocarcinoma and Immune Checkpoint Inhibitors: The Gray Curtain of Immunotherapy and Spikes of Lights
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond

by
Sree M. Lanka
1,
Nicholas A. Zorko
2,
Emmanuel S. Antonarakis
2 and
Pedro C. Barata
3,*
1
Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA
2
Department of Hematology and Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
3
Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(4), 4246-4256; https://doi.org/10.3390/curroncol30040323
Submission received: 24 March 2023 / Revised: 15 April 2023 / Accepted: 18 April 2023 / Published: 19 April 2023

Abstract

:
The therapeutic landscape of several genitourinary malignancies has been revolutionized by the development of immune checkpoint inhibitors (ICIs); however, the utility of immunotherapies in prostate cancer has been limited, partly due to the immunologically “cold” tumor terrain of prostate cancer. As of today, pembrolizumab is the only immune checkpoint inhibitor approved for the treatment of metastatic castration resistant prostate cancer (mCRPC) in a select group of patients with high microsatellite instability (MSI-H), deficient mismatch repair (dMMR), or high tumor mutational burden (TMB). Looking ahead, several combinatorial approaches with ICIs involving radioligands, radiotherapy, PARP inhibitors, interleukin inhibitors, and cancer vaccines are exploring a potential synergistic effect. Furthermore, B7-H3 is an alternative checkpoint that may hold promise in adding to the treatment landscape of mCRPC. This review aims to summarize previous monotherapy and combination therapy trials of ICIs as well as novel immunotherapy combination therapeutic strategies and treatment targets in mCRPC.

1. Background

As a major public health concern, prostate cancer is a leading cause of cancer worldwide and the second leading cause of cancer death in men in the United States. In 2022, prostate cancer had an incidence of 268,490 and a 5-year-relative survival of 96.8%. However, in patients with distant metastases, the 5-year-relative survival dramatically decreased to 32.3% [1]. Although the treatment strategies for prostate cancer have progressed significantly in recent decades, the expected survival for patients with metastatic castration-resistant prostate cancer (mCRPC) remains poor. Immunotherapies targeting programmed death-1 (PD-1)/programmed death- ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), otherwise known as immune checkpoint inhibitors (ICIs), have revolutionized the therapeutic landscape of genitourinary malignancies such as renal cell carcinoma and urothelial carcinoma. However, they have not yet been shown to be quite as efficacious in mCRPC [2].
As an immunologically “cold” malignancy, prostate cancer may have had limited benefits from ICIs in previous trials due to low tumor burden, low major histocompatibility class I expression, dysfunctional interferon signaling, and a complex tumor microenvironment [3,4]. However, there is an unmet need for the personalization of treatment in mCRPC and although ICI monotherapy is not as efficacious, there may be utility in exploring combination therapies with ICIs and other targeted treatments. The utilization of histologic-agnostic agents remains to be an area that is largely unexplored in prostate cancer, with the exception of pembrolizumab [5]. It is key to take into account the tumor landscape of mCRPC when developing new combination treatments or personalized treatment regimens for patients [6]. To combat the difficult terrain of mCRPC, novel combination treatment strategies and therapeutic targets are being evaluated to advance the use of ICIs in prostate cancer. The goal of this review is to summarize previous and ongoing trials of ICIs in mCRPC and discuss ICI combination strategies and new therapeutic targets for patients with mCRPC.

2. Selection of Trials

In this review, the authors aimed to review the existing literature on monotherapy and combination therapy trials of immune checkpoint inhibitors, specifically in the context of mCRPC. A literature search of existing trials was conducted by searching PubMed for all relevant publications from its inception to 5 April 2023. Titles and abstracts were screened for relevance and full texts of articles were analyzed for eligibility in the review. In addition, breakthrough results of more recent trials that were presented at conferences were also included. There was an emphasis on landmark phase II and III trials that elucidated the extent of the utility of ICIs in mCRPC. In addition, more recent studies that explored ICIs in combination with other agents in mCRPC were also included.

3. Monotherapy: Previous Trials

As of today, pembrolizumab, an ICI targeting PD-1, is the only Food and Drug Administration (FDA) approved ICI for prostate cancer, but it is only specifically approved for mCRPC patients with high microsatellite instability (MSI-H), deficient mismatch repair (dMMR, or tumor mutational burden (TMB) ≥10 mut/Mb. Furthermore, it is recommended only as a subsequent therapy for mCRPC patients who progressed on docetaxel and a novel hormonal therapy [7]. This was extrapolated from a phase II KEYNOTE-158 study that investigated the utility of pembrolizumab in patients with previously treated, advanced non-colorectal MSI-H and dMMR tumors [8]. This study observed objective response in 20 out of the 102 patients with a TMB ≥10 mut/Mb and 43 out of 688 patients with less than 10 mut/Mb. Pembrolizumab was approved for use in the treatment of patients with unresectable or metastatic MSI-H or dMMR solid tumors that had progressed on prior treatment in 2017 [7]. In addition, in a study of a cohort of 65 dMMR mCRPC patients, 19 were treated with anti-PD1 therapy and showed a PSA response rate of 65% and median PFS of 24 weeks [9].
There have been several trials that have investigated ICI monotherapies in mCRPC including pembrolizumab. However, studies have shown that there is a very limited response to single-agent checkpoint inhibitor therapy (Table 1). For instance, in KEYNOTE-199, 258 mCRPC patients who were previously treated with docetaxel and one or more targeted endocrine therapy were given pembrolizumab [10]. In this study, three cohorts of patients were evaluated with pembrolizumab monotherapy. This study also included two cohorts or patients who progressed on enzalutamide, who were given combination pembrolizumab and enzalutamide, an androgen receptor inhibitor. The pembrolizumab monotherapy group had low objective and PSA response rates; however, the patients who did respond to monotherapy tended to have more durable responses. Furthermore, in the enzalutamide and pembrolizumab groups, the overall response rate (ORR) seemed numerically better than the monotherapy group. However, the phase III KEYNOTE-641 trial, which evaluated pembrolizumab in combination with enzalutamide and ADT in mCRPC, was discontinued after an interim analysis showed no improvement in rPFS or OS [11]. In addition, ipilimumab monotherapy was studied in two large phase III trials in chemotherapy-naïve and in docetaxel-pretreated mCRPC patients; however, improved overall survival (OS) was not met in either study [12,13]. Ipilimumab did, however, prolong progression free survival (PFS) and PSA responses in a subset of mCRPC patients. There has also been a phase 1 study that investigated the use of atezolizumab in 35 mCRPC patients who had progressed on sipuleucel-T or enzalutamide, which showed a minimal PSA response rate of 8.6% [14]. Combination with enzalutamide was studied in the IMbassador250 trial [15]. Furthermore, nivolumab was studied in a phase 1 trial, which included patients with melanoma, non-small-cell lung cancer, mCRPC, renal cell carcinoma, and colorectal carcinoma [16]. In the prostate cancer subset of patients, however, there were no objective responses to nivolumab monotherapy.

4. Combination Strategies

There have also been trials that have investigated immunotherapy combination treatments in prostate cancer (Table 2). For instance, in the phase III IMbassador250 trial, atezolizumab plus enzalutamide compared to enzalutamide therapy alone was studied in 759 mCRPC patients. These patients had already progressed on abiraterone and were ineligible for taxane-based therapy [15]. This study, however, did not meet the primary endpoint of improved overall survival in unselected patients. Similar results were found in the phase III KEYNOTE-641 trial, which evaluated pembrolizumab in combination with enzalutamide and ADT in mCRPC, which was discontinued after an interim analysis showed no improvement in rPFS or OS [11]. In another study, the phase II CheckMate 9KD trial, the utility of nivolumab and docetaxel was studied in 41 chemo-naïve mCRPC patients on ongoing androgen deprivation therapy (ADT) [17]. The ORR in patients with measurable disease was 36.8%, whereas the PSA response was 46.3%. It is thought that treatment with immunotherapy may augment the effects of docetaxel. There is now an ongoing phase III clinical trial, CheckMate 7DX to further investigate these results [18]. In cohort B of the KEYNOTE-365 trial, combination therapy with pembrolizumab plus docetaxel and prednisone was studied in chemotherapy-naïve mCRPC patients [19]. Out of the 104 patients who were treated, there was a PSA response rate of 28% with an ORR of 18%. The phase III KEYNOTE-921 trial evaluated pembrolizumab and docetaxel and prednisone in chemotherapy-naïve mCRPC patients [20]. The results of the KEYNOTE-921 trial were presented at the GU ASCO 2023 meeting, showing that the primary endpoints of rPFS and OS were not met [21]. There have also been trials to investigate dual immune-checkpoint inhibitor therapy including the phase II CheckMate 650 trial, which investigated the utility of ipilimumab and nivolumab in mCRPC patients who were previously treated with docetaxel [22]. It was shown that particularly in patients with high tumor mutational burden (TMB), ipilimumab and nivolumab combination therapy had clinical activity; however, treatment discontinuation occurred due to early toxicity. Additional results presented at ASCO GU 2023 included newly enrolled patients with an alternative ipilimumab and nivolumab regimen versus ipilimumab alone versus cabazitaxel. Several patients in the ipilimumab and nivolumab combination cohorts had a reduction (75–100%) in tumor size and PSA [23].

5. Novel Immunotherapeutic Targets beyond ICI

It is important to investigate whether the combination of immunotherapy with various other treatments could achieve synergistic effects in patients with prostate cancer (Table 3). To do this, it is essential to take a closer look at the complex tumor microenvironment in prostate cancer and identify other therapeutic targets that can be used alongside ICIs [24]. For instance, cytokines, specifically interleukins (ILs) such as IL-6, IL-8, IL-15, and IL-23, play a major role in the proliferation of cancer. Specifically in prostate cancer, IL-6 has been shown to be involved in radiotherapy resistance [25]. Although there have been several pre-clinical studies on therapies targeting cytokines in prostate cancer, there have not been many successful studies that have led to clinical outcomes [26]. In a phase II trial, siltuximab, a monoclonal antibody against IL-6, was evaluated in mCRPC patients who were previously treated with chemotherapy; however, there was only a 3.8% PSA response rate, and the results were largely disappointing [27]. There have not been many dedicated trials that have investigated combination regimens with interleukin inhibitors.
Furthermore, cancer vaccines such as sipuleucel-T were the initial backbone of prostate cancer management. There have been studies that have investigated the utility of sipuleucel-T with ICIs, for instance, the phase Ib study that studied a combination of sipuleucel-T with atezolizumab in 37 asymptomatic or minimally symptomatic mCRPC patients [28]. Of the 23 patients with measurable disease, only 4.3% had an objective response. Although the combination was well-tolerated, further studies are needed in larger cohorts to determine whether the combination is truly beneficial.
In addition, poly-ADP ribose polymerase (PARP) inhibitors, which work by interfering with DNA damage repair mechanisms, could be combined with ICIs. Even though there are studies that have investigated PARP inhibitor treatment such as olaparib and rucaparib, few have investigated their synergistic effects with ICIs [29]. However, cohort A of the KEYNOTE 365 study investigated pembrolizumab plus olaparib in 102 docetaxel-pretreated mCRPC patients with disease progression [30]. A total of 59 patients had measurable disease, with an ORR of 8.5%. Radiographic progression free survival (rPFS) was 4.5 months and the median OS was 14 months. Additionally, there is the KEYLYNK-010 phase III study, in which the pembrolizumab plus olaparib combination is being compared to enzalutamide monotherapy or abiraterone monotherapy in enzalutamide or abiraterone pretreated mCRPC patients who progressed on chemotherapy [31]. The primary endpoints being measured are overall survival and rPFS. The results of KEYLYNK-010 were presented at the European Society for Medical Oncology Congress in 2022, and it was shown that the primary endpoints of rPFS and OS were not met [32]. Furthermore, the phase II trial that investigated durvalumab, a PD-L1 inhibitor, and olaparib in mCRPC, showed a PSA response of more than 50% in 47% of patients [33]. This was especially notable in patients with DNA damage repair mutations, alluding that patients with these mutations may benefit further with a combination of immunotherapy and PARP inhibitor treatment.
Furthermore, although there have been a few studies looking into ICIs and radioligand therapy combinations, it may be worthwhile exploring this option as radioligand therapies could sensitize immunologically “cold” prostate tumors to ICIs [34]. The phase 1b/II PRINCE trial of pembrolizumab in combination with 177-Lu-PSMA-617 in mCRPC patients indicated a PSA response rate of 76% and an ORR of 78%. In addition, there is an ongoing phase II EVOLUTION trial introduced at ASCO GU 2023 that is investigating ipilimumab and nivolumab in combination with 177-Lu-PSMA-617. The primary endpoint is 12-month PSA progression-free survival. There are ongoing translational studies looking into the tumor microenvironment effects and predictors of response to this combination therapy [35]. There are further preclinical studies investigating the utility of actinium in addition to ICIs that are currently underway [36]. For instance, in the murine model study by Czernin et al., mice were treated with 225Ac-PSMA617, an anti-PD-1 antibody, or both. It was shown that combination therapy improved the time to progression and survival compared to monotherapy alone [36]. Finally, there have been many preclinical and phase II studies that have shown that radiotherapy in combination with ICIs can result in tumor regression. However, in subsequent phase III trials, there was no significant difference between the ICI plus radiotherapy. In a study that evaluated atezolizumab and radium-223 in mCRPC patients with bone and lymph node and/or visceral metastases, there was no clear clinical benefit of combination therapy [37]. In addition, the combination regimen had greater toxicity compared to either drug alone.

6. B7-H3 as an Alternative Immune Checkpoint in Prostate Cancer

In addition to exploring combination strategies with classical immune checkpoints (PD-1, PD-L1, CTLA4), an alternative approach is to validate additional checkpoints as potential therapeutic targets in prostate cancer. To this end, the B7 superfamily molecule B7-H3 (also known as PD-L3, or CD276) has emerged as a new target in prostate cancer [38]. Relative to PD-L1 and PD-L2, B7-H3 is expressed at much higher levels in prostate cancer [39] and is found in >80% of primary and castration-resistant tumor specimens [40]. While B7-H3 was initially proposed to be immunostimulatory, accumulated evidence supports its negative regulatory role for immune response [41,42]. Consistent with these findings, high B7-H3 expression is associated with lower CD3+ T cell density and higher Treg density and is inversely correlated with Black race (expression is higher in Caucasian patients relative to African ancestry patients) [43]. Interestingly, B7-H3 expression is strongly correlated with AR signaling and AR cofactors (FOXA1, HOXB13), and mechanistic studies have suggested that the B7-H3 promoter and distal enhancer regions are directly bound by AR and its co-factors, implying that B7-H3 expression is under AR transcriptional control [44]. In a recent neoadjuvant study using the B7-H3 targeting monoclonal antibody, enoblituzumab, in 32 patients with high-risk localized prostate cancer, B7-H3 inhibition produced several PSA reductions and Gleason score reductions while inducing the upregulation of CD8+ T cells, adaptive increases in PD-1/PD-L1, and the evidence of immune activation (elevated granzyme B, IFN-gamma signaling, and myeloid inflammation) [45]. Larger randomized adjuvant studies of enoblituzumab in high-risk prostate cancer are currently being designed. Finally, given its high expression across prostate cancer treatment states, B7-H3 is also being pursued by several groups as an antigen target without the goal of interrupting its immunoregulatory roles. Multiple strategies including antibody-drug conjugates (DS-7300 and MGC018), Tri-Specific NK cell engagers (TriKE), and chimeric antigen receptor CAR-T and CAR-NK cells are in various states of pre-clinical development and early phase clinical trials [46,47,48,49,50]. While not functioning via the typical mechanism for immune checkpoint inhibition by focusing on blocking the inhibitory role of immune checkpoint molecules, these strategies may provide a novel alternative for exploiting the high levels of B7-H3 expression in prostate cancer.

7. Discussion

Although monotherapy ICI trials in mCRPC have not been very promising, there are many trials that have combined ICIs with standard chemotherapy as well as targeted therapies. There is an unmet need for the personalization of treatment in mCRPC, and ICI combination therapies may play a role in targeted therapy for mCRPC patients. Except for pembrolizumab, tumor-agnostic drugs have not otherwise been approved in prostate cancer [6]. Utilizing other targets in the signaling pathway of the prostate cancer microenvironment can result in a synergistic effect with ICIs. For instance, as discussed, combining ICIs with chemotherapy, radioligands, radiotherapy, PARP inhibitors, interleukin inhibitors, and cancer vaccines holds promise in the future of mCRPC treatment. Not only that, but exploring different checkpoint targets other than the conventional PD-1, PD-L1, and CTLA-4 can create a novel treatment strategy that may benefit patients. Further large-cohort studies must be conducted on various combinations of ICIs to determine which patients benefit the most from different treatment targets.

8. Conclusions

In conclusion, even though immune checkpoint inhibitors have revolutionized the treatment landscape of many malignancies, they tended to have a lag in utility in mCRPC. In the setting of treatment resistance and immunologically “cold” characteristics of prostate cancer, it is important to delve further into the tumor microenvironment and identify additional targets for therapy and for combination regimens that sensitize tumors to ICIs. There have been several trials that have studied the efficacy and utility of ICIs in prostate cancer; however, few clinical trials have approved combination therapies. Certain combinatorial approaches with ICIs involving radioligands, radiotherapy, PARP inhibitors, interleukin inhibitors, and cancer vaccines could create a synergistic effect and may be the future of mCRPC therapy. In addition, alternative checkpoints (such as B7-H3) may hold more promise as better therapeutic targets in this disease.

Author Contributions

Conceptualization, S.M.L. and P.C.B.; methodology, all authors; validation, all authors; formal analysis, all authors; writing—original draft preparation, all authors; writing—review and editing, all authors; supervision, P.C.B. 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.

Data Availability Statement

Not applicable.

Conflicts of Interest

N.A.Z. is an unpaid consultant for Caris Life Sciences and supported by a Prostate Cancer Foundation Young Investigator Award and a Department of Defense Early Investigator Award W81XWH-22-1-0242. E.S.A. has served as a paid consultant/advisor for Sanofi, Dendreon, Janssen Biotech, Merck, AstraZeneca, Clovis Oncology, Pfizer, Curium, Lilly, and Bayer; has received research funding from Janssen Biotech, Johnson & Johnson, Sanofi, Dendreon, Aragon Pharmaceuticals, Exelixis, Millennium, Genentech, Novartis, Astellas Pharma, Tokai Pharmaceuticals, Merck, AstraZeneca, Clovis Oncology and Constellation Pharmaceuticals; and is a co-inventor of a biomarker technology owned by Johns Hopkins University and licensed to Qiagen. P.C.B. has received consultant fees from Astellas, Eisai, AVEO Oncology, Janssen, EMD Serono; Dendreon; Pfizer, Seattle Genetics, BMS, Bayer, Guardant Health, Caris Life Sciences and Myovant (unrelated to this work).

References

  1. Seer Cancer Statistics. Available online: https://seer.cancer.gov/statfacts/html/prost.html (accessed on 18 March 2023).
  2. Mitsogiannis, I.; Tzelves, L.; Dellis, A.; Issa, H.; Papatsoris, A.; Moussa, M. Prostate Cancer Immunotherapy. Expert Opin. Biol. Ther. 2022, 22, 577–590. [Google Scholar] [CrossRef]
  3. Claps, M.; Mennitto, A.; Guadalupi, V.; Sepe, P.; Stellato, M.; Zattarin, E.; Gillessen, S.S.; Sternberg, C.N.; Berruti, A.; De Braud, F.G.M.; et al. Immune-Checkpoint Inhibitors and Metastatic Prostate Cancer Therapy: Learning by Making Mistakes. Cancer Treat Rev. 2020, 88, 102057. [Google Scholar] [CrossRef]
  4. Venkatachalam, S.; McFarland, T.R.; Agarwal, N.; Swami, U. Immune Checkpoint Inhibitors in Prostate Cancer. Cancers 2021, 13, 2187. [Google Scholar] [CrossRef]
  5. Iannantuono, G.M.; Torino, F.; Rosenfeld, R.; Guerriero, S.; Carlucci, M.; Sganga, S.; Capotondi, B.; Riondino, S.; Roselli, M. The Role of Histology-Agnostic Drugs in the Treatment of Metastatic Castration-Resistant Prostate Cancer. Int. J. Mol. Sci. 2022, 23, 8535. [Google Scholar] [CrossRef]
  6. Pestana, R.C.; Sen, S.; Hobbs, B.P.; Hong, D.S. Histology-Agnostic Drug Development—Considering Issues beyond the Tissue. Nat. Rev. Clin. Oncol. 2020, 17, 555–568. [Google Scholar] [CrossRef] [PubMed]
  7. National Comprehensive Cancer Network Guideline Version 1.2023 Prostate Cancer. Available online: https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf (accessed on 18 March 2023).
  8. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.-P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results from the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  9. Sena, L.A.; Fountain, J.; Isaacsson Velho, P.; Lim, S.J.; Wang, H.; Nizialek, E.; Rathi, N.; Nussenzveig, R.; Maughan, B.L.; Velez, M.G.; et al. Tumor Frameshift Mutation Proportion Predicts Response to Immunotherapy in Mismatch Repair-Deficient Prostate Cancer. Oncologist 2021, 26, e270–e278. [Google Scholar] [CrossRef] [PubMed]
  10. 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]
  11. Merck News Release. Available online: https://www.merck.com/news/merck-provides-update-on-phase-3-trials-keynote-641-and-keynote-789/ (accessed on 18 March 2023).
  12. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; van den Eertwegh, A.J.M.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; et al. Ipilimumab versus Placebo after Radiotherapy in Patients with Metastatic Castration-Resistant Prostate Cancer That Had Progressed after Docetaxel Chemotherapy (CA184-043): A Multicentre, Randomised, Double-Blind, Phase 3 Trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef]
  13. 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]
  14. Petrylak, D.P.; Loriot, Y.; Shaffer, D.R.; 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, 27, 3360–3369. [Google Scholar] [CrossRef]
  15. Powles, T.; Yuen, K.C.; Gillessen, S.; Kadel, E.E.; Rathkopf, D.; Matsubara, N.; Drake, C.G.; Fizazi, K.; Piulats, J.M.; Wysocki, P.J.; et al. Atezolizumab with Enzalutamide versus Enzalutamide Alone in Metastatic Castration-Resistant Prostate Cancer: A Randomized Phase 3 Trial. Nat. Med. 2022, 28, 144–153. [Google Scholar] [CrossRef]
  16. 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]
  17. Fizazi, K.; González Mella, P.; Castellano, D.; Minatta, J.N.; Rezazadeh Kalebasty, A.; Shaffer, D.; Vázquez Limón, J.C.; Sánchez López, H.M.; Armstrong, A.J.; Horvath, L.; et al. Nivolumab plus Docetaxel in Patients with Chemotherapy-Naïve Metastatic Castration-Resistant Prostate Cancer: Results from the Phase II CheckMate 9KD Trial. Eur. J. Cancer 2022, 160, 61–71. [Google Scholar] [CrossRef]
  18. Drake, C.G.; Saad, F.; Clark, W.R.; Ciprotti, M.; Sharkey, B.; Subudhi, S.K.; Fizazi, K. 690TiP A Phase III, Randomized, Double-Blind Trial of Nivolumab or Placebo Combined with Docetaxel for Metastatic Castration-Resistant Prostate Cancer (MCRPC; CheckMate 7DX). Ann. Oncol. 2020, 31, S546. [Google Scholar] [CrossRef]
  19. Yu, E.Y.; Kolinsky, M.P.; Berry, W.R.; Retz, M.; Mourey, L.; Piulats, J.M.; Appleman, L.J.; Romano, E.; Gravis, G.; Gurney, H.; 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 Cohort B Study. Eur. Urol. 2022, 82, 22–30. [Google Scholar] [CrossRef]
  20. Petrylak, D.P.; Li, B.; Schloss, C.; Fizazi, K. KEYNOTE-921: Phase III Study of Pembrolizumab (Pembro) plus Docetaxel and Prednisone for Enzalutamide (Enza)- or Abiraterone (Abi)-Pretreated Patients (Pts) with Metastatic Castration-Resistant Prostate Cancer (MCRPC). Ann. Oncol. 2019, 30, v351. [Google Scholar] [CrossRef]
  21. Petrylak, D.P.; Ratta, R.; Matsubara, N.; Korbenfeld, E.P.; Gafanov, R.; Mourey, L.; Todenhöfer, T.; Gurney, H.; Kramer, G.; Bergman, A.M.; et al. Pembrolizumab plus Docetaxel for Patients with Metastatic Castration-Resistant Prostate Cancer (MCRPC): Randomized, Double-Blind, Phase 3 KEYNOTE-921 Study. JCO 2023, 41 (Suppl. S6), 19. [Google Scholar] [CrossRef]
  22. Sharma, P.; Pachynski, R.K.; Narayan, V.; Fléchon, A.; Gravis, G.; Galsky, M.D.; Mahammedi, H.; Patnaik, A.; Subudhi, S.K.; Ciprotti, M.; et al. Nivolumab Plus Ipilimumab for Metastatic Castration-Resistant Prostate Cancer: Preliminary Analysis of Patients in the CheckMate 650 Trial. Cancer Cell 2020, 38, 489–499.e3. [Google Scholar] [CrossRef]
  23. Sharma, P.; Krainer, M.; Saad, F.; Castellano, D.; Bedke, J.; Kwiatkowski, M.; Patnaik, A.; Procopio, G.; Wiechno, P.; Kochuparambil, S.T.; et al. Nivolumab plus Ipilimumab for the Treatment of Post-Chemotherapy Metastatic Castration-Resistant Prostate Cancer (MCRPC): Additional Results from the Randomized Phase 2 CheckMate 650 Trial. JCO 2023, 41 (Suppl. S6), 22. [Google Scholar] [CrossRef]
  24. Heidegger, I.; Necchi, A.; Pircher, A.; Tsaur, I.; Marra, G.; Kasivisvanathan, V.; Kretschmer, A.; Mathieu, R.; Ceci, F.; van den Bergh, R.C.N.; et al. A Systematic Review of the Emerging Role of Immune Checkpoint Inhibitors in Metastatic Castration-Resistant Prostate Cancer: Will Combination Strategies Improve Efficacy? Eur. Urol. Oncol. 2021, 4, 745–754. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, X.; Chen, F.; Ren, Y.; Weng, G.; Xu, L.; Xue, X.; Keng, P.C.; Lee, S.O.; Chen, Y. IL-6 Signaling Contributes to Radioresistance of Prostate Cancer through Key DNA Repair-Associated Molecules ATM, ATR, and BRCA 1/2. J. Cancer Res. Clin. Oncol. 2019, 145, 1471–1484. [Google Scholar] [CrossRef] [PubMed]
  26. Mughees, M.; Kaushal, J.B.; Sharma, G.; Wajid, S.; Batra, S.K.; Siddiqui, J.A. Chemokines and Cytokines: Axis and Allies in Prostate Cancer Pathogenesis. Semin. Cancer Biol. 2022, 86 Pt 3, 497–512. [Google Scholar] [CrossRef] [PubMed]
  27. Dorff, T.B.; Goldman, B.; Pinski, J.K.; Mack, P.C.; Lara, P.N.; Van Veldhuizen, P.J.; Quinn, D.I.; Vogelzang, N.J.; Thompson, I.M.; Hussain, M.H.A. Clinical and Correlative Results of SWOG S0354: A Phase II Trial of CNTO328 (Siltuximab), a Monoclonal Antibody against Interleukin-6, in Chemotherapy-Pretreated Patients with Castration-Resistant Prostate Cancer. Clin Cancer Res. 2010, 16, 3028–3034. [Google Scholar] [CrossRef]
  28. Dorff, T.; Hirasawa, Y.; Acoba, J.; Pagano, I.; Tamura, D.; Pal, S.; Zhang, M.; Waitz, R.; Dhal, A.; Haynes, W.; et al. Phase Ib Study of Patients with Metastatic Castrate-Resistant Prostate Cancer Treated with Different Sequencing Regimens of Atezolizumab and Sipuleucel-T. J. Immunother. Cancer 2021, 9, e002931. [Google Scholar] [CrossRef]
  29. 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]
  30. Yu, E.Y.; Piulats, J.M.; Gravis, G.; Fong, P.C.C.; Todenhöfer, T.; Laguerre, B.; Arranz, J.A.; Oudard, S.; Massard, C.; Heinzelbecker, J.; 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. [Google Scholar] [CrossRef]
  31. LeVee, A.; Lin, C.Y.; Posadas, E.; Figlin, R.; Bhowmick, N.A.; Di Vizio, D.; Ellis, L.; Rosser, C.J.; Freeman, M.R.; Theodorescu, D.; et al. Clinical Utility of Olaparib in the Treatment of Metastatic Castration-Resistant Prostate Cancer: A Review of Current Evidence and Patient Selection. Onco Targets Ther. 2021, 14, 4819–4832. [Google Scholar] [CrossRef]
  32. ESMO Congress 2022 OncologyPro. Available online: https://oncologypro.esmo.org/meeting-resources/esmo-congress/pembrolizumab-olaparib-vs-abiraterone-abi-or-enzalutamide-enza-for-patients-pts-with-previously-treated-metastatic-castration-resistant-pro. (accessed on 18 March 2023).
  33. Karzai, F.; VanderWeele, D.; Madan, R.A.; Owens, H.; Cordes, L.M.; Hankin, A.; Couvillon, A.; Nichols, E.; Bilusic, M.; Beshiri, M.L.; et al. Activity of Durvalumab plus Olaparib in Metastatic Castration-Resistant Prostate Cancer in Men with and without DNA Damage Repair Mutations. J. Immunother. Cancer 2018, 6, 141. [Google Scholar] [CrossRef]
  34. Bellavia, M.C.; Patel, R.B.; Anderson, C.J. Combined Targeted Radiopharmaceutical Therapy and Immune Checkpoint Blockade: From Preclinical Advances to the Clinic. J. Nucl. Med. 2022, 63, 1636–1641. [Google Scholar] [CrossRef]
  35. Sandhu, S.; Subramaniam, S.; Hofman, M.S.; Stockler, M.R.; Martin, A.J.; Pokorski, I.; Goh, J.C.; Pattison, D.A.; Dhiantravan, N.; Gedye, C.; et al. Evolution: Phase II Study of Radionuclide 177 Lu-PSMA-617 Therapy versus 177 Lu-PSMA-617 in Combination with Ipilimumab and Nivolumab for Men with Metastatic Castration-Resistant Prostate Cancer (MCRPC; ANZUP 2001). JCO 2023, 41 (Suppl. S6), TPS271. [Google Scholar] [CrossRef]
  36. Czernin, J.; Current, K.; Mona, C.E.; Nyiranshuti, L.; Hikmat, F.; Radu, C.G.; Lückerath, K. Immune-Checkpoint Blockade Enhances 225Ac-PSMA617 Efficacy in a Mouse Model of Prostate Cancer. J. Nucl. Med. 2021, 62, 228–231. [Google Scholar] [CrossRef] [PubMed]
  37. Fong, L.; Morris, M.J.; Sartor, O.; Higano, C.S.; Pagliaro, L.; Alva, A.; Appleman, L.J.; Tan, W.; Vaishampayan, U.; Porcu, R.; et al. A Phase Ib Study of Atezolizumab with Radium-223 Dichloride in Men with Metastatic Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2021, 27, 4746–4756. [Google Scholar] [CrossRef]
  38. Shenderov, E.; Antonarakis, E.S. B7-H3 and Prostate Cancer: New Therapeutic Dance Partners. Eur. Urol. 2023, 83, 239–240. [Google Scholar] [CrossRef] [PubMed]
  39. 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]
  40. Guo, C.; Figueiredo, I.; Gurel, B.; Neeb, A.; Seed, G.; Crespo, M.; Carreira, S.; Rekowski, J.; Buroni, L.; Welti, J.; et al. B7-H3 as a Therapeutic Target in Advanced Prostate Cancer. Eur. Urol. 2023, 83, 224–238. [Google Scholar] [CrossRef]
  41. 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-γ Production. Nat. Immunol. 2001, 2, 269–274. [Google Scholar] [CrossRef]
  42. Kontos, F.; Michelakos, T.; Kurokawa, T.; Sadagopan, A.; Schwab, J.H.; Ferrone, C.R.; Ferrone, S. B7-H3: An Attractive Target for Antibody-Based Immunotherapy. Clin. Cancer Res. 2021, 27, 1227–1235. [Google Scholar] [CrossRef]
  43. Mendes, A.A.; Lu, J.; Kaur, H.B.; Zheng, S.L.; Xu, J.; Hicks, J.; Weiner, A.B.; Schaeffer, E.M.; Ross, A.E.; Balk, S.P.; et al. Association of B7-H3 Expression with Racial Ancestry, Immune Cell Density, and Androgen Receptor Activation in Prostate Cancer. Cancer 2022, 128, 2269–2280. [Google Scholar] [CrossRef]
  44. Shi, X.; Day, A.; Bergom, H.E.; Tape, S.; Baca, S.C.; Sychev, Z.E.; Larson, G.; Bozicevich, A.; Drake, J.M.; Zorko, N.; et al. Integrative Molecular Analyses Define Correlates of High B7-H3 Expression in Metastatic Castrate-Resistant Prostate Cancer. npj Precis. Onc. 2022, 6, 80. [Google Scholar] [CrossRef]
  45. Shenderov, E.; De Marzo, A.M.; Lotan, T.L.; Wang, H.; Chan, S.; Lim, S.J.; Ji, H.; Allaf, M.E.; Chapman, C.; Moore, P.A.; et al. Neoadjuvant enoblituzumab in localized prostate cancer: A single-arm, phase 2 trial. Nat. Med. 2023, 1–10. [Google Scholar] [CrossRef]
  46. Doi, T.; Patel, M.; Falchook, G.S.; Koyama, T.; Friedman, C.F.; Piha-Paul, S.; Gutierrez, M.E.; Abdul-Karim, R.; Awad, M.; Adkins, D.R.; et al. 453O DS-7300 (B7-H3 DXd Antibody-Drug Conjugate [ADC]) Shows Durable Antitumor Activity in Advanced Solid Tumors: Extended Follow-Up of a Phase I/II Study. Ann. Oncol. 2022, 33, S744–S745. [Google Scholar] [CrossRef]
  47. Jang, S.; Powderly, J.D.; Spira, A.I.; Bakkacha, O.; Loo, D.; Bohac, G.C.; Sharma, M. Phase 1 Dose Escalation Study of MGC018, an Anti-B7-H3 Antibody-Drug Conjugate (ADC), in Patients with Advanced Solid Tumors. JCO 2021, 39 (Suppl. S15), 2631. [Google Scholar] [CrossRef]
  48. Miller, J.S.; Zorko, N.; Merino, A.; Phung, G.; Khaw, M.; Howard, P.; Hamsher, H.; Davis, Z.; Cichocki, F.; Berk, G.I.; et al. 755P B7H3-Targeted Tri-Specific Killer Engagers Deliver IL-15 to NK Cells but Not T-Cells, and Specifically Target Solid Tumors as a Pan-Tumor Antigen Strategy Mediated through NK Cells. Ann. Oncol. 2022, 33, S889. [Google Scholar] [CrossRef]
  49. Zhang, Y.; He, L.; Sadagopan, A.; Ma, T.; Dotti, G.; Wang, Y.; Zheng, H.; Gao, X.; Wang, D.; DeLeo, A.B.; et al. Targeting Radiation-Resistant Prostate Cancer Stem Cells by B7-H3 CAR T Cells. Mol. Cancer Ther. 2021, 20, 577–588. [Google Scholar] [CrossRef]
  50. Zorko, N.; Cichocki, F.; Goulding, J.; Hancock, B.; Blum, R.; Pribadi, M.; Gaertner, B.; Lee, T.; Felices, M.; Bjordahl, R.; et al. Abstract 2761: FT573: Preclinical Development of Multiplexed-Engineered IPSC-Derived NK Cells Expressing a Novel Camelid Nanobody Chimeric Antigen Receptor (CAR) Targeting Pan-Cancer Antigen B7-H3. Cancer Res. 2022, 82 (Suppl. S12), 2761. [Google Scholar] [CrossRef]
Table 1. Immune checkpoint inhibitor monotherapy evidence in mCRPC.
Table 1. Immune checkpoint inhibitor monotherapy evidence in mCRPC.
Trial Name/NCT PhasePatient PopulationTreatmentPrimary EndpointResults
CA184-095 (NCT01057810)IIIChemotherapy-naïve patients with mCRPC without visceral metastases Ipilimumab monotherapy OS28.7 months (95% CI, 24.5 to 32.5 months) in the ipilimumab arm versus 29.7 months (95% CI, 26.1 to 34.2 months) in the placebo arm (hazard ratio, 1.11; 95.87% CI, 0.88 to 1.39; p = 0.3667)
KEYNOTE-199 (NCT02787005)IIPretreated mCRPC patients in 3 cohorts (cohort 1 PD-L1 + disease, cohort 2 PD-L1 negative disease, cohort 3 bone predominant disease regardless of PD-L1 expression)Pembrolizumab monotherapyORR 5% (95% CI, 2% to 11%) in cohort 1 and 3% (95% CI, <1% to 11%) in cohort 2
CA184-043 (NCT00861614)IIIDocetaxel-pretreated mCRPC patients Ipilimumab vs. placebo after radiotherapy OS11.2 months (95% CI 9.5–12.7) for ipilimumab and 10.0 months (95% CI 8.3–11.0) for placebo (HR 0.85, 95% CI 0.71–1.00; p = 0.053)
CA184-095 (NCT01057810)IIIAsymptomatic or minimally symptomatic chemo-naïve patients with mCRPCIpilimumab vs. placebo OS28.7 months (95% CI, 24.5 to 32.5) for ipilimumab and 29.7 (95% CI, 26.1 to 34.2) in placebo (HR 1.11; 95% CI 0.88 to 1.39; p = 0.3667)
PCD4989g (NCT01375842)IbmCRPC patients who have progressed on sipuleucel-T or enzalutamideAtezolizumab monotherapySafety and tolerability of atezolizumabTreatment- related adverse events in 60% of patients
Abbreviations: CI confidence interval; HR hazard ratio; mCRPC metastatic castration-resistant prostate cancer; ORR objective response rate; OS overall survival; PD-L1 programmed death-1 ligand 1.
Table 2. Immune checkpoint inhibitor combination therapy evidence in mCRPC.
Table 2. Immune checkpoint inhibitor combination therapy evidence in mCRPC.
Trail Name/NCTPhasePatient PopulationTreatmentPrimary EndpointResults
IMbassador250 (NCT03016312)IIImCRPC patients who had progressed on abirateroneAtezolizumab + enzalutamide vs. enzalutamide aloneOSStopped early due to low probability of trial achieving primary endpoint given risk of immune-mediated adverse events
KEYNOTE-641 (NCT03834493)IIIChemo-naïve mCRPC patients who are abiraterone-naïve or are intolerant to or progressed on abirateronePembrolizumab + enzalutamide vs. placebo + enzalutamide OS, rPFSDiscontinued after an interim analysis showed no improvement in rPFS or OS
CheckMate 9KD (NCT03338790)IIChemo-naïve mCRPC patients with ongoing ADT and ≤2 prior novel hormonal therapiesNivolumab and docetaxel with prednisone and then nivolumab ORR, PSA response rateConfirmed ORR (95% CI) was 40.0% (25.7–55.7), and the confirmed PSA50-RR (95% CI) was 46.9% (35.7–58.3)
CheckMate 7DX (NCT04100018)IIIChemo-naïve mCRPC patients with ongoing ADT and ≤2 prior novel hormonal therapiesNivolumab + docetaxel vs. placebo + docetaxelrPFS, OS Pending
KEYNOTE-365 (NCT02861573) Cohort B1b/IIChemo-naïve mCRPC patients who progressed on 4 weeks or more of abiraterone or enzalutamidePembrolizumab + docetaxel + prednisoneSafety, PSA response rate, ORRConfirmed PSA response rate was 34% and the confirmed ORR was 23%. TRAEs occurred in 100 patients (96%). Grade 3–5 TRAEs occurred in 46 patients (44%). Seven AE-related deaths (6.7%) occurred (2 due to treatment-related pneumonitis)
KEYNOTE-921 (NCT03834506)IIIChemo-naïve mCRPC patients who progressed on 4 weeks or more of abiraterone or enzalutamidePembrolizumab + docetaxel vs.
docetaxel alone
OS, rPFSResults presented at ASCO GU 2023 Conference: rPFS (median 8.6 mo with pembrolizumab + docetaxel vs. 8.3 mo with placebo + docetaxel; HR 0.85, 95% CI 0.7121.01; p = 0.0335) and OS (median 19.6 months vs. 19.0 months; HR 0.92, 95% CI 0.7821.09; p = 0.1677) were not met
CheckMate 650 (NCT02985957)IIAsymptomatic/minimally symptomatic patients who progressed after 2nd-generation hormone therapy and have not received chemotherapy for mCRPC (cohort 1) and patients who progressed after taxane-based chemotherapy (cohort 2) Ipilimumab + nivolumab ORR; rPFSMedian rPFS (95% CI) in all treated patients was 5.5 (3.5–7.1) and 3.8 months (2.1–5.1) in cohorts 1 and 2. In patients with TMB above vs. below the median, the ORR was 50.0% (95% CI 26.0–74.0) vs. 5.3% (95% CI 0.1–26.0)
CheckMate 650 (NCT02985957) additional resultsIImCRPC patients previously treated with docetaxelNivolumab + ipilimumab q3weeks for 4 doses then nivolumab q4weeks (cohort 1) vs. nivolumab q3weeks for 8 doses and ipilimumab q6w for 4 doses then nivolumab q4weeks (cohort 2) vs. ipilimumab alone (cohort 3) vs. cabaziaxel (cohort 4) ORR, PSA response rate, rPFSORR 9% (cohort 1) vs. 15% (cohort 2) vs. 4% (cohort 3) vs. 11% (cohort 4). PSA response rate 14% (cohort 1) vs. 18% (cohort 2) vs. 5% (cohort 3) vs. 24% (cohort 4)
Abbreviations: ADT androgen deprivation therapy; CI confidence interval; HR hazard ratio; mCRPC metastatic castration-resistant prostate cancer; ORR objective response rate; OS overall survival; PD-L1 programmed death-1 ligand 1; PSA prostate-specific antigen; rPFS radiographic progression-free survival; RR response rate; TMB tumor mutational burden; TRAEs treatment-related adverse events.
Table 3. Combination strategies and targets with ICIs in mCRPC.
Table 3. Combination strategies and targets with ICIs in mCRPC.
Trial/NCTPhasePatient PopulationTreatmentPrimary EndpointResults
ICIs + cytokines
SWOG S0354 (NCT00433446)IImCRPC patients with prior taxane therapy Siltuximab every 2 weeks for 12 cycles PSA RR defined as 50% reduction Overall PSA RR of 3.8% (95% CI: 0.5%, 13.0%)
ICIs + cancer vaccines
NCT030242161bAsymptomatic or minimally symptomatic mCRPC patientsAtezolizumab followed by sipuleucel-T (Arm 1) or sipuleucel-T followed by atezolizumab (Arm 2)SafetyAt least one treatment-related AE was reported in 31 subjects (83.8%), including 7 (18.9%) with at least one grade 3 treatment-related AE
ICIs + PARP inhibitors
KEYNOTE-365 (NCT02861573) Cohort A1b/IIDocetaxel-pretreated mCRPC patients who progressed within 6 months of screening and were molecularly unselected Pembrolizumab + olaparibSafety, PSA response rate, ORRThe confirmed PSA response rates in patients with a baseline PSA measurement were 15% (15/102) for the total population and 19% (11/59) for patients with RECIST-ORR was 8.5% (five PRs) in patients with RECIST-measurable disease. All 102 treated patients (100%) experienced at least one all-cause AE, and grade 3–5 AEs occurred in 74 patients (73%) Treatment-related AEs occurred in 93 patients (91%)
KEYLYNK-010 (NCT03834519)IIImCRPC patients who progressed after chemotherapy and either abiraterone or enzalutamidePembrolizumab + olaparib vs. next-generation hormonal agentOS, rPFSrPFS (median 4.4 months with pembrolizumab + olaparib vs. 4.2 months with next-generation hormonal agent; HR 1.02, 95% CI 0.82–1.25; p = 0.55) and OS (15.8 mo vs. 14.6 mo; HR 0.94, 95% CI 0.77–1.14; p = 0.26) were not met. Study was stopped for futility.
NCT02484404IImCRPC patients who had received prior enzalutamide and/or abiraterone unselected for somatic or germline mutationsDurvalumab + olaparib rPFS, PSA response9 of 17 patients (53%) had a PSA decline of ≥50%. Median rPFS for all patients is 16.1 months (95% CI: 4.5–16.1 months) with a 12-month rPFS of 51.5% (95% CI: 25.7–72.3%)
ICIs + radioligand therapies
PRINCE (NCT03658447)1bmCRPC patients with high PSMA expression (SUVmax ≥20 in an index lesion, SUVmax >10 for all lesions ≥10 mm), and no FDG positive/PSMA negative lesions on paired baseline PET/CT177Lu-PSMA-617 + pembrolizumabSafety, PSA response ratePSA response rate was 76% (28/37 [95% CI 59–88]) and 7/10 (70%) patients with RECIST-measurable disease had a partial response
NCT028146691bmCRPC patients with bone and lymph node and/or visceral metastases that progressed after androgen pathway inhibitor treatmentatezolizumab + radium-223Safety, ORR All 44 patients had ≥1 all-cause AE; 23 (52.3%) had a grade 3/4 AE. 15 (34.1%) grade 3/4 and 3 (6.8%) grade 5 AEs were related to atezolizumab; none were related to radium-223. Confirmed ORR was 6.8% [95% CI, 1.4–18.7]
NCT05150236IImCRPC patients with progression on prior androgen receptor pathway inhibitors, no more than one line of prior chemotherapy, significant PSMA avidity on 68GaPSMA-11 PET/CT (SUVmax ≥15 at one disease site and SUVmax ≥10 at measurable sites of disease. 10 mm), no FDG positive/PSMA negative disease and no contraindications to ICIIpilimumab + Nivolumab + 177Lu-PSMA-617 12-month PSA PFSOngoing
Abbreviations: ADT androgen deprivation therapy; AEs adverse events; CI confidence interval; FDG [18] F-fluorodeoxyglucose; HR hazard ratio; ICIs immune checkpoint inhibitors; mCRPC metastatic castration-resistant prostate cancer; ORR objective response rate; OS overall survival; PD-L1 programmed death-1 ligand 1; PFS progression free survival; PR partial response; PSA prostate-specific antigen; PSMA prostate-specific membrane antigen; rPFS radiographic progression-free survival; RR response rate; SUVmax maximum standardized uptake value; TMB tumor mutational burden.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lanka, S.M.; Zorko, N.A.; Antonarakis, E.S.; Barata, P.C. Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond. Curr. Oncol. 2023, 30, 4246-4256. https://doi.org/10.3390/curroncol30040323

AMA Style

Lanka SM, Zorko NA, Antonarakis ES, Barata PC. Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond. Current Oncology. 2023; 30(4):4246-4256. https://doi.org/10.3390/curroncol30040323

Chicago/Turabian Style

Lanka, Sree M., Nicholas A. Zorko, Emmanuel S. Antonarakis, and Pedro C. Barata. 2023. "Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond" Current Oncology 30, no. 4: 4246-4256. https://doi.org/10.3390/curroncol30040323

Article Metrics

Back to TopTop