Download PDF
Review  |  Open Access  |  24 Sep 2023

A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer

Views: 612 |  Downloads: 203 |  Cited:   3
Cancer Drug Resist 2023;6:656-73.
10.20517/cdr.2023.48 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Immunotherapy has become integral in cancer therapeutics over the past two decades and is now part of standard-of-care treatment in multiple cancer types. While various biomarkers and pathway alterations such as dMMR, CDK12, and AR-V7 have been identified in advanced prostate cancer to predict immunotherapy responsiveness, the vast majority of prostate cancer remain intrinsically immune-resistant, as evidenced by low response rates to anti-PD(L)1 monotherapy. Since regulatory approval of the vaccine therapy sipuleucel-T in the biomarker-unselected population, there has not been much success with immunotherapy treatment in advanced prostate cancer. Researchers have looked at various strategies to overcome immune resistance, including the identification of more biomarkers and the combination of immunotherapy with existing effective prostate cancer treatments. On the horizon, novel drugs using bispecific T-cell engager (BiTE) and chimeric antigen receptors (CAR) technology are being explored and have shown promising early efficacy in this disease. Here we discuss the features of the tumour microenvironment that predispose to immune resistance and rational strategies to enhance antitumour responsiveness in advanced prostate cancer.

Keywords

Prostate cancer, immunotherapy, immune checkpoint inhibitor, immune resistance, tumour microenvironment

INTRODUCTION

Prostate Cancer has the second highest cancer incidence worldwide and is the 5th leading cause of cancer death in men[1]. The cornerstone treatment of locally-advanced and metastatic prostate cancer centres upon androgen deprivation therapy. Patients who experience disease progression while having castrate levels of testosterone are considered castration-resistant. In the advanced prostate cancer setting, additional treatment modalities include novel hormonal agents (NHAs), chemotherapy, radioligand therapy, poly(ADP)-ribose polymerase (PARP) inhibitors, and immunotherapy. Successive waves of clinical trials in the past decade have brought these treatment modalities forth from the castration-resistant setting into the hormone-sensitive setting, showing improved survival with early introduction of chemotherapy, NHAs, or combinations of these[2]. Despite these advances in prostate cancer treatment, the 5-year survival for metastatic prostate cancer patients in 2022 remains low at 32.3%[3].

Immunotherapy, in the form of sipuleucel-T, received FDA approval in 2010 for the treatment of patients with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). In patients with deficient mismatch repair or microsatellite-high (dMMR/MSI-H) tumours, pembrolizumab and dostarlimab are FDA-approved options[4,5]. However, the prevalence of dMMR/MSI-H in prostate cancer is dismal at 1%, with MSH2 being the most frequently implicated (other MMR genes being MSH6, MLH1, PMS2)[6]. Owing to an immunologically “cold” microenvironment in unselected acinar prostate adenocarcinoma, to date, no other immunotherapeutic agents have shown to be beneficial in the current treatment of advanced prostate cancer. In this review, we look at the current treatment paradigm, the role of immunotherapy, and existing and up-and-coming methods to overcome immune therapy resistance in prostate cancer.

IMMUNE REGULATION IN THE TUMOUR MICROENVIRONMENT (TME) OF PROSTATE CANCER

Immuno-oncology has changed the treatment paradigm of multiple tumour types, including melanoma, renal cell carcinoma, and lung carcinoma. The cancer-immunity cycle is depicted in Figure 1, explaining how the innate immune system fends off cancer cells and the various points at which therapeutic targets act. Despite successes in these typically immunogenic tumours, prostate cancer has traditionally been considered to have an immunologically “cold” tumour microenvironment (TME) characterized by T cell exclusion, low neoantigen load, and a highly immunosuppressive microenvironment comprising a high proportion of myeloid-derived suppressor cells (MDSCs)[7,8]. Factors that suggest a maladaptive immune response against tumour cells include lack of tumour-infiltrating lymphocytes (TILs), presence of M2-polarized tumour-associated macrophages (TAMs) and MDSCs, with evidence that increment in such cell populations within the TME is correlated with tumour progression[9]. MDSCs are immune cells that are activated in cancers and display potent immunosuppressive effects leading to prostate cancer resistance to anti-hormonal therapy[10]. Furthermore, CRPCs frequently exhibit PTEN loss, which is associated with increased MDSC infiltration[11] and may interact with the interferon-1 pathway required for innate immune activation[12]. Other immune-suppressive factors within the TME, such as soluble tumour necrosis factor (sTNF), interleukin-1 beta (IL-1β), TGF-β, and IL-10, promote chronic inflammation and increase myeloid cell differentiation into MDSCs[13,14].

A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer

Figure 1. The cancer immunity cycle and where various classes of drugs act on.

Reduced immune stimulatory factors can also contribute to the immunologically cold TME in prostate cancer. CRPC patients have decreased peripheral natural killer (NK) cell pools, and this may be due to increased NK cell group 2 member D (NKG2D) serum receptor levels from the tumour[15]. This phenomenon is more pronounced with metastatic disease[9]. NK cells are lymphocytes that have roles in innate and adaptive immunity, whereas NKG2D is an activating cell surface receptor expressed on NK cells, NKT cells, and subsets of γδ T cells. Although initially thought to enhance immune responses against cancer, it appears that when NKG2D ligands are expressed chronically, this can instead lead to inhibition of immune cell function[16]. Low tumour mutational burden (TMB) in prostate cancer is associated with reduced neoantigen load recognised by the immune system[17]. These mechanisms enable immune evasion by cancer cells and directly impact the therapeutic response to anti-PD(L)1/anti-CTLA4 immune checkpoint inhibitors (ICIs)[18]. Figure 2 illustrates the interplay amongst the immune cells, cancer cells and vascular supply within the TME.

A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer

Figure 2. The immunologically “cold” tumour microenvironment in prostate cancer.

Potential biomarkers for ICI response include dMMR/MSI-H as mentioned above and tumours with DNA damage repair (DDR) pathway deficiencies. Tumours with DDR pathway deficiencies have increased mutational load as a result of decreased DNA repair capacity, leading to genomic instability[19]. Patients with somatic alterations in genes involved in DNA replication or repair have been shown to express higher neoantigen load, higher mutational burdens, higher levels of CD3+ and CD8+ TILs and higher PD-1/PD-L1 levels, all of which correlate with sustained ICI responses[20-24]. Despite this, dMMR and CDK12-altered prostate cancers have more aggressive biology[25,26]. A retrospective study of prostate cancer patients from the Royal Marsden Hospital showed that 8.1% of the patients had dMMR, which was correlated with decreased survival (median OS 4.1 years for dMMR vs. 8.5 years for proficient MMR)[26]. CDK12 alterations were found in 6% of advanced prostate cancer in one study[25], and were typically linked to poor prognosis as well as insensitivity to PARP inhibitors[27]. However, these tumours have increased neoantigen load and tumoural lymphocyte infiltration, which may increase their response to ICIs[27].

ICI MONOTHERAPY IN THE UNSELECTED PROSTATE CANCER PATIENT

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a receptor found on the surface of T lymphocytes. When APCs activate T cells in response to the presence of foreign antigens, there is involvement of costimulatory molecules such as CD28 and B-7, which enhance the immune response. CTLA-4 acts as an immune checkpoint by binding to B-7, counteracting the costimulatory effect of CD28 and overall cause suppression of the immune response[28,29]. Cancer cells can downregulate the immune response by exploiting CTLA-4, and this forms the basis of targeting CTLA-4 with monoclonal antibodies such as ipilimumab. Inhibition of CTLA-4 activity causes activation and proliferation of cytotoxic T cells against tumour cells[30,31]. To date, two phase 3 trials have looked at the activity of ipilimumab in mCRPC patients. The first study, CA 184-043, recruited 799 mCRPC patients with at least one bone metastasis and have progressed on docetaxel chemotherapy. Patients were randomised to receive either one fraction of bone-directed radiation therapy followed by ipilimumab at 10 mg/kg or placebo. There was no overall survival benefit seen in this study (median OS 11.2 vs. 10 months, HR 0.85, 95% CI 0.72-1.00), but a progression-free survival (PFS) benefit (4.0 vs. 3.1 months, HR 0.70, 95% CI 0.61-0.82) was seen[32]. The second study by Beer et al. (2017) randomised 602 mCRPC patients who were chemotherapy-naive and had no visceral metastases to ipilimumab at 10 mg/kg vs. placebo. The study showed no overall survival benefit (median OS 28.7 vs. 29.7 months; HR 1.11, 95% CI 0.88-1.39), although a PFS benefit (median PFS 5.6 vs. 3.8 months; HR 0.67; 95% CI 0.55-0.81) was observed. Exploratory analyses further showed a higher prostate-specific antigen (PSA) response rate with ipilimumab (23%) than with placebo (8%)[33]. Taken together, the PFS and PSA response with ipilimumab suggests antitumour activity despite the lack of survival benefit.

PD-1 is a transmembrane glycoprotein found on the surfaces of activated cytotoxic T cells, B cells, dendritic cells, NK cells, and macrophages[34]. The binding of PD-1 to its ligands programmed death ligands 1 and 2 (PD-L1 and PD-L2) found on cancer cells delivers inhibitory signals for T-cell activation, suppressing an immune response[35,36]. Monoclonal antibodies targeting PD-1/PD-L1, such as nivolumab and pembrolizumab, have shown activity in multiple cancer types, leading to regulatory approval for their use[37,38]. Pembrolizumab was studied in the phase 1b KEYNOTE-028 and phase 2 KEYNOTE-199 trials as monotherapy in mCRPC, showing poor responses[39,40]. The objective response rate (ORR) was 5% in PD-L1 combined positive score (CPS) ≥ 1 patients in KEYNOTE-199, compared with 3% for patients with a negative PD-L1 expression[39]. Three phase 1 dose-escalation trials of nivolumab monotherapy in mCRPC patients likewise showed no objective response[41-43]. As mentioned, the paucity of PD-L1 expression in the TME in prostate cancer patients could account for this. Despite the glaringly low response rates for anti-PD(L)1/anti-CTLA4 monotherapies in unselected prostate cancer, the expression of immune checkpoints has been reported to be dynamic, and various agents such as ipilimumab, sipuleucel-T and enzalutamide can increase T cell infiltration into the TME and modulate response to anti-PD(L)1 therapy[44]. This sets the stage for combination of various therapies with ICIs to improve immunotherapeutic response in prostate cancer.

ONGOING STRATEGIES TO OVERCOME IMMUNE RESISTANCE

Several strategies have been examined to modulate antitumour immunity in advanced prostate cancer.

PARP inhibitors and ICIs

PARP inhibitors are small molecules that prevent the repair of single-strand DNA breaks. Pathogenic DDR gene alterations are found in 23% of mCRPCs[45], with BRCA2, ATM, CHEK2, and BRCA1 being the most frequently implicated genes[46]. The resulting homologous recombination deficiency (HRD) leads to sensitivity to PARP inhibition as a result of synthetic lethality[47]. Presently in mCRPC patients, the FDA has approved rucaparib for use in those with germline/somatic BRCA mutation and olaparib for those with germline/somatic homologous recombination repair (HRR) gene mutations. This is based on a high ORR of 50.8% seen with rucaparib use in the phase 2 TRITON2 study and improved radiologic PFS with olaparib use over enzalutamide/abiraterone in the phase 3 PROfound study[48,49]. The phase 3 TRITON3 study showed similar results[50]. Furthermore, efforts made in examining PARP inhibition in unselected patients have been successful as well, with the phase 3 PROpel trial showing improvement in radiologic PFS with combination abiraterone plus olaparib over abiraterone plus placebo as first-line treatment of mCRPC patients, overall suggesting an increasing role in PARP inhibition[51].

Increased micronuclei and cytosolic double-stranded DNA release after PARP inhibitor treatment as a result of PARP-DNA trapping and DNA damage leads to increased neoantigen formation, increased PD-L1 expression, increased intra-tumoural CD8 T cell infiltration and increased interferon production in the TME, forming the basis for ICI-PARP inhibitor combinations, and potentially expanding the benefit of PARP inhibitors beyond tumours harbouring alterations[52,53]. A phase 2 open-label clinical trial combining durvalumab with olaparib in men with mCRPC showed a response (radiographic or biochemical) in 9 out of 17 patients. Five of the 9 responders were found to have dysfunctional DDR genes based on genomic analysis and the presence of mutated DDR genes was associated with significantly higher 12-month PFS than those without (83.3% vs. 36.4%). Interestingly, patients with fewer peripheral MDSCs were more likely to respond[54]. This study showed early evidence of combining PARP inhibitors and ICIs, and other ongoing studies looking at similar combinations are listed in Table 1.

Table 1

Trials looking at ICI combinations in treatment of advanced prostate cancer

Trial numberPhaseIntervention arm(s)PopulationOutcomeStatus
ICIs + PARP inhibitor
NCT024844042Durvalumab + OlaparibmCRPC after progression with 1 NHA or DocetaxelORR, safety, DOR, PSA responseCompleted
NCT043369432Durvalumab + OlaparibRecurrent prostate cancer with immunogenic signaturePSA responseActive, recruiting
NCT038345193Pembrolizumab + Olaparib
NHA (Abiraterone or Enzalutamide)
mCRPC after progression with 1 NHA and chemotherapyOS, rPFSActive, not recruiting
NCT028615731/2Pembrolizumab + Olaparib
Multiple cohorts
mCRPCORR, safety, PSA responseActive, recruiting
NCT055685502Pembrolizumab + Olaparib + RT
Pembrolizumab + RT
High-risk localised PCPSA responseNot yet recruiting
NCT033387902Nivolumab + Rucaparib
Nivolumab + Docetaxel
Nivolumab + Enzalutamide
mCRPCORR, PSA responseActive, not recruiting
NCT045922372Maintenance Cetrelimab + Niraparib
Maintenance Niraparib
Aggressive variant mPC given induction Cabazital + Carboplatin + CetrelimabPFSActive, recruiting
ICIs + vaccines
NCT030242161Atezolizumab + Sipuleucel-TmCRPCSafetyCompleted
NCT018328701Ipilimumab + Sipuleucel-TCRPC eligible to receive Sipuleucel-T in accordance to FDA-approved labelingAntigen-specific T cell response, antibody responseCompleted
NCT001139841MDX-010 (anti-CTLA-4) + PROSTVAC-V/TRICOM (virus vaccine)mCRPC after progression with anti-androgens and ≤ 1 chemotherapySafetyCompleted
NCT029332551/2Nivolumab + PROSTVAC-V/FmCRPC
Neoadjuvant therapy for localised PC planned for surgery
Safety, changes in T-cell infiltrationActive, recruiting
NCT033158712M7824 (anti-PD-L1/TGFβ) + PROSTVAC + CV301 (virus vaccine)CRPCPSA responseActive, recruiting
NCT035322171Ipilimumab + Nivolumab + PROSTVAC-V/F + Neoantigen DNA vaccinemHSPCDLT, safety, immune responseCompleted
NCT034939451/2M7824 (anti-PD-L1/TGFβ) + BN-Brachyury (virus vaccine)+ N-803 (IL-15 superagonist complex) + Epacadostat (IDO1 inhibitor)CRPCCBRActive, recruiting
NCT023255571/2Pembrolizumab + ADXS31-142 (bacteria vaccine)mCRPC after progression on ≤ 3 systemic therapiesSafetyUnknown
NCT024998351/2pTVG-HP + concurrent Pembrolizumab
pTVG-HP + sequential Pembrolizumab
mCRPCORR, safety, PSA response, PFSActive, not recruiting
NCT040905282Pembrolizumab + pTVG-HP (DNA vaccine) + pTVG-AR HP (DNA vaccine)
Pembrolizumab + pTVG-HP
mCRPCPFSActive, recruiting
NCT043828981/2Cemiplimab + BNT112
BNT112 (RNA vaccine)
mCRPC after progression on 2-3 therapies including NHA and/or chemotherapy DLT, ORR, SafetyActive, recruiting
ICIs + tyrosine kinase inhibitors
NCT044461173Atezolizumab + Cabozantinib + NHA (Abiraterone or Enzalutamide)mCRPC after progression on 1 NHAPFS, OSActive, recruiting
NCT031709601/2Atezolimab + CarbozantinibmCRPC after progression on ≤ 1 NHADLT, ORRActive, not recruiting
NCT044775121Nivolumab + Cabozantinib + AbirateronemHSPCDLTActive, recruiting
NCT041598962Nivolumab + ESK981 (Pan-VEGFR/TIE2 TKI)mCRPC after progression on 1 NHA and 1 chemotherapySafety, PSA responseUnknown
Combination ICIs
NCT047171542Ipilimumab + NivolumabmCRPC with immunogenic signatureDCRActive, recruiting
NCT035706192Ipilimumab + NivolumabmCRPC with CDK12 aberrationORR, PSA responseActive, not recruiting
NCT030615392Ipilimumab + NivolumabmCRPC with immunogenic signature after progression on 1 systemic therapyORR, PSA responseActive, not recruiting
NCT029859572Ipilimumab + Nivolumab
Ipilimumab
Cabazitaxel
mCRPCORR, rPFSActive, not recruiting
NCT033336162Ipilimumab + NivolumabNon-adenocarcinoma PCORRActive, recruiting
NCT027887732Durvalumab + TremelimumabmCRPC with prior exposure to 1 NHAORRActive, not recruiting
ICIs + androgen receptor antagonist
NCT030163123Atezolizumab + Enzalutamide
Enzalutamide
mCRPC with prior exposure to 1 NHA and 1 chemotherapyOSCompleted
NCT027870052Pembrolizumab + EnzalutamidemCRPC progressing on EnzalutamideORRCompleted
NCT041910963Pembrolizumab + Enzalutamide
Enzalutamide
mHSPCrPFS, OSActive, not recruiting
NCT038344933Pembrolizumab + Enzalutamide
Enzalutamide
mCRPC, allows for prior Abiraterone exposurerPFS, OSActive, not recruiting
NCT023125572Pembrolizumab + EnzalutamidemCRPC after progression on EnzalutamidePSA responseActive, not recruiting
NCT033387902Nivolumab + Rucaparib
Nivolumab + Docetaxel
Nivolumab + Enzalutamide
mCRPCORR, PSA responseActive, not recruiting
NCT016884921/2Ipilimumab + AbirateronemCRPCSafety, PFSActive, not recruiting
ICIs + chemotherapy
NCT03338790
2Nivolumab + DocetaxelmCRPCORR, PSA responseActive, not recruiting
NCT041000183Nivolumab + Docetaxel
Nivolumab
mCRPC after progression on 1-2 NHAsrPFS, OSActive, recruiting
NCT038345063Pembrolizumab + Docetaxel
Docetaxel
mCRPC with prior exposure to 1 NHArPFS, OSActive, not recruiting
NCT028615731/2Pembrolizumab + Docetaxel
Multiple cohorts
mCRPCORR, safety, PSA responseActive, recruiting
NCT034094581/2Avelumab + PT-112 (Platinum + Pyrophosphate ligand)mCRPCSafety, PSA responseActive, not recruiting
NCT026010142Nivolumab + IpilimumabAR-V7-expressing mCRPCPSA responseCompleted
NCT027887732Durvalumab + Tremelimumab
Durvalumab
mCRPC with prior exposure to 1 NHAORRActive, not recruiting
ICIs + radiopharmaceuticals
NCT028146691Atezolizumab + Radium-223mCRPC after progression on 1 NHA and 1 chemotherapyORR, safetyCompleted
NCT041097291/2Nivolumab + Radium-223mCRPC with symptomatic bone metastasesSafety, ctDNA reductionActive, recruiting
NCT036584471/2Pembrolizumab + 177Lu-PSMAmCRPC after progression on 1 NHASafety, PSA responseCompleted

As mentioned, CDK12-altered prostate cancers typically carry poor prognosis and do not respond well to PARP inhibition, yet they present increased neoantigen load and lymphocytic infiltration, which may increase responsiveness to anti-PD1 therapy[25,27]. A retrospective study of 60 men with CDK12-altered advanced prostate cancer showed that of the 9 men who received PD-1 inhibitor therapy, 33% had a PSA response and the median PFS was 5.4 months[27,55]. Similarly, the ongoing phase 2 IMPACT trial has shown a 21.4% PSA response with ipilimumab-nivolumab combination in these patients[55].

Vaccines and ICIs

Anti-cancer vaccines can be classified into four groups: cell-based, viral-based, DNA/RNA-based, and peptide-based vaccines[56,57]. The goal of vaccine therapy is to stimulate the host’s adaptive immune response against tumour-associated antigens (TAA). Prostate cancer is suitable for vaccine therapy because it has many TAAs such as PSA, prostate-specific membrane antigen (PSMA), prostate acid phosphatase (PAP), prostate stem cell antigen (PSCA), prostate cancer antigen 3 (PCA3), mucin-1, and six-transmembrane epithelial antigens of the prostate (STEAP)[58].

Sipuleucel-T is a therapeutic dendritic cell-based vaccine that has received FDA approval for use in the treatment of patients with asymptomatic or minimally symptomatic mCRPC, based on overall survival (OS) benefit seen from the phase 3 IMPACT trial[59]. It is prepared from autologous peripheral blood mononuclear cells obtained by leukapheresis, and pulsed ex vivo with PAP2024, a unique fusion protein of granulocyte-macrophage colony-stimulating factor (GM-CSF) and prostatic acid phosphatase (PAP). GM-CSF fosters the maturation of dendritic cells and other APCs to present PAP to the patient’s T cells, resulting in PAP-specific T-cell proliferation targeting the PAP-expressing prostate cancer cells for killing. Both humoural and cellular responses have been reported, with peripheral immune responses to PAP and measures of APC activation correlating with improvements in OS[60,61]. Despite success with the use of sipuleucel-T, other vaccines studied have not been as successful. G-VAX is another cell-based GM-CSF-secreting vaccine that utilises irradiated TAAs[62]. The TAAs are derived from two cell lines: one hormone-sensitive (LNCaP) and one hormone-resistant (PC3)[63]. Despite initially promising results in asymptomatic mCRPC, the phase 3 VITAL 1 and VITAL 2 trials in asymptomatic mCRPC and symptomatic mCRPC patients, respectively, failed to show the OS benefit of G-VAX plus docetaxel against docetaxel alone. Both studies were terminated early based on futility assessments. A viral-based vaccine, PROSTVAC, utilizes recombinant poxviruses that express PSA with immune-enhancing costimulatory molecules to stimulate immune response[64,65]. In addition to induced modified human PSA, they contain three costimulatory domains for T cells (B7.1, leukocyte function-associated antigen-3, and intercellular adhesion molecule-1), called TRICOM[66]. The phase 3 PROSPECT trial was unable to demonstrate the OS benefit of PROSTVAC against placebo control[67].

Given the increase in T cell infiltration and inflammation within TME with sipuleucel-T[60,61], it is therefore postulated that synergy might be observed with the combined use of vaccines and ICIs. Ipilimumab and PROSTVAC were combined in a phase 1 dose-escalation trial, showing evidence of improved clinical and immunologic outcomes. The median OS was 34.4 months[68], which appears to be numerically larger than PROSTVAC alone in its original study[67]. There was a PSA reduction in 54% of patients and a PSA decline of more than 50% was seen in 25% of patients. ADXS31-142 is a live, attenuated, bioengineered listeria-based vaccine targeting PSA. It is being studied as part of the KEYNOTE-046 trial, with current results showing a median OS of 33.7 months for patients treated with combination vaccine and pembrolizumab[69]. Other ongoing studies of vaccine therapy with ICIs are listed in Table 1.

Tyrosine kinase inhibitors and ICIs

Prostate cancers have dysregulated vasculature that promotes an immunosuppressive TME[7,8]. These include promoting a shift in TAMs toward M2-like immunosuppressive phenotype, reduced maturation of dendritic cells which results in reduced antigen presentation, and increased PD-L1 expression[70]. Vascular endothelial growth factor (VEGF) overexpression has been found to prevent the differentiation of monocytes into dendritic cells[71]. Meanwhile, an improvement in the regulation of local vascular in preclinical models was associated with the assimilation of TAMs with M1-like immune-stimulatory phenotype, increased CD4+ and CD8+ T-cell infiltration into the TME, and reduction of MDSCs[72-75]. These suggest that targeting angiogenesis in tumours can inhibit tumour-induced dysregulation of local vasculature and promote immunogenicity in the TME, forming the basis of combining antiangiogenesis agents with ICIs. Indeed, it has been shown in renal cell carcinoma that anti-VEGF therapy leads to a reduction in immune inhibitory stimuli such as regulatory T-cells and MDSCs[76,77]. Aside from VEGFR targeting, the TAM family of receptor tyrosine kinases comprising TYRO3, AXL and MER has been shown to promote immune suppression as well, making it an attractive target[78,79].

Cabozantinib is a multi-kinase inhibitor targeting MET, VEGFR-1, -2 and -3, AXL, RET, ROS1, TYRO3, MER, KIT, TRKB, FLT-3, and TIE-2[80]. Preclinical data suggests that it has an effect on the TME by reprogramming M2 TAMs to “pro-inflammatory” M1 macrophages, in addition to reducing MDSCs and T regulatory cells[81]. A dose-expansion cohort in the phase 1b COSMIC-021 trial evaluated the combination of cabozantinib with atezolizumab (anti-PD1) in mCRPC patients who have had disease progression following treatment with novel hormonal agents such as abiraterone or enzalutamide. An ORR of 32% was observed in 132 patients treated with the combination, with a disease control rate (DCR) of more than 80%. This effect was consistent in patients with visceral disease as well[82]. Due to promising results from this study, this combination is now being evaluated in a phase 3 clinical trial for mCRPC patients. Other ongoing studies looking at combination anti-VEGF therapy with ICIs are listed in Table 1.

Combination ICIs

CheckMate-650 is a phase 2 study looking at various dosing combinations of nivolumab with ipilimumab in asymptomatic or minimally symptomatic mCRPC patients who have progressed on novel hormone therapy in two cohorts (chemotherapy-naive and chemotherapy-exposed). In the chemotherapy-naive cohort, nivolumab/ipilimumab achieved an ORR of 25% with a median radiological PFS of 5.5 months and a median OS of 19.0 months. In the chemotherapy-exposed cohort, the ORR was 10%, with a median radiological PFS of 3.8 months and a median OS of 15.2 months[83]. Exploratory analyses revealed that PD-L1 ≥ 1%, the presence of DDR or homologous recombination deficiency (if at least one gene in the relevant gene panel had a deleterious mutation/homozygous deletion) were associated with higher ORR[83]. In this study, 44 patients had quality-controlled whole-exome sequencing data, giving rise to a median TMB of 74.5 mutations/patient. Tumours harbouring TMB exceeding this median were associated with higher ORR, PSA response rate, radiologic PFS, and median OS[83].

Combination nivolumab and ipilimumab has been examined in AR-V7 expressing mCRPC patients as well. Androgen receptor splice variant 7 (AR-V7) expression is found in approximately 20% of mCRPC patients and is associated with alterations in a greater number of DDR genes, which could increase susceptibility to ICIs[84]. The STARVE-PC trial is a phase 2 non-randomised study that evaluated the activity of nivolumab and ipilimumab in 15 AR-V7 expression mCRPC patients, showing an ORR of 25%, PSA response rate of 13% and OS of 8.2 months[85]. Responses were more pronounced in six of the patients who were found to have mutations in DDR genes (three in BRCA2, two in ATM, and one in ERCC4)[86]. Lastly, an ongoing phase 2 randomised study is looking at mCRPC patients following progression on novel hormonal agents, randomising them to receive durvalumab or combination durvalumab plus ipilimumab. The ORR with combination ICI was 16% vs. 0% with durvalumab monotherapy in this study[87]. Other ongoing trials evaluating the efficacy of combination ICIs are listed in Table 1.

Androgen receptor antagonists and ICIs

How prostate cancer treatment impacts the immune response is variable. ADT enhances lymphopoiesis, which can mitigate immune tolerance to prostate cancer antigens[88]. On the other hand, androgen receptor antagonists have been shown to inhibit T cell responses[89].

ADT and anti-androgens can both target the AR signalling pathway and have been shown to result in an increase in the number of TILs, and a decrease in the number of regulatory T cells supporting an antitumour response to ADT[90,91]. Animal models confirm that while ADT induces pro-inflammatory conditions initially, the subsequent development of castration resistance and immune tolerance to prostate cancer antigens reduces this[92,93]. Therefore, the combination of AR-signalling blockade with ICIs, especially during its pro-inflammatory state, may be beneficial in the treatment of advanced prostate cancer.

The phase 2 IMbassador250 trial examined 759 advanced CRPC patients who had progressed on abiraterone and docetaxel, randomising them to receive combination enzalutamide and atezolizumab vs. enzalutamide alone. The study was closed prematurely due to futility (combination therapy vs. enzalutamide monotherapy, 15.2 vs. 16.6 months; HR 1.12, 95% CI 0.91-1.37). However, pre-planned exploratory analyses showed a longer PFS with combination therapy in patients with high PD-L1 IC2/3, CD8 expression[94]. The study also performed an unbiased RNA sequencing-based analysis of immune-related gene expression that had previously correlated with mCRPC responses to immunotherapy[95], and found longer PFS with combination therapy in patients harbouring genes related to pre-existing immunity such as TAP-1, CXCL9, interferon signalling[94]. The multicohort phase 2 KEYNOTE-199 trial examined combination pembrolizumab with enzalutamide in mCRPC patients whose disease were refractory to enzalutamide. In the cohorts with measurable disease and bone-predominant disease (cohorts 4 and 5), the disease control rate was 51% and ORR was 12%. The duration of response was almost 6 months in 60% of responders[96]. This strategy is being evaluated further in an ongoing phase 3 trial [Table 1].

Systemic chemotherapy and ICIs

Chemotherapy may potentiate antitumour immunity by various mechanisms, including the release of TAAs and enhancing antigen presentation, stimulating the activity of cytotoxic T lymphocytes[97,98]. Importantly, chemotherapy may reduce immunosuppressive cell populations such as MDSCs and regulatory T cells, known to maintain prostate cancer immune evasion[99,100]. Preclinical studies have suggested that chemotherapy does improve antitumour immune responses, showing that the addition of taxanes can cause a shift in macrophage populations toward the M1-like (immune-activating) phenotype and reduce regulatory T cell and MDSC populations in mouse models[101,102]. The multicohort phase 2 trial CheckMate 9KD showed that combination nivolumab and docetaxel in 41 chemotherapy-naive mCRPC patients who have progressed on novel hormonal agents achieved an ORR of 36.8%, radiologic PFS of 8.2 months and PSA response of 46.3%[103]. KEYNOTE-365 is an ongoing multicohort phase 1b/2 study examining combination pembrolizumab and docetaxel in mCRPC patients, yielding an ORR of 18%, PSA response of 28%, radiologic PFS of 8.3 months, and OS of 20.4 months[104]. Ongoing phase 3 trials (CheckMate7DX and KEYNOTE-921) evaluating the superiority of combination chemotherapy with immunotherapy over chemotherapy alone will shed light in this area [Table 1].

Radiopharmaceuticals and ICIs

177Lu-PSMA-617 has gained regulatory approval for the treatment of mCRPC patients who have been treated with androgen receptor (AR) pathway inhibition and taxane chemotherapy, based on positive results on a phase 3 trial[105]. In murine models, targeted radionuclide therapy (TRT) may increase PD-L1 expression on T cells and the combination of TRT with ICIs leads to increased infiltration of CD8 T cells[106]. There is, hence, interest in combining radionuclide therapy with ICIs. Despite low clinical response (ORR 6.8%, PSA response 4.5%, radiologic PFS 3 months) seen on a phase 1b trial combining Atezolizumab and Radium-223 in mCRPC[107], the interim results of another phase 1b/2 PRINCE trial are relatively promising. In this study, 37 mCRPC patients who have progressed on a novel hormonal agent and docetaxel were treated with pembrolizumab and 177Lu-PSMA-617, yielding an ORR of 78%, PSA response of 73%, and 24-week radiologic PFS of 65%[108] [Table 1].

FUTURE DIRECTIONS AND CONCLUSIONS

Research is ongoing to identify more immunogenic targets and pair them with the multiple TAAs that prostate cancer expresses. Amongst these, cellular-based therapy is an area that deserves special mention. Adoptive cell therapy involves the engineering of patients’ T lymphocytes to target specific viruses or tumours. The use of chimeric antigen receptors (CAR) allows for the creation of artificial T-cell receptors used in adoptive cell therapy[109]. A first-in-human phase 1 study of 13 CRPC patients tested PSMA-targeting CAR T cells armoured with a dominant-negative TGF-β receptor. TGF-β is an inhibitory factor found at high levels within the prostate TME. In this study, 4 patients had a ≥ 30% reduction in PSA and 1 patient had a > 98% reduction in PSA. Five patients experienced grade 2 or higher cytokine-release syndrome (CRS)[110]. Another CAR T therapy using P-PSMA-101, which targets PSMA, was evaluated in 10 heavily-pre-treated CRPC patients, yielding PSA decline in 7 patients, with 4 patients having > 50% reduction in PSA. CRS was seen in 60% of patients[111]. Other CAR T products targeting Epithelial cell adhesion molecule (EpCAM) and Natural Killer Group 2D (NKG2D) have shown activity in prostate cancer patients as well[112,113]. Other potential targets of interest with adoptive cell therapy include PSA, PAP, PSCA, and B7-H3[114], and Table 2 shows a list of ongoing clinical trials.

Table 2

Trials looking at novel therapies in advanced prostate cancer

Trial numberPhaseIntervention arm(s)PopulationOutcomeStatus
CAR T
NCT042272751CART-PSMA-TGFβRDNmCRPC after progression on 2 NHAsDLT, safetyActive, not recruiting
NCT030892031CART-PSMA-TGFβRDNmCRPC after progression on ≥ 1 systemic therapySafetyActive, recruiting
NCT040530621LIGHT-PSMA-CARTmCRPC after progression on Abiraterone and chemotherapySafetySuspended
NCT042499471P-PSMA-101 CAR-TmCRPCORR, DLT, safetyActive, not recruiting
NCT038738051Anti-PSCA-CAR-4-1BB/TCRzeta-CD19t-expressing T-lymphocytesPSCA+ mCRPCDLT, safetyActive, recruiting
NCT027442871/2BPX-601 (PSCA-specific CAR-T cells)PSCA+ mCRPCDLT, safetyActive, recruiting
NCT030137121/2EpCAM-specific CAR T CellsEpCAM+ mCRPCSafetyUnknown
BiTE
NCT041046071CC-1 (PSMAxCD3)mCRPC after progression on ≥ 3 systemic therapiesSafetyActive, recruiting
NCT037928411Acapatamab (PSMAxCD3)mCRPC after progression on 1 NHA and 1 chemotherapyDLT, safetyActive, not recruiting
NCT011403731/2HPN424 (PSMAxCD3)mCRPC after progression on ≥ 2 systemic therapiesORR, DLTActive, not recruiting
NCT039726571/2REGN5678 (PSMAxCD28) + CemiplimabmCRPC after progression on ≥ 2 systemic therapiesORR, DLT, safetyActive, recruiting
NCT042215421AMG 509 (STEAP1xCD3)mCRPC after progression on 1 NHA and 1 chemotherapyDLT, safetyActive, recruiting
NCT034068582HER2Bi-armed activated T cells (HER2xCD3) + PembrolizumabmCRPCPFSActive, not recruiting

Bispecific T cell engager (BiTE) antibodies is another technology that has been developed to target TAAs such as PSMA in prostate cancer cells. Structurally, these are bispecific monoclonal antibodies that can crosslink TAAs with the coreceptors on T cells, generating an antitumour immune response. Pasotuxizumab is a bispecific monoclonal antibody that crosslinks CD3 and PSMA, and its efficacy has been studied in 16 mCRPC patients on a phase 1 trial, showing ≥ 50% decline in PSA in 3 patients, of which two were long-term responders treated for 14.0 and 19.4 months, respectively. 81% of the patients had adverse events of grade ≥ 3[115]. The efficacy of AMG 160, a BiTE product that binds CD3 on T cells and PSMA on cancer cells, was evaluated in mCRPC patients on a phase 1 trial. In the preliminary report, 27% of patients had confirmed PSA responses and 84% of patients experienced CRS (10% grade ≥ 3)[116]. The study also had a subset of patients who received AMG 160 with pembrolizumab, and such a combination will likely be examined in future studies as well. Other potential BITE targets including STEAP, CEACAM5, DLL3, HER2 are being studied[117,118], and a list of ongoing trials can be seen in Table 2. Figure 3 shows a schematic diagram of BiTE therapy.

A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer

Figure 3. Bispecific T cell engager binding CD3 on T cell with PSMA on prostate cancer cell. BiTE: Bispecific T-cell engager; PSMA: prostate-specific membrane antigen.

On the horizon, relevant and novel targets to modulate antitumour immunity in prostate cancer may include the targeting of relevant immune-metabolic pathways, such as the adenosine receptor[119-121], or cytokine-directed efforts, such as IL-8 involved in the differentiation of TAM to M2 phenotype (promotes immune resistance and tumour metastasis)[122,123], IL-23 which is a cytokine secreted by MDSCs[124] and TGF-β which promotes tumour growth and immunosuppression in the TME[81]. Targeting cell signalling pathways such as the phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway has also been shown to downregulate immunosuppressive cells such as T regulatory cells and may have a role in improving ICI efficacy in prostate cancer[125,126]. For example, in prostate cancer mouse models, intermittent PI3K inhibition was able to alleviate PTEN-null cancer cell-intrinsic immunosuppressive activity and turn “cold” tumours into T cell-inflamed ones[127]. Novel immune checkpoints may be worth exploiting in prostate cancer. Increased expression of V domain Ig suppressor of T Cell activation (VISTA) was found to promote immune resistance following Ipilimumab treatment, which may serve as a new immunotherapeutic target in advanced prostate cancer[128].

There are presently limited biomarkers that can identify prostate cancer patients who may benefit from ICI therapy. It appears that combination strategies to promote immunogenicity within the “cold” TME of prostate cancer can increase the effect of ICIs. We recognise that the majority of the existing efforts are presently in the preclinical or early phase setting and may not be ready for use in the clinics yet. It would nevertheless be interesting to monitor this space for future developments.

DECLARATIONS

Authors’ contributions

Conceptualisation: Sooi K, Wong A, Ngoi N

Methodology: Sooi K, Walsh R, Wong A, Ngoi N

Writing: Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N

Supervision: Kumarakulasinghe N, Wong A, Ngoi N

Visualisation: Sooi K

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209-49.

2. Sekhoacha M, Riet K, Motloung P, Gumenku L, Adegoke A, Mashele S. Prostate cancer review: genetics, diagnosis, treatment options, and alternative approaches. Molecules 2022;27:5730.

3. Cancer stat facts: prostate cancer. Available from: https://seer.cancer.gov/statfacts/html/prost.html. [Last accessed on 19 Sep 2023].

4. FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-first-tissuesite-agnostic-indication. [Last accessed on 19 Sep 2023].

5. FDA grants accelerated approval to dostarlimab-gxly for dMMR advanced solid tumors. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-dostarlimab-gxly-dmmr-advanced-solid-tumors. [Last accessed on 19 Sep 2023].

6. Sedhom R, Antonarakis ES. Clinical implications of mismatch repair deficiency in prostate cancer. Future Oncol 2019;15:2395-411.

7. de Bono JS, Guo C, Gurel B, et al. Prostate carcinogenesis: inflammatory storms. Nat Rev Cancer 2020;20:455-69.

8. 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.

9. Gannon PO, Poisson AO, Delvoye N, Lapointe R, Mes-Masson AM, Saad F. Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients. J Immunol Methods 2009;348:9-17.

10. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162-74.

11. Garcia AJ, Ruscetti M, Arenzana TL, et al. Erratum for Garcia et al., Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression. Mol Cell Biol 2014;34:2017-28.

12. Jamaspishvili T, Berman DM, Ross AE, et al. Clinical implications of PTEN loss in prostate cancer. Nat Rev Urol 2018;15:222-34.

13. Zhang S, Ma X, Zhu C, Liu L, Wang G, Yuan X. The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS One 2016;11:e0164514.

14. Gonda K, Shibata M, Ohtake T, et al. Myeloid-derived suppressor cells are increased and correlated with type 2 immune responses, malnutrition, inflammation, and poor prognosis in patients with breast cancer. Oncol Lett 2017;14:1766-74.

15. Liu G, Lu S, Wang X, et al. Perturbation of NK cell peripheral homeostasis accelerates prostate carcinoma metastasis. J Clin Invest 2013;123:4410-22.

16. Wensveen FM, Jelenčić V, Polić B. NKG2D: a master regulator of immune cell responsiveness. Front Immunol 2018;9:441.

17. 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.

18. Cai L, Michelakos T, Yamada T, et al. Defective HLA class I antigen processing machinery in cancer. Cancer Immunol Immunother 2018;67:999-1009.

19. Mouw KW, Goldberg MS, Konstantinopoulos PA, D’Andrea AD. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov 2017;7:675-93.

20. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8.

21. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165:35-44.

22. Strickland KC, Howitt BE, Shukla SA, 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;7:13587-98.

23. McAlpine JN, Porter H, Köbel M, et al. BRCA1 and BRCA2 mutations correlate with TP53 abnormalities and presence of immune cell infiltrates in ovarian high-grade serous carcinoma. Mod Pathol 2012;25:740-50.

24. Clarke B, Tinker AV, Lee CH, et al. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol 2009;22:393-402.

25. Chung JH, Dewal N, Sokol E, et al. Prospective comprehensive genomic profiling of primary and metastatic prostate tumors. JCO Precis Oncol 2019;3:1-23.

26. Rodrigues DN, Rescigno P, Liu D, et al. Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer. J Clin Invest 2018;128:4441-53.

27. Antonarakis ES, Isaacsson Velho P, Fu W, 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.

28. Alme AKB, Karir BS, Faltas BM, Drake CG. Blocking immune checkpoints in prostate, kidney, and urothelial cancer: an overview. Urol Oncol 2016;34:171-81.

29. Goswami S, Aparicio A, Subudhi SK. Immune checkpoint therapies in prostate cancer. Cancer J 2016;22:117-20.

30. Walunas TL, Lenschow DJ, Bakker CY, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1994;1:405-13.

31. Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res 2013;1:32-42.

32. Kwon ED, Drake CG, Scher HI, 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;15:700-12.

33. Beer TM, Kwon ED, Drake CG, 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.

34. Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 2004;4:336-47.

35. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008;26:677-704.

36. Fife BT, Pauken KE, Eagar TN, et al. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol 2009;10:1185-92.

37. Raedler LA. Opdivo (Nivolumab): second PD-1 inhibitor receives FDA approval for unresectable or metastatic melanoma. Am Health Drug Benefits 2015;8:180-3.

38. Kazandjian D, Suzman DL, Blumenthal G, et al. FDA approval summary: nivolumab for the treatment of metastatic non-small cell lung cancer with progression on or after platinum-based chemotherapy. Oncologist 2016;21:634-42.

39. Antonarakis ES, Piulats JM, Gross-Goupil M, 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.

40. Hansen AR, Massard C, Ott PA, et al. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol 2018;29:1807-13.

41. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443-54.

42. Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 2014;20:5064-74.

43. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167-75.

44. Martin AM, Nirschl TR, Nirschl CJ, et al. Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis 2015;18:325-32.

45. Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015;161:1215-28.

46. Dias A, Kote-Jarai Z, Mikropoulos C, Eeles R. Prostate cancer germline variations and implications for screening and treatment. Cold Spring Harb Perspect Med 2018;8:a030379.

47. McCabe N, Turner NC, Lord CJ, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res 2006;66:8109-15.

48. Abida W, Campbell D, Patnaik A, et al. Non-BRCA DNA damage repair gene alterations and response to the PARP inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase II TRITON2 study. Clin Cancer Res 2020;26:2487-96.

49. de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med 2020;382:2091-102.

50. Fizazi K, Piulats JM, Reaume MN, et al. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med 2023;388:719-32.

51. Clarke NW, Armstrong AJ, Thiery-vuillemin A, et al. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evidence 2022;1:EVIDoa2200043.

52. Shen J, Zhao W, Ju Z, et al. PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness. Cancer Res 2019;79:311-9.

53. Pham MM, Ngoi NYL, Peng G, Tan DSP, Yap TA. Development of poly(ADP-ribose) polymerase inhibitor and immunotherapy combinations: progress, pitfalls, and promises. Trends Cancer 2021;7:958-70.

54. Karzai F, VanderWeele D, Madan RA, 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.

55. Alva AS, Li J, Chou J, et al. Phase 2 trial of immunotherapy in tumors with CDK12 inactivation (IMPACT): results from cohort A of patients (pts) with metastatic castration resistant prostate cancer (mCRPC) receiving dual immune checkpoint inhibition (ICI). J Clin Oncol 2022;40:103.

56. López-Campos F, Gajate P, Romero-Laorden N, et al. Immunotherapy in advanced prostate cancer: current knowledge and future directions. Biomedicines 2022;10:537.

57. Movassaghi M, Chung R, Anderson CB, Stein M, Saenger Y, Faiena I. Overcoming immune resistance in prostate cancer: challenges and advances. Cancers 2021;13:4757.

58. Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological complexity of the prostate cancer microenvironment influences the response to immunotherapy. Adv Exp Med Biol 2019;1210:121-47.

59. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22.

60. Sheikh NA, Petrylak D, Kantoff PW, et al. Sipuleucel-T immune parameters correlate with survival: an analysis of the randomized phase 3 clinical trials in men with castration-resistant prostate cancer. Cancer Immunol Immunother 2013;62:137-47.

61. GuhaThakurta D, Sheikh NA, Fan LQ, et al. Humoral immune response against nontargeted tumor antigens after treatment with sipuleucel-t and its association with improved clinical outcome. Clin Cancer Res 2015;21:3619-30.

62. Warren TL, Weiner GJ. Uses of granulocyte-macrophage colony-stimulating factor in vaccine development. Curr Opin Hematol 2000;7:168-73.

63. Simons JW, Carducci MA, Mikhak B, et al. Phase I/II trial of an allogeneic cellular immunotherapy in hormone-naïve prostate cancer. Clin Cancer Res 2006;12:3394-401.

64. Rauch S, Jasny E, Schmidt KE, Petsch B. New vaccine technologies to combat outbreak situations. Front Immunol 2018;9:1963.

65. Bouard D, Alazard-Dany N, Cosset FL. Viral vectors: from virology to transgene expression. Br J Pharmacol 2009;157:153-65.

66. Madan RA, Arlen PM, Mohebtash M, Hodge JW, Gulley JL. Prostvac-VF: a vector-based vaccine targeting PSA in prostate cancer. Expert Opin Investig Drugs 2009;18:1001-11.

67. Gulley JL, Borre M, Vogelzang NJ, et al. Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J Clin Oncol 2019;37:1051-61.

68. Madan RA, Mohebtash M, Arlen PM, et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012;13:501-8.

69. Stein MN, Fong L, Mega AE, et al. KEYNOTE-046 (Part B): effects of ADXS-PSA in combination with pembrolizumab on survival in metastatic, castration-resistant prostate cancer patients with or without prior exposure to docetaxel. J Clin Oncol 2020;38:126.

70. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol 2018;15:325-40.

71. Ott PA, Hodi FS, Buchbinder EI. Inhibition of immune checkpoints and vascular endothelial growth factor as combination therapy for metastatic melanoma: an overview of rationale, preclinical evidence, and initial clinical data. Front Oncol 2015;5:202.

72. Shelley M, Harrison C, Coles B, Staffurth J, Wilt TJ, Mason M. Chemotherapy for hormone-refractory prostate cancer. Cochrane Database Syst Rev 2006;18:CD005247.

73. Redman JM, Gibney GT, Atkins MB. Advances in immunotherapy for melanoma. BMC Med 2016;14:20.

74. Rolfo C, Caglevic C, Santarpia M, et al. Immunotherapy in NSCLC: a promising and revolutionary weapon. Adv Exp Med Biol 2017;995:97-125.

75. Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A 2012;109:17561-6.

76. Adotevi O, Pere H, Ravel P, et al. A decrease of regulatory T cells correlates with overall survival after sunitinib-based antiangiogenic therapy in metastatic renal cancer patients. J Immunother 2010;33:991-8.

77. Ko JS, Zea AH, Rini BI, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res 2009;15:2148-57.

78. Graham DK, DeRyckere D, Davies KD, Earp HS. The TAM family: phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat Rev Cancer 2014;14:769-85.

79. Lemke G, Rothlin CV. Immunobiology of the TAM receptors. Nat Rev Immunol 2008;8:327-36.

80. FDA Prescribing Information - Cabometyx. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/208692s010lbl.pdf. [Last accessed on 25 Sep 2023].

81. Tripathi M, Nandana S, Billet S, et al. Modulation of cabozantinib efficacy by the prostate tumor microenvironment. Oncotarget 2017;8:87891-902.

82. Agarwal N, McGregor B, Maughan BL, et al. Cabozantinib in combination with atezolizumab in patients with metastatic castration-resistant prostate cancer: results from an expansion cohort of a multicentre, open-label, phase 1b trial (COSMIC-021). Lancet Oncol 2022;23:899-909.

83. Sharma P, Pachynski RK, Narayan V, 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.

84. Joshi H, Pinski JK. Association of ARV7 expression with molecular and clinical characteristics in prostate cancer. J Clin Oncol 2016;34:109.

85. Shenderov E, Boudadi K, Fu W, 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.

86. Boudadi K, Suzman DL, Anagnostou V, et al. Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018;9:28561-71.

87. Hotte SJ, Winquist E, Chi KN, et al. CCTG IND 232: a phase II study of durvalumab with or without tremelimumab in patients with metastatic castration resistant prostate cancer (mCRPC). Ann Oncol 2019;30:v885.

88. Aragon-Ching JB, Williams KM, Gulley JL. Impact of androgen-deprivation therapy on the immune system: implications for combination therapy of prostate cancer. Front Biosci 2007;12:4957-71.

89. Foster WR, Car BD, Shi H, et al. Drug safety is a barrier to the discovery and development of new androgen receptor antagonists. Prostate 2011;71:480-8.

90. Page ST, Plymate SR, Bremner WJ, et al. Effect of medical castration on CD4+CD25+ T cells, CD8+ T cell IFN-γ expression, and NK cells: a physiological role for testosterone and/or its metabolites. Am J Physiol Endocrinol Metab 2006;290:E856-63.

91. Gamat M, McNeel DG. Androgen deprivation and immunotherapy for the treatment of prostate cancer. Endocr Relat Cancer 2017;24:T297-310.

92. Shen YC, Ghasemzadeh A, Kochel CM, et al. Combining intratumoral Treg depletion with androgen deprivation therapy (ADT): preclinical activity in the Myc-CaP model. Prostate Cancer Prostatic Dis 2018;21:113-25.

93. Kissick HT, Sanda MG, Dunn LK, et al. Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation. Proc Natl Acad Sci U S A 2014;111:9887-92.

94. Powles T, Yuen KC, Gillessen S, 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.

95. The Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 2015;163:1011-25.

96. Hoimes CJ, Graff JN, Tagawa ST, et al. KEYNOTE-199 cohorts (C) 4 and 5: phase II study of pembrolizumab (pembro) plus enzalutamide (enza) for enza-resistant metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2020;38:5543.

97. Hodge JW, Garnett CT, Farsaci B, et al. Chemotherapy-induced immunogenic modulation of tumor cells enhances killing by cytotoxic T lymphocytes and is distinct from immunogenic cell death. Int J Cancer 2013;133:624-36.

98. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015;28:690-714.

99. Dosset M, Vargas TR, Lagrange A, et al. PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology 2018;7:e1433981.

100. Alizadeh D, Trad M, Hanke NT, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res 2014;74:104-18.

101. Vicari AP, Luu R, Zhang N, et al. Paclitaxel reduces regulatory T cell numbers and inhibitory function and enhances the anti-tumor effects of the TLR9 agonist PF-3512676 in the mouse. Cancer Immunol Immunother 2009;58:615-28.

102. Garnett CT, Schlom J, Hodge JW. Combination of docetaxel and recombinant vaccine enhances T-cell responses and antitumor activity: effects of docetaxel on immune enhancement. Clin Cancer Res 2008;14:3536-44.

103. Fizazi K, Gonzalez Mella P, Castellano D, et al. Efficacy and safety of nivolumab in combination with docetaxel in men with metastatic castration-resistant prostate cancer in CheckMate 9KD. Ann Oncol 2019;30:v885-6.

104. Sridhar SS, Kolinsky MP, Gravis G, et al. Pembrolizumab (pembro) plus docetaxel and prednisone in patients (pts) with abiraterone acetate (abi) or enzalutamide (enza)-pretreated metastatic castration-resistant prostate cancer (mCRPC): KEYNOTE-365 cohort B efficacy, safety and, biomarker results. J Clin Oncol 2020;38:5550.

105. Sartor O, de Bono J, Chi KN, et al. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med 2021;385:1091-103.

106. Chen H, Zhao L, Fu K, et al. Integrin αvβ3-targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics 2019;9:7948-60.

107. Morris MJ, Fong L, Petrylak DP, 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.

108. Sandhu SK, Joshua AM, Emmett L, et al. 577O PRINCE: interim analysis of the phase Ib study of 177Lu-PSMA-617 in combination with pembrolizumab for metastatic castration resistant prostate cancer (mCRPC). Ann Oncol 2021;32:S626-7.

109. Fu J, Shang Y, Qian Z, et al. Chimeric Antigen receptor-T (CAR-T) cells targeting Epithelial cell adhesion molecule (EpCAM) can inhibit tumor growth in ovarian cancer mouse model. J Vet Med Sci 2021;83:241-7.

110. Narayan V, Barber-Rotenberg JS, Jung IY, et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat Med 2022;28:724-34.

111. Slovin SF, Dorff TB, Falchook GS, et al. Phase 1 study of P-PSMA-101 CAR-T cells in patients with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2022;40:98.

112. Bębnowska D, Grywalska E, Niedźwiedzka-Rystwej P, et al. CAR-T cell therapy - an overview of targets in gastric cancer. J Clin Med 2020;9:1894.

113. He C, Zhou Y, Li Z, et al. Co-expression of IL-7 improves NKG2D-based CAR T cell therapy on prostate cancer by enhancing the expansion and inhibiting the apoptosis and exhaustion. Cancers 2020;12:1969.

114. Yang S, Wei W, Zhao Q. B7-H3, a checkpoint molecule, as a target for cancer immunotherapy. Int J Biol Sci 2020;16:1767-73.

115. Hummel HD, Kufer P, Grüllich C, et al. Phase I study of pasotuxizumab (AMG 212/BAY 2010112), a PSMA-targeting BiTE (Bispecific T-cell Engager) immune therapy for metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2020;38:124.

116. Tran B, Horvath L, Dorff T, et al. 609O results from a phase I study of AMG 160, a half-life extended (HLE), PSMA-targeted, bispecific T-cell engager (BiTE®) immune therapy for metastatic castration-resistant prostate cancer (mCRPC). Ann Oncol 2020;31:S507.

117. Lin TY, Park JA, Long A, Guo HF, Cheung NV. Novel potent anti-STEAP1 bispecific antibody to redirect T cells for cancer immunotherapy. J Immunother Cancer 2021;9:e003114.

118. Dorff TB, Narayan V, Forman SJ, et al. Novel redirected T-cell immunotherapies for advanced prostate cancer. Clin Cancer Res 2022;28:576-84.

119. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 2017;17:709-24.

120. Schmiel SE, Yang JA, Jenkins MK, Mueller DL. Cutting edge: adenosine A2a receptor signals inhibit germinal center T follicular helper cell differentiation during the primary response to vaccination. J Immunol 2017;198:623-8.

121. Lappas CM, Rieger JM, Linden J. A2A adenosine receptor induction inhibits IFN-γ production in murine CD4+ T cells1. J Immunol 2005;174:1073-80.

122. Ha H, Debnath B, Neamati N. Role of the CXCL8-CXCR1/2 axis in cancer and inflammatory diseases. Theranostics 2017;7:1543-88.

123. Yuen KC, Liu LF, Gupta V, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med 2020;26:693-8.

124. Calcinotto A, Spataro C, Zagato E, et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 2018;559:363-9.

125. Shorning BY, Dass MS, Smalley MJ, Pearson HB. The PI3K-AKT-mTOR pathway and prostate cancer: at the crossroads of AR, MAPK, and WNT signaling. Int J Mol Sci 2020;21:4507.

126. Conciatori F, Bazzichetto C, Falcone I, et al. Role of mTOR signaling in tumor microenvironment: an overview. Int J Mol Sci 2018;19:2453.

127. Qi Z, Xu Z, Zhang L, et al. Overcoming resistance to immune checkpoint therapy in PTEN-null prostate cancer by intermittent anti-PI3Kα/β/δ treatment. Nat Commun 2022;13:182.

128. Gao J, Ward JF, Pettaway CA, et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med 2017;23:551-5.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. Cancer Drug Resist 2023;6:656-73. http://dx.doi.org/10.20517/cdr.2023.48

AMA Style

Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. Cancer Drug Resistance. 2023; 6(3): 656-73. http://dx.doi.org/10.20517/cdr.2023.48

Chicago/Turabian Style

Sooi, Kenneth, Robert Walsh, Nesaretnam Kumarakulasinghe, Alvin Wong, Natalie Ngoi. 2023. "A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer" Cancer Drug Resistance. 6, no.3: 656-73. http://dx.doi.org/10.20517/cdr.2023.48

ACS Style

Sooi, K.; Walsh R.; Kumarakulasinghe N.; Wong A.; Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. Cancer Drug Resist. 2023, 6, 656-73. http://dx.doi.org/10.20517/cdr.2023.48

About This Article

Special Issue

© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
612
Downloads
203
Citations
3
Comments
0
8

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 3 clicks
Like This Article 8 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/