Accepted Manuscript Title: A Comprehensive Review of Immunotherapies in Prostate Cancer Authors: Manuel Caitano Maia, Aaron R. Hansen PII: DOI: Reference:
S1040-8428(17)30023-9 http://dx.doi.org/doi:10.1016/j.critrevonc.2017.02.026 ONCH 2337
To appear in:
Critical Reviews in Oncology/Hematology
Received date: Revised date: Accepted date:
13-1-2017 28-2-2017 28-2-2017
Please cite this article as: Maia Manuel Caitano, Hansen Aaron R.A Comprehensive Review of Immunotherapies in Prostate Cancer.Critical Reviews in Oncology and Hematology http://dx.doi.org/10.1016/j.critrevonc.2017.02.026 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Review Article
A Comprehensive Review of Immunotherapies in Prostate Cancer Manuel Caitano Maia1, Aaron R. Hansen2,3
1 – Department of Medical Oncology, Instituto do Câncer do Estado de São Paulo (ICESP). Av. Dr Arnaldo, 251 – Cerqueira César – CEP 01246-000. São Paulo, Brazil. 2 – Department of Medical Oncology and Hematology, Princess Margaret Hospital. 610 University Ave, Toronto, ON, Canada. 3- Department of Medicine, University of Toronto. Medical Sciences Building, 1 King's College Cir#3172, Toronto, ON, Canada. Correspondence should be addressed to: Manuel Caitano Maia, MD Division of Medical of Oncology Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina, Universidade de São Paulo. Av. Dr Arnaldo, 251, São Paulo, SP - Brazil Postal Code 01246-000 Phone: +55(11) 3893-2815; Email:
[email protected]
Highlights:
Although prostate cancer expresses many tumor associated antigens, its microenvironment is relative immunosuppressive. Sipuleucel-T is the only approved immunotherapy for prostate cancer and has driven major enthusiasm for testing new agents in this disease. Various other immune agents have been tested but failed to show clinical benefit in prostate cancer. Appropriate patient selection and trial design are crucial; and need to be tailored to account for the unique pharmacodynamics and clinical outcomes of immunotherapies. Here we review the data on completed trials and conclude with future directions, highlighting important aspects that need to be addressed to improve the evaluation of immunotherapies in prostate cancer.
Abstract Prostate cancer is the second most common malignant neoplasm in men worldwide and the fifth cause of cancer‐related death. Although multiple new agents have been approved for metastatic castration resistant prostate cancer over the last decade, it is still an incurable disease. New strategies to improve cancer control are needed and agents targeting the immune system have shown encouraging results in many tumor types. Despite being attractive for immunotherapies due to the expression of various tumor associated antigens, the microenvironment in prostate cancer is relatively immunosuppressive and may be responsible for the failures of various agents targeting the immune system in this disease. To date, Sipuleucel‐T is the only immunotherapy that has shown significant clinical efficacy in this setting, although the high cost and potential trial flaws have precluded its widespread incorporation into clinical practice. Issues with patient selection and trial design may have contributed to the multiple failures of immunotherapy in prostate cancer and provides an opportunity to tailor future studies to evaluate these agents more accurately. We have reviewed all the completed immune therapy trials in prostate cancer and highlight important considerations for the next generation of clinical trials.
Keywords: prostate cancer; metastatic; castration‐resistant; immunotherapy; immune checkpoint inhibitor; vaccines; oncolytic viruses
Introduction Prostate cancer (pCa) is the second most common cancer in men, with an estimated 1.1 million patients diagnosed worldwide yearly.1 Furthermore it is the fifth leading cause of cancer mortality in men accounting for 6.6% of total male deaths.1 The majority of new cases are localized or locally advanced, with 20% to 30% of these patients relapsing after curative intent therapy.2, 3 Less than 5% of patients present with de novo metastatic pCa.2 Eight clinical states of pCa have been described and metastatic disease is divided into hormone sensitive (HSPC) and castrate refractory
(CRPC) settings.4 For men with metastatic CRPC (mCRPC), the median overall survival (OS) in recent phase 3 studies has ranged from 12.2 to 34.7 months.5‐10 Table 1 outlines the various treatment options for mCRPC with hormonal, chemotherapy, radiopharmaceutical and immunotherapy agents. These therapies have demonstrated significant improvements in overall survival but ultimately metastatic pCa currently remains incurable. Targeting the immune system represents an appealing option for the development of anticancer treatment. There are several classes of immune therapies such as immune checkpoint inhibitors, co‐stimulatory antibodies, vaccines, adoptive cell transfer, tumor infiltrating lymphocytes, oncolytic viruses and cytokines. The cancer vaccine sipuleucel‐T has been approved for use in mCRPC by the US Food and Drug Administration (FDA) and recent early phase trials of programmed cell death protein‐1 (PD‐1) inhibitors have reported promising activity, which support the enthusiasm to develop immune therapies in pCa.5, 11, 12 Notwithstanding there have been several notable immunotherapy failures in pCa and correlative studies have demonstrated that the prostate tumor microenvironment is predisposed toward immunosuppression.13‐15 Here we review immunotherapies that have been tested in pCa, highlighting the spectrum of new agents and combinations. We also examine potential biomarkers and aspects of clinical trial design for immunotherapies.
Cancer and the immune system
Tumors occur in the setting of a dysregulated immune system.16 The immune system provides several protective mechanisms such as removing viruses that can induce tumor formation, suppressing tumorigenic inflammatory reactions and eliminating tumor cells. Cancer initiation occurs following oncogenic cellular transformation and failure of intrinsic tumor suppressor processes. Beyond these events the cancer immunoediting concept describes 3 phases that regulate the immune system‐tumor interaction.17
The elimination phase involves innate and adaptive immunity removing tumor cells. Those tumor cells that remain enter a state of quiescence and exist in equilibrium with adaptive immune cells. Over time under the ongoing pressure applied by the immune system, tumor cells escape the equilibrium to proliferate unchecked without regulation by immune cells.18 Tables 2 and 3 summarize the described concepts. The anti‐tumor immune response is cyclical, starting with recognition of tumor neo‐ antigens by specialized antigen‐presenting cells (APCs). Subsequently, APCs present antigen to effector T cells and when this occurs in the setting of an appropriate secondary signal, it leads to activation of T cells that then migrate to the tumor microenvironment where they remove cancer cells expressing those antigens.19 However, an intricate network of stimulatory and inhibitory signals regulates this response that will ultimately produce ongoing immune cell activation or suppression within the resultant infiltrate and has been associated with patient prognosis. 20‐24 The type, density and spatial distribution of infiltrating lymphocytes are strongly correlated with survival,25‐27 which supports the development of immune therapies to enhance antitumor immune responses.28 Figure 1 summarizes the immunoediting process and the mechanisms of action of immune agents tested in pCa.
Prostate Cancer Microenvironment Observations about the pCa microenvironment suggest that it is predominantly immunosuppressive. The findings that support this include a low cytolytic activity of NK cells within prostate tumor bed,14 higher secretion of TGF‐beta by prostate tissue, which inhibits NK cell and lymphocyte function,29 and the recruitment and accumulation of T regulatory cells (Tregs) and TH17 lymphocytes that down‐regulate antitumor immunity.30, 31 Levels of TGF‐beta in prostate tissue are associated with high gleason scores, higher
pathologic tumor stage and increased likelihood of post‐operative residual tumors in localized pCa.32 Furthermore in the metastatic setting, TGF‐beta concentrations are correlated with tumor burden.33 The prostate immune microenvironment is dynamic, changing over time, clinical states and with treatment exposure. The latter is characterized by a series of phenotypic alterations leading to immunomodulation34 such as increased tumor infiltrating lymphocytes (TIL) in the prostate bed following androgen deprivation therapy (ADT)35, 36 or sensitization of tumor cells to T‐cell mediated lysis following enzalutamide and abiraterone exposure,34 and higher levels of PD‐1 ligand (PD‐L1) and PD‐L2 expression on enzalutamide resistant prostate cancer cells.37
Immunotherapy in Prostate Cancer Immunotherapy is focused on agents developed to harness the host’s immune system to target and destroy malignant cells. It may involve various immune mechanisms, such as stimulating recognition and elimination of non‐self antigens, augmenting or propagating the antigen presentation process, priming T cells, enhancing T cell mediated lysis, B lymphocyte activation and the production of humoral responses. There are some characteristics that make pCa attractive for immunotherapy.33 The tumor microenvironment has many specific tumor associated antigens (TAAs) such as prostate specific antigen [PSA], prostate acid phosphatase [PAP] and prostate‐ specific membrane antigen [PSMA]. In addition, a proportion of tumors have slow growth kinetics which provides sufficient time for the immune system to mount an anti‐tumor response.38, 39 Here we discuss different classes of immunotherapy in pCa, involving antigen‐specific vaccination, immune checkpoint inhibitors and immunomodulators. In general, antigen‐specific vaccination can be divided in two major categories: passive or active. Passive vaccination involves the transference of immune effectors already activated (primed) to kill cancer cells developed ex vivo. Active vaccination is based on agents capable of generating active T effector cells against TAAs that will
lead to cell lysis as well as recruiting and or regulating other inflammatory cells.33, 40 Active vaccination is the main focus of the next section and is subdivided in two broad components: cell‐based and vector‐based approaches. Details are provided in Table 4.
Cell‐based Vaccines Cell‐based vaccination consists of autologous or allogenic whole cells that are modified in order to induce anti‐tumor immune responses. Autologous vaccines activate and prime the host’s immune cells for re‐infusion, while allogenic cell vaccinations are developed by culturing tumor cell lines ex‐vivo with immunological stimulators before infusion into patients. Autologous Vaccines Sipuleucel‐T is an autologous vaccine which is a personalized immunotherapy agent processed following peripheral dendritic cell (DC) collection via leukapheresis which is then incubated in GCS‐F and PAP (PA2024) protein.5, 40 After a 36 to 44 hour period, the primed DC are re‐infused into the patient in order to generate a PAP‐ specific CD4+ and CD8+ T cell response.33 Sipuleucel‐T was the first vaccine approved as a cancer treatment and was based on three phase III clinical trials that assessed its efficacy in the mCRPC scenario, mainly in patients with asymptomatic or minimally symptomatic disease with no visceral metastasis. The IMPACT trial enrolled 512 patients and treated them with 4 cycles of sipuleucel‐T. This study demonstrated an improvement of 4.1 months in OS compared with placebo, with a 22% reduction of the risk of death. Of note, no difference was noted in time to objective disease progression, which was secondary endpoint. Also, no statistically significant difference in PSA response was shown. The reasons for not improving time to progression (TTP) are not fully understood, but may be related to the delayed onset of anti‐tumor immune response after completion of immunotherapy 5 and is consistent with the findings from other phase
3 trials in this setting.41, 42 Safety data showed the treatment was overall well tolerated with no high frequency severe adverse events. 43 However, some authors have called attention to the fact that the improved OS seen may have been related to excess harm in the control arm due to leukapheresis, especially in the elderly population (≥65 years), since this intervention differed between groups. Of note, the placebo group had most of their circulating lymphocytes withdrawn but only received back a small portion of them. Furthermore, while ex vivo these lymphocytes were exposed to conditions that may have resulted in lysis or rendered them non‐functional.44 In addition, the elderly population had a lower than expected survival, possibly due to the harmful intervention.44 Immunologic assessment of patients treated in the IMPACT trial showed that those who had higher antibody titers (above 400) against PA2024 benefited the most from treatment, with a higher overall survival.5 Perhaps even more interesting is the presence of secondary antigen spread (release of non‐targeted antigens) after sipuleucel‐T treatment, inducing IgG humoral responses to non‐targeted antigens released after tumor cell lysis. This phenomena may act as a surrogate for longer OS.45 An increased benefit was also shown in subgroups of earlier disease states with less aggressive features like lower PSA baseline values, gleason scores, lactate dehydrogenase (LDH) and alkaline phosphatase.5, 46, 47 Despite its efficacy and good safety profile, the use of sipuleucel‐T has not been widely adopted mainly due to the high level of required resources and infra‐ structure and lack of cost‐effectiveness. 48, 49 Allogenic Vaccines GVAX (BioSante) is another type of cell‐based vaccine, which is composed of both castrate‐sensitive and castrate resistant allogenic prostate cancer cell lines (LNCaP and PC3, respectively) and transduced with a replication defective retrovirus genetically modified to bear GM‐CSF, resulting in APC recruitment to the injection site.50, 51 Although an initial phase 1/2 trial showed promising results,52 two phase 3
trials (VITAL‐1 and VITAL‐2) failed to show improved outcomes and were closed early due to futility. VITAL‐1 trial randomized patients with asymptomatic mCRPC (n=626) to GVAX or docetaxel‐prednisone and was terminated early after an interim analysis showed a likelihood of less 30% to meet its primary endpoint of OS.53 VITAL‐2 trial compared GVAX alone with GVAX plus docetaxel/prednisone in symptomatic mCRPC patients (n=408) and also closed early due to an increased death rate among patients assigned to the intervention‐group.54 These trials had several flaws, which may have contributed to the disappointing outcomes. GVAX had not been compared with a placebo control and thus moving forward with a trial comparing it with chemotherapy in VITAL‐1 was premature. Furthermore, the recommended phase 2 dose for the combination of GVAX and docetaxel had not been determined prior to VITAL‐2. 33, 55
Vector‐based Vaccines These consist of genetically engineered nucleic acids that encode specific TAAs transmitted by vectors such as bacterial plasmids or viruses. DNA‐based vaccines DNA‐based vaccines can be incorporated by host cells and generate an immune response by recruiting APCs.56 Several early phase trials of various DNA vaccines alone or in combination with cytokines and growth factors have been tested in different prostate cancer settings. In general these agents have modulated the immune system leading to T cell and humoral changes. In addition these vaccines have had an anti‐tumor effect as evidenced by slowing the PSA doubling time. The most studied DNA vaccine in prostate cancer to date has been pTVG‐HP, a plasmid, which encodes PAP protein. It has been tested in phase I and II trials and shown to generate PAP‐specific T cell responses in men with biochemically recurrent prostate cancer, resulting in an increase in PSA doubling time and long‐term T cell responses.57, 58 A phase I/II trial in non‐metastatic CRPC patients with pTVG‐HP showed an increased PSA doubling time from 6.5 months to 9.3 months after 1 year
follow‐up after treatment.58 Viral‐vector based vaccines The mechanism of action involves infection of epithelial cells that when lysed liberate antigens that will be taken up by APCs and presented to CD4+ and CD8+ T cells, generating the immune response. In order to increase the immunogenicity, three co‐stimulatory molecules have been incorporated (B7.1, ICAM‐1 and LFA‐3, also known as TRICOM).59,60 PROSTVAC‐VF is a PSA‐target pox‐virus‐based vaccine that has been developed using a semi‐heterologous prime‐boost strategy where sequential administration of a vaccinia virus prime followed by a recombinant fowlpox‐PSA virus boost is performed. Fowlpox vectors have demonstrated sustained immune response after an initial priming event with vaccinia‐based vaccines since they do not yield late viral gene products and thus do not elicit significant amounts of host antibodies.61 PROSTVAC‐VF has been shown to increase PSA progression‐free survival in 63% of patients for more than 6 months as well as significantly slowing the PSA doubling time from 5.3 months to 7.7 months in a phase II study in non‐metastatic pCa patients.62 In another phase II study with PROSTVAC‐VF, 125 patients with minimally symptomatic mCRPC were enrolled and randomized to receive the vaccine or placebo. Although the study was negative for its primary endpoint (PFS), OS after 3 years of follow up was significantly increased by 8.5 months (25.1 vs 16.6 months; HR 0.56; p=0.0061).63 A subsequent phase III study of PROSTVAC‐VF has completed accrual and final results are awaited (NCT01322490).64
Adenovirus type 5 (Ad5) has also been used as vector in pCa. A phase I trial tested Ad5‐PSA in mCRPC and reported 34% of patients produced PSA antibodies, 68% produced an anti‐PSA T cell responses and increased PSA doubling time in 48% of the treated patients,65 leading to a phase II trial that is currently underway (NCT00583024).66
Personalized peptide Vaccination (PPV) To overcome the challenges of inter‐patient immunological diversity, PPV identifies
peptides that are recognized by the highest frequency of precursor cytotoxic T lymphocytes (CTL) for each individual. These peptides bind to MHC‐class I antigens and when administered induce CTL activation and subsequent anti‐tumor response. The safety of PPV has been tested in several phase I trials with the most common toxicity being an injection‐site reaction.67 Further testing in a phase II trial of HLA‐ A2+ or HLA‐A24+ in metastatic CRPC was undertaken by randomizing patients to either PPV plus low‐dose estramustine or standard dose estramustine alone. The trial demonstrated an increase in PFS (primary endpoint) from 2.8 months to 8.5 months with HR of 0.28 (95% CI, 0.14–0.61; log‐rank P = 0.0012) in the PPV group.68 A recently published phase 2 trial of PPV plus metronomic low‐dose cyclophosphamide compared with PPV alone showed no significant differences in either PFS or OS, although patients who developed a positive immune response showed a longer OS regardless of treatment arm.69 A phase III, randomized, placebo‐ controlled trial testing PPV in HLA‐A24+ patients is underway in Japan (UMIN000011308), with an aim to recruit 333 docetaxel‐refractory mCRPC patients.
Immune checkpoint inhibition Checkpoint inhibitors are antibodies that target regulatory or co‐inhibitory molecules, such as cytotoxic T‐lymphocyte associated protein 4 (CTLA‐4), PD‐1 and its ligand (PD‐L1). These checkpoints typically down‐regulate the immune system by inhibiting T‐cell activation and promoting tolerance.38
Anti‐CTLA‐4 Ipilimumab is a monoclonal antibody that blocks CTLA‐4. A phase III clinical trial of 799 metastatic CRPC patients who had progressed on docetaxel‐chemotherapy were randomized to ipilimumab or placebo after bone‐directed radiotherapy (8Gy in one fraction). No significant difference was noted in the primary endpoint of OS (11.2 months vs 10 months; HR 0.85; p=0.053), but a modest benefit was observed in PFS with ipilimumab over placebo (4.0 vs 3.1 months, respectively; HR 0.70, p < 0.0001). Patients in the ipilimumab arm more frequently had greater than 50% reduction in the PSA (13.1% vs 5.3%). Additionally, a post‐hoc analysis of pre‐defined subgroups
showed a greater benefit in patients with more favorable prognostic factors, such as alkaline phosphatase concentration of less than 1.5 times ULN, a hemoglobin of 110 g/L or higher and no visceral metastases. In this group, median OS was 22.7months with ipilimumab and 15.8 months with placebo (HR 0.62; 95%CI 0.45–0.86; p=0.0038).70 The higher clinical benefit found in favorable subgroups has been already shown in other immunotherapy trials and reasons may involve a lower tumor burden and a less immunosuppressive tumor microenvironment. Also, slow growing tumors may be more likely to mount an anti‐tumor immune response.71, 72 Failure to improve OS in the overall population could have resulted from a lack of efficacy of ipilimumab or a sub‐optimal schedule that combined radiation with ipilimumab. Preclinical studies have reported that higher radiation doses as well as fractionation may be preferable when combining with immune checkpoint therapies.73‐76 A phase 3 trial assessing ipilimumab in the chemotherapy‐naïve mCRPC setting randomized a total of 400 patients to ipilimumab 10mg/kg or placebo. Median OS was not significantly different between arms (28.7mo vs 29.7mo; HR 1.11; p=0.37). However, ipilimumab‐treated patients derived a modest PFS benefit of approximately 2 months (5.6mo x 3.8mo; HR 0.67; P < 0.05) and a higher PSA response (23% vs 8%; p value not reported).77 Interestingly, this trial did not corroborate the subgroup analysis from the radiation and ipilimumab trial, whereby patients with favorable prognostic factors did not perform better with the immune therapy compared with placebo.
Anti‐PD1/PDL‐1 The T‐cell surface molecule PD‐1 interacts with its ligand PD‐L1 (or B7‐H1) leading to T cell inhibition. Studies have demonstrated that higher expression of PD‐L1 in tumor infiltrating lymphocytes is associated with poor survival.78 Blocking this interaction enhances anti‐tumor immune response. Agents targeting the PD‐1 pathway, such as nivolumab, pembrolizumab and more recently PD‐L1 inhibitors such as atezolizumab, have received regulatory approval in other tumor types, including
melanoma, renal cell, lung and bladder cancer.79‐82 Currently, there are many clinical trials underway exploring the role of pembrolizumab either alone or in combination with other immune therapies, such as vaccines or cryosurgery, in HSPC and mCRPC (NCT02312557, NCT02499835, NCT02489357, NCT02787005), as well as a combination of nivolumab and ipilimumab in mCRPC (NCT02601014). Preliminary findings from a phase 1b study with pembrolizumab in heavily pretreated PD‐L1 positive advanced prostate cancer patients reported an ORR of 13%, with a median duration of response (DOR) of 59 weeks and stable disease (SD) reached in 39% of enrolled patients.11 A phase II study of pembrolizumab in combination with enzalutamide in mCRPC patients upon progression on enzalutamide alone showed a PSA decline of ≥50% in 20% of the patients and some of them remained progression‐free for up to 60 weeks.12
Immunomodulators Immunomodulatory agents that target the tumor microenvironment have also been explored in pCa. Tasquinimod (ABR‐215050; Active Biotech, Lund, Sweden), is a second‐generation quinoline‐3‐carboxamide that blocks the immunomodulatory protein S100A9, which plays a key role in the function of regulatory myeloid cells. This drug has been shown to have anti‐tumor activity in preclinical models and clinical efficacy in randomized double blind placebo controlled trial in pCa.83 By modulating regulatory myeloid cells, it decreases immunosuppression and angiogenesis in the tumor microenvironment, preventing metastatic spread.84 In a phase II study with 206 asymptomatic or minimally symptomatic, chemotherapy‐naïve mCRPC patients assigned to tasquinimod or placebo, disease progression was significantly delayed (the primary endpoint) from 3.3 to 7.6 months (p=0.0042) favoring the experimental therapy. Tasquinimod was reported to have an acceptable toxicity profile.85 In a long‐term survival analysis of this trial, there was a trend for longer OS in the subgroup with bone only metastasis [34.2 versus 27.1 months (P = 0.19; HR, 0.73; 95% CI, 0.46–1.17)].86 In this regard, a phase III trial was
initiated which corroborated the improvement in radiographic PFS found in the phase II trial (7.0 vs 4.4 months; HR 0.64; p=0.001), but it failed to demonstrate an improvement in OS (21.3 months with tasquinimod vs 24 months with placebo; HR 1.10; p=0.25).87
Future Directions To date most immunotherapies have yielded disappointing results in pCa in comparison with other tumor types. There are two potential strategies to improve these results. Firstly, the development of more effective immune therapies or rational combinations of immune treatments. Secondly, to better select patients for these treatments using biomarkers.
Combination Immune Therapies The purpose of combination immune therapies is to enhance anti‐tumor T cell responses. Combinations may include dual immune therapies or immune treatments with chemotherapy, hormonal therapy, targeted therapy, radiation or surgery. Table 5 summarizes current ongoing clinical trials testing combinations involving immunotherapies. Androgen ablation has multiple immune effects and can modulate cancer cell sensitivity to T cells, regulate apoptotic genes34 and induce thymus regeneration, causing efflux of new and naive T cells.88 Other data have shown that ADT may mitigate self‐tolerance89 and eliminate tumor cells. Agents targeting the androgen pathway, such as enzalutamide, can modulate the immune system to render pCa cells more sensitive to immune‐mediated attack.90 In light of enzalutamide’s immunomodulatory capacity, it is an attractive combination with other immune treatments. A combination of enzalutamide and a vaccine targeting TWIST (an antigen involved in epithelial to mesenchymal transition and metastasis) has already shown promising results in pre‐clinical models.90 Clinical trials exploring this effect are underway with enzalutamide plus the vaccine
PROSTVAC/TRICOM in mCRPC (NCT01867333) and in non‐metastatic castration‐ sensitive pCa (NCT01875250). Based on preclinical studies showing that immunotherapy may have improved efficacy when given before androgen ablation,91 abiraterone, a cyp‐17 lyase inhibitor, is being evaluated in conjunction or in sequence with Sipuleucel‐T (NCT01487863). A phase II study reported the feasibility of this combination in spite of the requirement for steroids during abiraterone use.92 Similarly, other combinations of immunotherapies and androgen pathway targeted agents are being studied, such as sipuleucel‐T with ADT (NCT0141391) and ketoconazole in conjunction with the 177 lutetium PSMA labelled with the monoclonal antibody J591 (177Lu‐PSMA‐J591) (NCT00859781). Preclinical data have reported that cytotoxic treatments, such as chemotherapy and radiation, can produce regulatory T cell inhibition, effector T and B cell activation and TAA release leading to immunogenic cell death.93‐95 These findings have led to the development of clinical trials evaluating various cytotoxic combinations: neoadjuvant low‐dose cyclophosphamide followed by GVAX and ADT compared to ADT alone in localized pCa patients before prostatectomy (NCT01696877); standard docetaxel/prednisone chemotherapy in combination with increasing doses of the anti‐PSMA
mAb
177Lu‐J591
in
mCRPC
patients
(NCT00916123);
docetaxel/prednisone alone for up to 12 cycles or the same regimen preceded by 12 weeks of ProstVac/PSA‐TRICOM in slowly progressing mCRPC patients (NCT01145508). Immune therapies combined with other immune agents are being tested in multiple trials. A phase I dose‐escalation trial of ipilimumab and a fixed dose of GVAX in chemotherapy‐naïve mCRPC patients reported results showing this combination was safe and well‐tolerated. Approximately 25% of patients were reported to have a PSA decline of ≥50%.96 Another phase I dose‐escalation trial assessed the combination of ipilimumab and a poxviral vaccine (with PSA‐TRICOM) in 30 mCRPC patients (24 of them were chemo‐naïve). The combination was well‐tolerated, with no increased
immune‐related toxicities compared to ipilimumab alone. Of note, 14 (58%) of the 24 patients who were chemotherapy‐naïve had a PSA decline, 6 of whom had ≥50% reduction.97 Other combinations of PD‐1 checkpoint inhibitors with chemotherapy, targeted therapy and hormonal therapy are in development in mCRPC (NCT02861573).
Biomarkers To date no approved therapy for prostate cancer is selected due to the presence or absence of a molecular aberration. While not related currently to immune therapy, BRCA abnormalities are being utilized to select patients with mCRPC for PARP inhibitor treatment.98 The development of predictive biomarkers that identify patients most likely to respond to immunotherapy is an area of active research. A summary of studied immune biomarkers is provided in table 6.
Snyder and colleagues reported that neo‐epitopic signatures predicted anti‐tumor response to CTLA‐4 blockade with ipilimumab or tremelimumab. According to the findings, patients with prolonged benefit from anti‐CTLA‐4 agents shared common neo‐epitopes produced by somatic mutations in tumors.99 Other data have demonstrated that high mutational load can be associated with increased benefit from checkpoint inhibitors.100‐102 Another potential predictive biomarker that has been explored in various solid tumors includes tumor cell or immune cell PD‐L1 expression. Several studies have reported that patients with tumor samples that express PD‐L1 have an increased benefit from anti‐PD‐1 or anti‐PD‐L1 therapies.82, 103‐105 In pCa the role of PD‐L1 expression has not been defined in relation to treatment selection. Furthermore issues surrounding how PD‐L1 positivity is defined, which tissue should be tested (primary versus metastasis) and which assay should be used remains unresolved.106‐ 109
The search for immune biomarkers in pCa is an active area of research and to date no definitive biomarkers have been found. Nevertheless, several markers appear promising. Most studies have focused on measuring immunological activation as surrogates for clinical benefit, certain genomic alterations like expression of HLA‐ DRB1*11 or HLA‐A*24 alleles,110 and humoral antigen spread (an IgG response to secondary antigens) after treatment exposure.45 Although most biomarkers used to date are based on measuring CTL responses to specific TAAs, it is important to note that using immunological activation as surrogates for clinical benefit is currently limited, since assays are still unreliable because of lack of reproductibility between laboratories.111,
112
In addition,
evaluating immune parameters in response to a specific TAA may not capture the role of epitope spreading or dynamic changes over time and with treatment exposure.113 The acquisition of serial biopsies is not practical, since patients may not have accessible tumors, may not be willing to do repeated biopsies and not all institutions have radiology services who can perform these investigations. Cost considerations involving all the aforementioned issues must also be taken into account.
Considerations for clinical trial design for immunotherapy in prostate cancer Duration of therapy Currently the optimal duration of immune therapy is not known. Different trials have either treated patients until progression, or for a set period of time for example 1 to 2 years. Addressing this in a clinical trial setting is a challenge and apart from Checkmate‐153 (NCT02066636) few studies are designed to answer this question. Such a trial will take a long time to complete and will require a large number of patients.
Endpoints Traditional endpoints used to evaluate chemotherapy may not assess accurately the clinical benefit from immune therapies. This could reflect that the mechanism of
action of most immune therapies is not directly cytotoxic. Developing an immune response can take several weeks and thus clinical changes may not be observed until that has occurred. Thus TTP and PSA endpoints may not be appropriate measures of benefit in clinical trials.33 In recent clinical trials of immune checkpoint inhibitors, an OS benefit was seen irrespective of PFS improvements. The phase III trial of nivolumab in metastatic renal cell carcinoma patients and the phase II study of atezolizumab in bladder cancer are prime examples.81, 82 In the phase III trial of sipuleucel‐T and the phase II trial of ProstVac VF despite no difference in PFS or TTP, an improvement in OS favoring the experimental arm was reported. While OS is the gold standard endpoint for any anticancer therapy, there are limitations with this endpoint especially in the first line metastatic setting where subsequent treatments are likely to be administered and thus dilute differences between the arms of the trial. Logistical considerations such as duration of study and cost must be accounted for given that OS endpoints typically prolong the trial time and thus increase expense. Ideally surrogate or intermediate endpoints utilizing clinical or pharmacodynamics biomarkers of early response or benefit could be used if they were validated. The ICECaP initiative is an international, multi‐institutional collaboration that plans to identify intermediate clinical endpoints in patients following radiation or prostatectomy.114 While this may not be applicable to pCa patients on immune therapies, similar efforts are needed in patients with metastatic CRPC to aid in clinical trial design. The conventional statistical considerations that underpin a trial may also need to be reviewed. A delayed separation of OS curves does not comport with a proportional hazards model and may reduce the power to detect differences between treatment arms.115 As a result, designing clinical trials with hazard ratios as a function of time, clearly separating them before and after curves divert could provide a more appropriate assessment of new immunotherapies in clinical trials.
Response Assessment RECIST criteria does not account completely for the different responses that can be
observed with immunotherapy. To address this an immune‐related response criteria known as ir‐RECIST was developed in order to assess response to immunotherapy.116 These criteria account for a variety of responses that range from an initial increase in tumor burden before shrinkage, to a reduction in target lesions, or slow, steady decline in total tumor volume, or reduction in total tumor burden after the appearance of new lesions. This revised response criteria is thought to provide a better evaluation of treatment response.117
Correlative studies Early phase clinical trials should aim at establishing dose and safety in addition to providing proof of principle of mechanism of action and an understanding of the biological impact of the treatment and immune modulation. Additionally, immunological biomarkers that are surrogates for OS are needed, since OS analysis requires several years. Reproducible and reliable assays are needed to develop effective immune biomarkers that can be used in clinical trials. Barriers to such biomarkers and correlative studies include cost, laboratory expertise, need for serial sampling of tumor tissue or blood and analytical validation of assays.
Patient Selection Ideal candidates for immunotherapy trials in pCa are those with early disease states, low tumor burden and slow growing tumors, who have sufficient time to mount an anti‐tumor immune response. Traditionally experimental therapies are often first tested in patients with advanced treatment refractory disease. Presumably the immune tumor interaction changes as the disease state changes which must be factored in when evaluating these therapies. Studies conducted in renal cell carcinoma, colorectal and lung cancers have provided evidence of the impact of disease stage in clinical benefit from vaccines, showing better outcomes in subgroups of less advanced disease.118‐120 In pCa, a greater benefit was seen with PROSTVAC‐VF vaccine among patients with less aggressive disease as predicted by the Halabi nomogram.72 Similarly, sipuleucel‐T provided a higher clinical benefit in early disease states and lower tumor burden.5, 121, 122
Greater tumor burden is associated with a more immunosuppressive microenvironment, with greater inhibitory effect on the immune system.113, 123 Patient selection for immunotherapy trials is challenging, given patients with early disease states have multiple effective treatment options. Possible strategies to circumvent this issue include administration of agents before a local treatment is applied (such as surgery or radiation) in a window of opportunity approach, focusing on immunological or pathological endpoints that could act as surrogates for clinical benefit for future clinical trials such as with the ICECaP initiative.
Conclusion Efficacious immunotherapies will need to overcome the immunosuppressive milieu in the tumor microenvironment in patients with pCa. Our understanding of the complex interactions between the immune system and pCa microenvironment is ever increasing, which may identify new targets for immunotherapy. Many clinical trials are exploring different combinations of immune therapies with cytotoxic agents, androgen pathway modulators, radiotherapy and targeted drugs, to investigate synergistic effects between these agents and define the appropriate sequence and dose. The design of clinical trials of immune treatments must also be tailored to assess more accurately the true effect of immunotherapies. Addressing these issues will permit the successful development and subsequent approval of immunotherapies , broadening the arsenal of treatment approaches for pCa. Funding: This research did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors.
References 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7‐30. 2. Howlader N NA, Krapcho M, Miller D, Bishop K, Altekruse SF, Kosary CL, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA SEER Cancer Statistics Review, 1975‐2013, National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/csr/1975_2013/, based on November 2015 SEER data submission, posted to the SEER web site, April 2016. 3. Abdollah F, Schmitges J, Sun M, Jeldres C, Tian Z, Briganti A, et al. Comparison of mortality outcomes after radical prostatectomy versus radiotherapy in patients with localized prostate cancer: a population‐based analysis. Int J Urol. 2012;19:836‐44. 4. Scher HI, Solo K, Valant J, Todd MB, Mehra M. Prevalence of Prostate Cancer Clinical States and Mortality in the United States: Estimates Using a Dynamic Progression Model. PLoS ONE. 2015;10:e0139440. 5. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel‐T immunotherapy for castration‐resistant prostate cancer. The New England journal of medicine. 2010;363:411‐22. 6. Berthold DR, Pond GR, Soban F, De Wit R, Eisenberger M, Tannock IF. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: Updated survival in the TAX 327 study. J Clin Oncol. 2008;26:242‐5. 7. De Bono JS, Oudard S, Ozguroglu M, Hansen Sr, MacHiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration‐resistant prostate cancer progressing after docetaxel treatment: A randomised open‐label trial. The Lancet. 2010;376:1147‐54. 8. Parker C, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fossa SD, et al. Alpha emitter radium‐223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369:213‐23. 9. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. PREVAIL Enzalutamide in Metastatic Prostate Cancer before Chemotherapy. The New England journal of medicine. 2014:1‐10. 10. Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PF, Sternberg CN, et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy‐naive men with metastatic castration‐resistant prostate cancer (COU‐AA‐302): final overall survival analysis of a randomised, double‐blind, placebo‐controlled phase 3 study. Lancet Oncol. 2015;16:152‐60. 11. A. Hansen, C. Massard, P.A. Ott, N. Haas, J. Lopez, S. Ejadi, J. Wallmark, B. Keam, J‐P. Delord, R. Aggarwal, M. Gould, P. Qiu, S. Saraf, S. Keefe, S.A. Piha‐Paul. Pembrolizumab for patients with advanced prostate adenocarcinoma: Preliminary results from the KEYNOTE‐028 study. Annals of Oncology. Volume 27, 2016 Supplement 6. Abstract 725PD. 12. J.N. Graff, J.J. Alumkal, C.G. Drake, G.V. Thomas, W.L. Redmond, M. Farhad, R. Slottke, T.M. Beer. First evidence of significant clinical activity of PD‐1 inhibitors in metastatic, castration resistant prostate cancer (mCRPC). Annals of Oncology. Volume 27, 2016 Supplement 6. Abstract 719O. .
13. Zhang Q, Liu S, Zhang Q, Xiong Z, Wang AR, Myers L, et al. Interleukin‐17 promotes development of castration‐resistant prostate cancer potentially through creating an immunotolerant and pro‐angiogenic tumor microenvironment. Prostate. 2014;74:869‐79. 14. Pasero C, Gravis G, Guerin M, Granjeaud S, Thomassin‐Piana J, Rocchi P, et al. Inherent and Tumor‐Driven Immune Tolerance in the Prostate Microenvironment Impairs Natural Killer Cell Antitumor Activity. Cancer Res. 2016;76:2153‐65. 15. Shafer‐Weaver Ka, Anderson MJ, Stagliano K, Malyguine A, Greenberg NM, Hurwitz Aa. Cutting Edge: Tumor‐specific CD8+ T cells infiltrating prostatic tumors are induced to become suppressor cells. Journal of immunology (Baltimore, Md : 1950). 2009;183:4848‐52. 16. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. 2011. p. 646‐74. 17. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329‐60. 18. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol. 2006;90:1‐50. 19. Chen DS, Mellman I. Oncology meets immunology: the cancer‐immunity cycle. Immunity. 2013;39:1‐10. 20. Clemente CG, Mihm MC, Jr., Bufalino R, Zurrida S, Collini P, Cascinelli N. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer. 1996;77:1303‐10. 21. Predina J, Eruslanov E, Judy B, Kapoor V, Cheng G, Wang LC, et al. Changes in the local tumor microenvironment in recurrent cancers may explain the failure of vaccines after surgery. Proc Natl Acad Sci U S A. 2013;110:E415‐24. 22. Pages F, Berger A, Camus M, Sanchez‐Cabo F, Costes A, Molidor R, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353:2654‐66. 23. Schreiber RD, Old LJ, Smyth MJ. Cancer Immunoediting: Integrating Immunity’s Roles in Cancer Suppression and Promotion. Science. 2011;331:1565‐70. 24. Fridman WH, Pages F, Sautes‐Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12:298‐306. 25. Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58:3491‐4. 26. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor‐infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538‐43. 27. van Houdt IS, Sluijter BJ, Moesbergen LM, Vos WM, de Gruijl TD, Molenkamp BG, et al. Favorable outcome in clinically stage II melanoma patients is associated with the presence of activated tumor infiltrating T‐lymphocytes and preserved MHC class I antigen expression. Int J Cancer. 2008;123:609‐15. 28. Yuan J, Page DB, Ku GY, Li Y, Mu Z, Ariyan C, et al. Correlation of clinical and immunological data in a metastatic melanoma patient with heterogeneous tumor responses to ipilimumab therapy. Cancer Immun. 2010;10:1.
29. Flavell RA, Sanjabi S, Wrzesinski SH, Licona‐Limón P. The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol. 2010;10:554‐67. 30. Miller AM, Lundberg K, Ozenci V, Banham AH, Hellström M, Egevad L, et al. CD4+CD25high T cells are enriched in the tumor and peripheral blood of prostate cancer patients. Journal of immunology (Baltimore, Md : 1950). 2006;177:7398‐405. 31. Sfanos KS, Bruno TC, Maris CH, Xu L, Thoburn CJ, Demarzo AM, et al. Phenotypic analysis of prostate‐infiltrating lymphocytes reveals T H17 and Treg skewing. Clin Cancer Res. 2008;14:3254‐61. 32. Reis STD, Pontes‐Júnior J, Antunes AA, Sousa‐Canavez JMd, Abe DK, Cruz JASd, et al. Tgf‐β1 expression as a biomarker of poor prognosis in prostate cancer. Clinics (São Paulo, Brazil). 2011;66:1143‐7. 33. Drake CG. Prostate cancer as a model for tumour immunotherapy. Nat Rev Immunol. 2010;10:580‐93. 34. Ardiani A, Gameiro SR, Kwilas AR, Donahue RN, Hodge JW. Androgen deprivation therapy sensitizes prostate cancer cells to T‐cell killing through androgen receptor dependent modulation of the apoptotic pathway. Oncotarget. 2014;5:9335‐48. 35. Thoma C. Prostate cancer: Towards effective combination of ADT and immunotherapy. Nature Reviews Urology. 2016:2016‐. 36. Gannon PO, Poisson AO, Delvoye N, Lapointe Rj, 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. 37. Bishop JL, Sio A, Angeles A, Roberts ME, Azad AA, Chi KN, et al. PD‐L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget. 2015;6:234‐42. 38. Noguchi M, Koga N, Moriya F, Itoh K. Immunotherapy in prostate cancer: challenges and opportunities. Immunotherapy. 2016;8:69‐77. 39. Wang X, Yu J, Sreekumar A, Varambally S, Shen R, Giacherio D, et al. Autoantibody signatures in prostate cancer. N Engl J Med. 2005;353:1224‐35. 40. Ren R, Koti M, Hamilton T, Graham CH, Nayak JG, Singh J, et al. A primer on tumour immunology and prostate cancer immunotherapy. Journal of the Canadian Urological Association. 2016;10:60‐5. 41. Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, Valone FH, et al. Placebo‐controlled phase III trial of immunologic therapy with sipuleucel‐T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24:3089‐94. 42. Higano CS, Schellhammer PF, Small EJ, Burch PA, Nemunaitis J, Yuh L, et al. Integrated data from 2 randomized, double‐blind, placebo‐controlled, phase 3 trials of active cellular immunotherapy with sipuleucel‐T in advanced prostate cancer. Cancer. 2009;115:3670‐9. 43. Hall SJ, Klotz L, Pantuck AJ, George DJ, Whitmore JB, Frohlich MW, et al. Integrated safety data from 4 randomized, double‐blind, controlled trials of autologous cellular immunotherapy with sipuleucel‐T in patients with prostate cancer. J Urol. 2011;186:877‐81. 44. Huber ML, Haynes L, Parker C, Iversen P. Interdisciplinary critique of sipuleucel‐T as immunotherapy in castration‐resistant prostate cancer. J Natl Cancer Inst. 2012;104:273‐9.
45. GuhaThakurta D, Sheikh NA, Fan LQ, Kandadi H, Meagher TC, Hall SJ, 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. 46. Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst. 2012;104:599‐613. 47. Gulley JL, Mulders P, Albers P, Banchereau J, Bolla M, Pantel K, et al. Perspectives on sipuleucel‐T: Its role in the prostate cancer treatment paradigm. OncoImmunology. 2016;5:e1107698‐e. 48. Longo DL. New therapies for castration‐resistant prostate cancer. N Engl J Med. 2010;363:479‐81. 49. Holko P, Kawalec P. Economic evaluation of sipuleucel‐T immunotherapy in castration‐resistant prostate cancer. Expert Rev Anticancer Ther. 2014;14:63‐73. 50. Simons JW, Sacks N. Granulocyte‐macrophage colony‐stimulating factor‐ transduced allogeneic cancer cellular immunotherapy: the GVAX vaccine for prostate cancer. Urol Oncol. 2006;24:419‐24. 51. Small EJ, Sacks N, Nemunaitis J, Urba WJ, Dula E, Centeno AS, et al. Granulocyte macrophage colony‐stimulating factor‐‐secreting allogeneic cellular immunotherapy for hormone‐refractory prostate cancer. Clin Cancer Res. 2007;13:3883‐91. 52. Higano CS, Corman JM, Smith DC, Centeno AS, Steidle CP, Gittleman M, et al. Phase 1/2 dose‐escalation study of a GM‐CSF‐secreting, allogeneic, cellular immunotherapy for metastatic hormone‐refractory prostate cancer. Cancer. 2008;113:975‐84. 53. Higano C SF, Somer B, Curti B, Petrylak DP, Drake CG, et al. . A phase III trial of GVAX immunotherapy for prostate cancer vs. docetaxel plus prednisone in asymptomatic castration‐resistant prostate cancer (CRPC). Genitourinary Cancer Symposium2009. 54. Small E DT, Gerritsen W et al. A Phase III trial of GVAX immunotherapy for prostate cancer in combination with docetaxel vs docetaxel plus prednisone in symptomatic, castration resistant prostate cancer (CRPC). Genitourinary Cancer Symposium2009. 55. Fernandez‐Garcia EM, Vera‐Badillo FE, Perez‐Valderrama B, Matos‐Pita AS, Duran I. Immunotherapy in prostate cancer: review of the current evidence. Clin Transl Oncol. 2015;17:339‐57. 56. Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer. 2008;8:108‐20. 57. Becker JT, Olson BM, Johnson LE, Davies JG, Dunphy EJ, McNeel DG. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long‐term T‐cell responses in patients with recurrent prostate cancer. J Immunother. 2010;33:639‐47. 58. McNeel DG, Dunphy EJ, Davies JG, Frye TP, Johnson LE, Staab MJ, et al. Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol. 2009;27:4047‐54. 59. 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.
60. Harrington LE, Most Rv R, Whitton JL, Ahmed R. Recombinant vaccinia virus‐ induced T‐cell immunity: quantitation of the response to the virus vector and the foreign epitope. J Virol. 2002;76:3329‐37. 61. Hodge JW, McLaughlin JP, Kantor JA, Schlom J. Diversified prime and boost protocols using recombinant vaccinia virus and recombinant non‐replicating avian pox virus to enhance T‐cell immunity and antitumor responses. Vaccine. 1997;15:759‐68. 62. DiPaola RS, Chen YH, Bubley GJ, Stein MN, Hahn NM, Carducci MA, et al. A national multicenter phase 2 study of prostate‐specific antigen (PSA) pox virus vaccine with sequential androgen ablation therapy in patients with PSA progression: ECOG 9802. Eur Urol. 2015;68:365‐71. 63. Kantoff PW, Schuetz TJ, Blumenstein BA, Michael Glode L, Bilhartz DL, Wyand M, et al. Overall survival analysis of a phase II randomized controlled trial of a poxviral‐based PSA‐targeted immunotherapy in metastatic castration‐resistant prostate cancer. J Clin Oncol. 2010;28:1099‐105. 64. James L. Gulley JLG, James Bradley Breitmeyer, Alexis J. Franzusoff, Dennis Panicali, Jeffrey Schlom, Philip W. Kantoff. A randomized double‐blind phase 3 efficacy study of PROSTVAC‐VF immunotherapy in men with asymptomatic/minimally symptomatic metastatic castration‐resistant prostate cancer. J Clin Oncol. 2015;33. 65. Lubaroff DM, Konety BR, Link B, Gerstbrein J, Madsen T, Shannon M, et al. Phase I clinical trial of an adenovirus/prostate‐specific antigen vaccine for prostate cancer: safety and immunologic results. Clin Cancer Res. 2009;15:7375‐ 80. 66. Lubaroff DM. Prostate cancer vaccines in clinical trials. Expert Rev Vaccines. 2012;11:857‐68. 67. Perez SA, Kallinteris NL, Bisias S, Tzonis PK, Georgakopoulou K, Varla‐ Leftherioti M, et al. Results from a phase I clinical study of the novel Ii‐Key/HER‐ 2/neu(776‐790) hybrid peptide vaccine in patients with prostate cancer. Clin Cancer Res. 2010;16:3495‐506. 68. Noguchi M, Kakuma T, Uemura H, Nasu Y, Kumon H, Hirao Y, et al. A randomized phase II trial of personalized peptide vaccine plus low dose estramustine phosphate (EMP) versus standard dose EMP in patients with castration resistant prostate cancer. Cancer Immunol Immunother. 2010;59:1001‐9. 69. Noguchi M, Moriya F, Koga N, Matsueda S, Sasada T, Yamada A, et al. A randomized phase II clinical trial of personalized peptide vaccination with metronomic low‐dose cyclophosphamide in patients with metastatic castration‐ resistant prostate cancer. Cancer Immunol Immunother. 2016;65:151‐60. 70. Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, 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. 71. Gulley JL, Madan RA, Schlom J. Impact of tumour volume on the potential efficacy of therapeutic vaccines. Current Oncology. 2011;18:150‐7. 72. Halabi S, Small EJ, Kantoff PW, Kattan MW, Kaplan EB, Dawson NA, et al. Prognostic model for predicting survival in men with hormone‐refractory metastatic prostate cancer. J Clin Oncol. 2003;21:1232‐7.
73. Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single‐dose radiotherapy induces an immune‐ mediated abscopal effect when combined with anti‐CTLA‐4 antibody. Clin Cancer Res. 2009;15:5379‐88. 74. Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. 2012;83:1306‐10. 75. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen‐specific effector cells that traffic to the tumor. J Immunol. 2005;174:7516‐23. 76. Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114:589‐95. 77. Tomasz M. Beer EDK, Charles G. Drake, Karim Fizazi, Christopher Logothetis, Gwenaelle Gravis, Vinod Ganju, Jonathan Polikoff, Fred Saad, Piotr Humanski, Josep M. Piulats, Pablo Gonzalez Mella, Siobhan S. Ng, Dirk Jaeger, Francis X. Parnis, Fabio A. Franke, Javier Puente, Roman Carvajal, Lisa Sengeløv, M. Brent McHenry, Arvind Varma, Alfonsus J. van den Eertwegh, and Winald Gerritsen. 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. 2016. 78. Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, et al. Costimulatory B7‐H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A. 2004;101:17174‐9. 79. Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti‐CTLA‐4 treatment (CheckMate 037): a randomised, controlled, open‐label, phase 3 trial. The Lancet Oncology. 2015:375‐84. 80. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015:150419053123009‐. 81. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus Everolimus in Advanced Renal‐Cell Carcinoma. The New England journal of medicine. 2015:1803‐13. 82. Rosenberg JE, Hoffman‐Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum‐ based chemotherapy: a single‐arm, multicentre, phase 2 trial. Lancet. 2016;387:1909‐20. 83. Gupta N, Al Ustwani O, Shen L, Pili R. Mechanism of action and clinical activity of tasquinimod in castrate‐resistant prostate cancer. Onco Targets Ther. 2014;7:223‐34. 84. Shen L, Sundstedt A, Ciesielski M, Miles KM, Celander M, Adelaiye R, et al. Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models. Cancer Immunol Res. 2015;3:136‐48. 85. Pili R, Haggman M, Stadler WM, Gingrich JR, Assikis VJ, Bjork A, et al. Phase II randomized, double‐blind, placebo‐controlled study of tasquinimod in men with
minimally symptomatic metastatic castrate‐resistant prostate cancer. J Clin Oncol. 2011;29:4022‐8. 86. Armstrong AJ, Haggman M, Stadler WM, Gingrich JR, Assikis V, Polikoff J, et al. Long‐term survival and biomarker correlates of tasquinimod efficacy in a multicenter randomized study of men with minimally symptomatic metastatic castration‐resistant prostate cancer. Clin Cancer Res. 2013;19:6891‐901. 87. Sternberg C, Armstrong A, Pili R, Ng S, Huddart R, Agarwal N, et al. Randomized, Double‐Blind, Placebo‐Controlled Phase III Study of Tasquinimod in Men With Metastatic Castration‐Resistant Prostate Cancer. J Clin Oncol. 2016. 88. Heng TS, Goldberg GL, Gray DH, Sutherland JS, Chidgey AP, Boyd RL. Effects of castration on thymocyte development in two different models of thymic involution. J Immunol. 2005;175:2982‐93. 89. Drake CG, Doody AD, Mihalyo MA, Huang CT, Kelleher E, Ravi S, et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer‐restricted antigen. Cancer Cell. 2005;7:239‐49. 90. Ardiani A, Farsaci B, Rogers CJ, Protter A, Guo Z, King TH, et al. Combination Therapy with a Second‐Generation Androgen Receptor Antagonist and a Metastasis Vaccine Survival in a Spontaneous Prostate Cancer Model. Clin Cancer Res. 2013;19. 91. Koh YT, Gray A, Higgins SA, Hubby B, Kast WM. Androgen ablation augments prostate cancer vaccine immunogenicity only when applied after immunization. Prostate. 2009;69:571‐84. 92. Small EJ, Lance RS, Gardner TA, Karsh LI, Fong L, McCoy C, et al. A Randomized Phase II Trial of Sipuleucel‐T with Concurrent versus Sequential Abiraterone Acetate plus Prednisone in Metastatic Castration‐Resistant Prostate Cancer. Clin Cancer Res. 2015;21:3862‐9. 93. 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. 94. Motoyoshi Y, Kaminoda K, Saitoh O, Hamasaki K, Nakao K, Ishii N, et al. Different mechanisms for anti‐tumor effects of low‐ and high‐dose cyclophosphamide. Oncol Rep. 2006;16:141‐6. 95. Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105:256‐65. 96. van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM, et al. Combined immunotherapy with granulocyte‐macrophage colony‐stimulating factor‐transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration‐ resistant prostate cancer: a phase 1 dose‐escalation trial. Lancet Oncol. 2012;13:509‐17. 97. Madan RA, Mohebtash M, Arlen PM, Vergati M, Rauckhorst M, Steinberg SM, 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. 98. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez‐Lopez R, et al. DNA‐Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med. 2015;373:1697‐708.
99. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA‐4 blockade in melanoma. N Engl J Med. 2014;371:2189‐99. 100. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD‐1 Blockade in Tumors with Mismatch‐Repair Deficiency. N Engl J Med. 2015:150530061707006‐. 101. Kubecek O, Trojanova P, Molnarova V, Kopecky J. Microsatellite instability as a predictive factor for immunotherapy in malignant melanoma. Med Hypotheses. 2016;93:74‐6. 102. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD‐1 blockade in non‐small cell lung cancer. Science. 2015;348:124‐8. 103. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti‐PD‐1 antibody in cancer. N Engl J Med. 2012;366:2443‐54. 104. Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Calio A, et al. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death‐Ligand‐1 (PD‐L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS ONE. 2015;10:e0130142. 105. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, 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. 106. Callea M, Albiges L, Gupta M, Cheng SC, Genega EM, Fay AP, et al. Differential Expression of PD‐L1 between Primary and Metastatic Sites in Clear‐Cell Renal Cell Carcinoma. Cancer Immunol Res. 2015;3:1158‐64. 107. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883‐92. 108. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al. Colocalization of inflammatory response with B7‐h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4:127ra37. 109. Choueiri TK, Fishman MN, Escudier BJ, Kim JJ, Kluger HM, Stadler WM, et al. Immunomodulatory activity of nivolumab in previously treated and untreated metastatic renal cell carcinoma (mRCC): biomarker‐based results from a randomized clinical trial. J Clin Oncol. 2014; 32(suppl):5s. abstr 5012. 110. Anastasopoulou EA, Voutsas IF, Keramitsoglou T, Gouttefangeas C, Kalbacher H, Thanos A, et al. A pilot study in prostate cancer patients treated with the AE37 Ii‐key‐HER‐2/neu polypeptide vaccine suggests that HLA‐A*24 and HLA‐DRB1*11 alleles may be prognostic and predictive biomarkers for clinical benefit. Cancer Immunol Immunother. 2015;64:1123‐36. 111. Hoos A, Eggermont AM, Janetzki S, Hodi FS, Ibrahim R, Anderson A, et al. Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst. 2010;102:1388‐97. 112. Hoos A, Britten CM, Huber C, O'Donnell‐Tormey J. A methodological framework to enhance the clinical success of cancer immunotherapy. Nat Biotechnol. 2011;29:867‐70.
113. Bilusic M, Gulley JL. Endpoints, patient selection, and biomarkers in the design of clinical trials for cancer vaccines. Cancer Immunol Immunother. 2012;61:109‐17. 114. Group ICW, Sweeney C, Nakabayashi M, Regan M, Xie W, Hayes J, et al. The Development of Intermediate Clinical Endpoints in Cancer of the Prostate (ICECaP). J Natl Cancer Inst. 2015;107:djv261. 115. Hoos A. Evolution of end points for cancer immunotherapy trials. Ann Oncol. 2012;23 Suppl 8:viii47‐52. 116. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune‐related response criteria. Clin Cancer Res. 2009;15:7412‐20. 117. George S, Motzer RJ, Hammers HJ, Redman BG, Kuzel TM, Tykodi SS, et al. Safety and Efficacy of Nivolumab in Patients With Metastatic Renal Cell Carcinoma Treated Beyond Progression: A Subgroup Analysis of a Randomized Clinical Trial. JAMA Oncol. 2016. 118. Hanna MG, Jr., Hoover HC, Jr., Pinedo HM, Finer M. Active specific immunotherapy with autologous tumor cell vaccines for stage II colon cancer: logistics, efficacy, safety and immunological Pharmacodynamics. Hum Vaccin. 2006;2:185‐91. 119. Butts C, Maksymiuk A, Goss G, Soulieres D, Marshall E, Cormier Y, et al. Updated survival analysis in patients with stage IIIB or IV non‐small‐cell lung cancer receiving BLP25 liposome vaccine (L‐BLP25): phase IIB randomized, multicenter, open‐label trial. J Cancer Res Clin Oncol. 2011;137:1337‐42. 120. Amato RJ, Hawkins RE, Kaufman HL, Thompson JA, Tomczak P, Szczylik C, et al. Vaccination of metastatic renal cancer patients with MVA‐5T4: a randomized, double‐blind, placebo‐controlled phase III study. Clin Cancer Res. 2010;16:5539‐ 47. 121. Sheikh NA SE, Quinn DI, Higano CS, Lin DW, Wesley JD, Hay‐ nes H, Stewart FP, Poehlein CH, Trager JB. Sipuleucel‐T product characterization across different disease states of prostate cancer. J Clin Oncol. 2012;30. 122. Schellhammer PF, Chodak G, Whitmore JB, Sims R, Frohlich MW, Kantoff PW. Lower baseline prostate‐specific antigen is associated with a greater overall survival benefit from sipuleucel‐T in the Immunotherapy for Prostate Adenocarcinoma Treatment (IMPACT) trial. Urology. 2013;81:1297‐302. 123. Muller AJ, Prendergast GC. Indoleamine 2,3‐dioxygenase in immune suppression and cancer. Curr Cancer Drug Targets. 2007;7:31‐40.
Figure 1 – The immunoediting process and immune targets of agents developed for prostate cancer treatment.
Escape
Cell Lysis
Legend Tregs
Tumor Cell
Normal Cell
An1gen‐presen1ng Cell/ Dendri1c Cell
T Regulatory Cell
MSDC
Myeloid‐derived Suppressor Cell
NK Cell
Natural Killer Cell
MP Macrophage
Table‐1 – Current FDA approved therapies for mCRPC.
Setting Comparator Arm (all mCRPC)
Agent Docetaxel6
Median OS
HR (p value)
-
Mitoxantrone
18.9m vs 16.5m
HR 0.76 (p=0.009)
Post-docetaxel
Mitoxantrone
15.1m vs 12.7m
HR 0.70 (p<0001)
Pre-docetaxel
Placebo + prednisone
34.7m vs 30.3m
HR 0.81 (p=0.0033)
Post-docetaxel
Placebo + prednisone
14.8m vs 10.9m
HR 0.65 (p<0.001)
Pre-docetaxel
Placebo + prednisone
32.4m vs 30.2m
HR 0.71 (p<0.001)
Post-docetaxel
Placebo + prednisone
18.4m vs 13.6m
HR 0.63 (p=0.001)
Sipuleucel-T5
Pre or postdocetaxel; asymptomatic or minimally symptomatic
Placebo
25.8m vs 21.7m
HR 0.78 (p=0.03)
Radium-2238
Symptomatic bone metastasis only
Placebo
14.0m vs 11.2m
HR 0.70 (P=0.002)
Cabazitaxel7
10
Abiraterone
9
Enzalutamide
Table 2: Types of Immune System
Innate System Cell type Natural Killer Cells (NK)
Function
Dendritic Cells (DC)
Antigen presenting cells (APC)
Roles
Activate T cells and stimulate NK cells
Macrophages
Adaptive System
Description Immediate response to tissue damage. Crucial role on immune activation and regulation. Linked to adaptive system by antigen presenting cells.
B lymphocytes
Production of antigenspecific antibodies CD4+ (helper)
T lymphocytes
CD8+ (effector)
Tregs
Participate in the humoral response to cancer. T Helper cells recruit other imune cells, activate effector cells. Cell lysis generates more Participate in cell TAAs, relysis initiating the cancer-immunity cycle Secretion of TGFbeta and IL-10 and expression. Supress effector T Of inhibitory cells molecules (CTLA-4 and PD1). Activate B lymphocytes and macrophages
Abbreviations: TAA = tumor associated antigen; Tregs = regulatory T cell; TGF = transforming growth factor; IL = interleukin; CTLA‐4 = cytotoxic ; PD‐1 = programmed cell death.
Table 3: Phases of Immune‐editing process. Phases Elimination
Immune Infiltrate Cytokines Important features Macrophages, NK, IFN-gama, Innate and adaptive Dendritic Cells MICA/B, IL-12, systems play together TNF
State of dormancy. Editing in progress. Development of immunoevasive mutations. Mainly Tregs and BCL2, VEGF, Immunossupressive. Evasion MDSC, but also TGF-beta, Expression of impaired CD8+ T galectin-1, IDO, inhibitory molecules cells IL-10. (CTAL4 and PD-1). Abbreviations: NK = natural killer; IFN = interferon; IL = interleukin; TNF = tumor necrosis factor; Treg = T regulatory cell; MDSC = myeloid derived suppressive cell; BCL2 = B‐cell lymphoma 2; VEGF = vascular endothelial growth factor; TGF = transforming growth factor; IDO = indoleamine 2,3‐dioxygenase; CTLA4 = cytotoxic T-lymphocyte associated protein 4; PD‐1 = programmed cell death 1. Equilibrium
CD8+ and CD4+ T IL-2, IFN-gama. cells
Table 4: Types of cancer vaccines. Description
Advantages
Allogenic Cell Vaccine
Derived from inactivated tumor cell lines
Easy preparation, targets various antigens, favorable safety profile
Autologous cell vaccine
Derived from host tumor cells
Target patients` own TAAs
Cost, time and laborconsuming, complexity, large amount of material required, must be cultured with cytokines and stimulants
DNA-based
Nucleic acids encoding genes for specific TAAs
Simplicity, stability, cost
pTVG‐HP Low immunogenicity; (PAP) need combination with proinflammatory molecules (IL-2, GM-CSF)
Viral-vector
Incorporation of genes within virus genome followed by Infection of epithelial cells that when lysed will release TAAs, which will be presented by APCs to naïve T cells.
Easy to manufacture high amount of genetic material, large experience, cheap.
Development of antibodies against viral vector coat proteins, leading to neutralization after 1st injection; low immunogenicity
Inflammatory responses caused by vectors recruit immune infiltrates to
Disadvantages
Example
Vaccine Type
GVAX Onyvax
Sipuleucel‐T
PROSTVAC /PSA‐TRICOM (pox + vaccinia viruses) Ad5‐PSA (adenovirus)
site. Broad propagation among APCs Personalized Peptide Vaccination
Utilizes HLAmatched CD8+ T cells Peptides based on host’s preexisting immunity
Harnesses the pre-existing immunity Direct towards specific tumor antigens
Cost, time and laborconsuming, Expensive, low immunological response
‐
Abbreviations: IL = interleukin; GM‐CSF = granulocyte macrophage colony stimulating fator.
Table 5. Ongoing Clinical Trals involving Immunotherapies in Prostate Cancer Localized Prostate Cancer Clinical Trial Pha se
Population
NCT0187525 0
II
Localized castratesensitive
NCT0141391
II
Nonmetastatic
NCT0058302 4
II
NCT0085978 1
II
Experimental arm
Control Endpoints Accru al Goal
Enzalutamide Enzaluta Primary = 160mg PO daily mide decrease in + PROSTVAC 160mg tumor reSC q2w for up PO daily growth to 7 doses rate; Secondary = immune response/ impacto in PSA. Sipuleucel-T followed by ADT (leuprolide acetate 45mg SC) q6mo
ADT (leuprolid e acetate 45mg SC) q6mo followed after 12 weeks by Sipuleuce l-T
58
Primary = imune parameters ; Secondary = safety; PK; PSA response
68
Hormonerefractory localized or metastatic
Adenoviurs/PS Not Primary = A vaccine for 3 Controlle PSA DT doses qmonthly d response; Secondary = PSA response and immue response
66
Nonmetastatic castrateresistante
Ketoconazol Ketocona Primary = 400mg PO q8h zol Metastasis for 4 weeks 400mg free followed by PO q8h survival; 177Lu-J591 for 4 Secondary Infusion weeks = PSA followed response. by Placebo
140
NCT0169687 7
I/II
NCT0141391
II
Localized Cyclophospham Degarelix Prostate ide 200mg/m2 240mg 14 Adenocarcin IV followed by days prior oma GVAX ID x 5 + to surgery ADT (degarelix 240mg SC) 14 days later on the day of surgery
Nonmetastatic
Sipuleucel-T followed by ADT (leuprolide acetate 45mg SC) q6mo
ADT (leuprolid e acetate 45mg SC) q6mo followed after 12 weeks by Sipuleuce l-T
Primary = intraprosta tic CD8+ T cell infiltration ; safety Secondary = PSA response and timeto-PSA recurrence; pCR; immune responses; intraprosta ttic CD4+ and tregs infiltration ;
29
Primary = imune parameters ; Secondary = safety; PK; PSA response
68
Metastatic Prostate Cancer NCT0260101 4
II
mCRPC ARV7 positive
Nivolumab + Not Primary= Ipilimumab Controlled PSA q3w for 12 response; weeks Secondary followed by = OS, nivolumab PFS, ORR; q2w for 36 safety; rate weeks or until of AR-V7 disease conversion progression
15
NCT0249983 5
I/II
mCRPC
pTVG-HP Not Primary = vaccine ID Controlled safety; q2w x 6 and PSA pembrolizuma response; b IV q3w x 4 6-mo PFS from D1 rate;
32
or the same schedule + pembrolizuma b from day 85.
median rPFS; ORR; Secondary = immune response and parameters
NCT0248935 7
II
HormoneADT Not Primary = sensitive (degarelix)+ Controlled proportion oligometastat Pembrolizuma of PSA < ic b q3w up to 6 0.6ng/ml doses + at 12mo; prostate gland Secondary cryoablation = PD-1 and PD-L1 expression
12
NCT0278700 5
II
mCRPC Pembrolizuma Not Primary = postb 200mg IV Controlled ORR; chemotherap q3w upto Secondary y 24mo = DCR; PSA response; safety;
250
NCT0186733 II 3
NCT0132249 0
III
mCRPC
mCRPC asymptomati c or minimally symptomatic
Enzalutamide Enzalutami Primary = 160mg PO de 160mg TTP; daily + PO daily Secondary PROSTVAC = OS; PSA SC q2w then PFS; qmonthly x imune 6mo then response q3months Prostvac-VF +/- GM-CSF
Placebo
Primary = OS; Secondary = eventfree survival
76
1298
NCT0114550 II 8
mCRPC
Prostvac/TRIC Docetaxel OM Vaccine 75mg/m2 SC q15d x 5 IV q3w followed by Docetacel 75mg/m2 IV q3w
Primary = 10 OS
NCT0148786 II 3
mCRPC
Concurrent Sipuleucel- Primary = 69 Sipuleucetel-T T + cumulative
+ abiraterone 1000 mg PO daily
abiraterona 100mg PO daily (after 6 weeks of last infusion)
sipuleucelT CD54 upregulati on; Secondary = PK; imune response; safety.
UMIN000011 III 308
mCRPC postdocetaxel (HLA-A24+)
ITK-1 (peptide Placebo vaccine) q1w x 6 followed by q2w for up to 30 times;
Primary = 333 OS; Secondary = PSA response; imune correlates; safety
NCT0231255 II 7
mCRPC upon progression on Enzalutamide
Pembrolizuma Not b q3w for 4 Controlled doses + Enzalutamide 160mg PO daily continously
Primary = 28 PSA response; Secondary = OR; imune parameters ; OS;
NCT0286157 Ib/II mCRPC 3
Multi-arm with Not backbone of Controlled Pembrolizuma be 200mg IV q3w; Arm 1: With Olaparib 400mg PO BID; Arm 2: With Docetaxel 75mg IV q3w. Arm 3: With Enzalutamide 160mg PO daily;
Primary = 210 PSA response (≥50%); safety; Secondary = ORR; OS; DCR;
NCT0091612 I 3
Docetaxel Not 75mg/m2 q3w Controlled + 177LuDOTA-J591 2 infusions (2040 mCi/dose) prior to cycle
Primary = 30 MTD; Secondary = toxicity profile
mCRPC
3. Abbreviations: NCT = national clinical trial; ADT = androgen deprivatin therapy; mCRPC = metastatic castration‐resistant prostate cancer; MTD = maximum tolerated dose; PK = pharmacokinectics; PSA = prostate especific antigen; PFS = progression‐free survival; OS = overall survival; pCR = pathologic complete response; DCR = disease control rate; ORR = overall response rate; TTP = time‐ to‐progression;
Table 6: Biomarkers Treatment AE37 polypeptide vaccination (PPV)
Biomarker
Description
HLA-DRB1*11 Better and/or HLA-A*24 immunological and alleles (positively); clinical responses (OS) Pre-existing IFN- Predicted gama (positively) immunological and TGF-beta responses (negatively) levels
Sipuleucel-T
Comment May be surrogates for increased benefits from this therapy. Needs validation.
Needs prospective Higher Increased validation eosinophils at week immunological activation, 6 post-treatment increased PCSS and trend for higher OS of Humoral (IgG) Higher OS and Importance CD4+ T cells on response to PSA durable responses activation of and LGALS3. humoral activity
Pox-viral Vaccine
PSA-specific T cell Higher OS; response; Favorable prognostic features
Anti-PD1 and anti-CTLA-4 agents
Indolent disease and less advanced states derive greater benefit from immunotherapies in general
Mutational Load
Mutational burden Higher ORR, PFS correlates with neo- and OS epitopes formation
PD-L1 expression
Level of expression in TIL or tumor cells may be quantified and cutoff level arbitrary defined
Issues with assay reproductibility; does not accurately predicts response or lack of response. Other still unknown coinhibitory molecules probably important.
Abbreviations: IFN = interferon; TGF = transforming growth fator; PCSS = prostate cancer especific survival; PSA = prostate especific antigen; PD-L1 = programmed cell death ligand 1; CTLA-4 = cytotoxic T-lymphocyte associated protein 4; TIL = tumor infiltrate lymphocyte; ORR = overall response rate; PFS = progression-free survival; OS = overall survival.