Purpose: Therapies targeting the programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway promote anti-tumor immunity and have shown promising results in various tumors. Preliminary data further indicate that immunohistochemically detected PD-L1 may be predictive for anti-PD-1 therapy. So far, no data are available on PD-L1 expression in primary prostate cancer.

Experimental Design: Following validation of a monoclonal antibody, immunohistochemical analysis of PD-L1 expression was performed in two independent, well-characterized cohorts of primary prostate cancer patients following radical prostatectomy (RP), and resulting data were correlated to clinicopathological parameters and outcome.

Results: In the training cohort (n = 209), 52.2% of cases expressed moderate to high PD-L1 levels, which positively correlated with proliferation (Ki-67, P < 0.001), Gleason score (P = 0.004), and androgen receptor (AR) expression (P < 0.001). Furthermore, PD-L1 positivity was prognostic for biochemical recurrence [BCR; P = 0.004; HR, 2.37; 95% confidence interval (CI), 1.32–4.25]. In the test cohort (n = 611), moderate to high PD-L1 expression was detected in 61.7% and remained prognostic for BCR in univariate Cox analysis (P = 0.011; HR, 1.49; 95% CI, 1.10–2.02). The correlation of Ki-67 and AR with PD-L1 expression was confirmed in the test cohort (P < 0.001). In multivariate Cox analysis of all patients, PD-L1 was corroborated as independently prognostic for BCR (P = 0.007; HR, 1.46; 95% CI, 1.11–1.92).

Conclusions: We provide first evidence that expression of the therapy target PD-L1 is not only highly prevalent in primary prostate cancer cells but is also an independent indicator of BCR, suggesting a biologic relevance in primary tumors. Further studies need to ascertain if PD-1/PD-L1–targeted therapy might be a treatment option for hormone-naïve prostate cancers. Clin Cancer Res; 22(8); 1969–77. ©2015 AACR.

Translational Relevance

Cancer immunotherapy represents a breakthrough in oncology. Among immunotherapeutical approaches, blocking the PD-1/PD-L1 pathway, and thereby promoting activity of tumor-specific effector T cells, has shown outstanding results. However, high response rates are only seen in a few cancer entities (e.g., melanomas). In patients with hormone-refractory metastatic prostate cancer, previous studies have not been able to provide evidence of response to anti-PD-L1 therapy. Our study revealed that PD-L1 is differentially expressed among primary prostate cancer patients. Furthermore, high PD-L1 expression was associated with a poor prognosis. Accordingly, PD-1/PD-L1–targeted therapy might be a promising novel treatment option for hormone-naïve prostate cancers.

Prostate cancer is the second most frequently diagnosed cancer in men, accounting for approximately 15% of all newly diagnosed male cancers worldwide (1). Overall, it is the fifth most common cause of death from cancer in men, with 307.000 estimated deaths (6.6% of all estimated deaths) in 2012 (1). First-line therapies for early-stage localized prostate cancer include surgery and radiotherapy, and the 5-year relative survival rate approaches 100% (2). For patients affected by metastatic prostate cancer, androgen deprivation therapy (ADT) still is the mainstay of treatment (3). Although surgical or chemical castration can initially be effective delaying disease progression, the majority of patients eventually develops castration-resistant prostate cancer (CRPC) and has an adverse prognosis (4, 5). Recent advances and the arrival of several new agents in the field of CRPC, however, have improved overall survival in this patient population. Enzalutamid, a novel androgen receptor (AR) inhibitor that reduces nuclear translocation of the AR complex and subsequent DNA binding, was shown to significantly prolong overall survival in two phase III randomized, placebo-controlled trials (6, 7). Furthermore, abiraterone, an inhibitor of cytochrome P450, family 17, subfamily A, polypeptide 1 (CYP17A1), suppresses androgen biosynthesis in combination with prednisone, increasing survival time and time to prostate-specific antigen (PSA) progression (8, 9). Above all, the early addition of docetaxel to ADT has recently been shown to extend survival for men with newly diagnosed hormone-sensitive prostate cancer by more than 13 months (10).

Among many concepts under investigation, augmenting immune responses to cancer has been proposed as a valid therapeutic option, providing an alternative approach to improve survival. In particular, checkpoint inhibitors have emerged as a complementary avenue of clinical research in prostate cancer (11, 12). These treatments target checkpoint molecules in the regulation of the immune system harnessing preexisting anticancer immune responses. The programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway plays a pivotal role in the regulation of T-cell activity at the time of inflammatory response. The PD-1 receptor thereby acts as a negative checkpoint regulator to prevent off-target immune activation of T-lymphocytes (13). Binding of PD-1 to its ligand PD-L1, the predominant mediator of immunosuppression, inhibits proliferation of activated T cells leading to “T-cell exhaustion” (14). PD-L1 itself is an immunomodulatory cell-surface glycoprotein primarily expressed by antigen-presenting cells on myeloid dendritic cells, activated T cells, and some non-hematopoietic tissues (15).

Recent evidence strongly suggests that the activation of the PD-1/PD-L1 pathway represents a mechanism allowing tumors to elude the host's immune system. Therapies targeting this signaling pathway promote marked antitumor immunity and have shown promising results in a subset of solid tumors (16, 17). As reported by Topalian and colleagues, blockade of PD-L1 using an anti-PD-1 antibody induced objective response rates of 6% to 17% and prolonged stabilization of disease in patients with several advanced cancers, that is, malignant melanoma, non–small cell lung cancer, and renal cell cancer (17). Further, immunohistochemical assessment of PD-L1 in pretreatment cancer specimens from 42 patients revealed that response to treatment was seen exclusively in PD-L1–positive tumors (9/25, 36%), indicating that PD-L1 expression might be a predictive biomarker for anti-PD-1 therapy (17). However, several studies have demonstrated response to treatment in patients with low or absent PD-L1 expression, although these patients seem to be in the minority (18–23). So far, no data are available to support that PD-1/PD-L1–targeted therapy may be effective in prostate cancer. This prompted us to analyze PD-L1 expression in primary prostate cancer.

Currently, only few PD-L1 antibodies have been validated for formalin-fixed paraffin embedded (FFPE) material, only one of which being commercially available ((E1L3N) XP; refs. 14, 24–31). The utilization of this antibody for predicting response to anti-PD1 or anti-PD-L1 therapy, however, remains unknown. The majority of clinical trials targeting the PD-1/PD-L1 pathway are using proprietary antibodies, implying that validation data for these particular antibodies are undefined. So far, no immunohistochemical detection method for quantifying PD-L1 expression is uniformly accepted as standard. We here comprehensively validated a novel monoclonal rabbit antibody against PD-L1 (clone EPR1161(2)) amenable to FFPE tissue and analyzed two large, well-characterized cohorts of primary prostate cancer after radical prostatectomy (RP) for PD-L1 expression.

Patients

Two previously described RP cohorts were analyzed in the present study (32–34). Clinicopathologic details, including Gleason grade grouping according to the latest International Society of Urological Pathology (ISUP) consensus (35), are given in Table 1. The training cohort included 262 patients having undergone radical prostatectomy at the University Hospital of Bonn between 1998 and 2008. Immunohistochemical PD-L1 staining was available for 209 cases. The test cohort comprised 640 patients diagnosed at the Charité University Hospital, Berlin between 1999 and 2005, and a total of 611 cases were eligible for PD-L1 immunhistochemistry. All research conducted was approved by the Charité Ethics Committee (EA1/06/2004) and the University Hospital of Bonn, Germany.

Table 1.

Baseline characteristics

Training cohortTest cohort
All patients (%)PD-L1 high (%)PD-L1 low (%)PAll patients (%)PD-L1 high (%)PD-L1 low (%)P
Patient number 209 (100.0) 109 (52.2) 100 (47.8)  611 (100.0) 377 (61.7) 234 (38.3)  
Mean follow-up (mo) 62.8    49.5    
Median follow-up (mo) 61.0    49.6    
Range follow-up (mo) 0–140    0–129    
Age 
 Range 45–83    43–74    
 Mean 64    62    
 Median 65    62    
 ≤Median 108 (51.7) 47 (22.5) 61 (29.2)  307 (50.2) 190 (31.1) 117 (19.1)  
 >Median 100 (47.8) 61 (29.2) 39 (18.7) P = 0.010a 304 (49.8) 187 (30.6) 117 (19.1) P = 0.92a 
 Unknown 1 (0.5)    0 (0.0)    
 <50 5 (2.4) 0 (0.0) 5 (2.4)  19 (3.1) 11 (1.8) 8 (1.3)  
 50–54 5 (2.4) 3 (1.4) 2 (1.0)  55 (9.0) 31 (5.1) 24 (3.9)  
 55–59 28 (13.4) 14 (6.7) 14 (6.7)  105 (17.2) 62 (10.1) 43 (7.0)  
 60–64 58 (27.8) 24 (11.5) 34 (16.3)  222 (36.3) 149 (24.4) 73 (11.9)  
 65–69 74 (35.4) 49 (23.4) 25 (12.0)  175 (28.6) 100 (16.4) 75 (12.3)  
 ≥70 38 (18.2) 18 (8.6) 20 (9.6) P = 0.077b 35 (5.7) 24 (3.9) 11 (1.8) P = 0.58b 
Pathologic stage 
 pT2 124 (59.3) 63 (30.1) 61 (29.2)  418 (68.4) 251 (41.1) 167 (27.3)  
 pT3/pT4 85(40.7) 46 (22.0) 39 (18.7) P = 0.64a 193 (31.6) 126 (20.6) 67 (11.0) P = 0.22a 
Preoperative PSA (ng/mL) 
 Range 0.7–163    0.8–39.0    
 Mean 11.6    8.5    
 Median 7.5    7.1    
 <Median 98 (46.9) 54 (25.8) 44 (21.1)  301 (49.3) 170 (27.8) 131 (21.4)  
 ≥Median 101 (48.3) 51 (24.4) 50 (23.9) P = 0.571a 302 (49.4) 201 (32.9) 101 (16.5) P = 0.011a 
 Unknown 10 (4.8)    8 (1.3)    
 ≤4.0 16 (7.7) 9 (4.3) 7 (3.3)  46 (7.5) 29 (4.7) 17 (2.8)  
 4.1–10 119 (56.9) 66 (31.6) 53 (25.4)  395 (64.6) 234 (38.3) 161 (26.4)  
 10.1–20 43 (20.6) 18 (8.6) 25 (12.0)  142 (23.2) 101 (16.5) 41 (6.7)  
 >20 21 (10.0) 12 (5.7) 9 (4.3) P = 0.53b 20 (3.3) 7 (1.1) 13 (2.1) P = 0.76b 
Surgical margin 
 Unknown 2 (1.0)    3 (0.5)    
 R1 83 (39.7) 50 (23.9) 33 (15.8)  169 (27.7) 107 (17.5) 62 (10.1)  
 R0 124 (59.3) 57 (27.3) 67 (32.1) P = 0.044a 439 (71.8) 268 (43.9) 171 (28.0) P = 0.61a 
Gleason grade 
 Unknown 5 (2.4)        
 G1 (<7) 99 (47.4) 42 (20.1) 57 (27.3)  216 (35.4) 126 (20.6) 90 (14.7)  
 G2 (7) 61 (29.2) 37 (17.7) 24 (11.5)  286 (46.8) 180 (29.5) 106 (17.3)  
 G3 (>7) 44 (21.1) 29 (13.9) 15 (7.2) P = 0.004b 109 (17.8) 71 (11.6) 38 (6.2) P = 0.20b 
Gleason grading group 
 Unknown 5 (2.4)        
 I (<7) 99 (47.4) 42 (20.1) 57 (27.3)  216 (35.4) 126 (20.6) 90 (14.7)  
 2 (3+4) 41 (19.6) 24 (11.5) 17 (8.1)  228 (37.3) 137 (22.4) 91 (14.9)  
 3 (4+3) 20 (9.6) 13 (6.2) 7 (3.3)  58 (9.5) 43 (7.0) 15 (2.5)  
 4 (8) 30 (14.3) 20 (9.6) 10 (4.8)  69 (11.3) 45 (7.4) 24 (3.9)  
 5 (>8) 14 (6.7) 9 (4.3) 5 (2.4) P = 0.007b 40 (6.5) 26 (4.3) 14 (2.3) P = 0.11b 
Nodal status (pN) 
 Unknown 1 (0.5)    303 (49.6)    
 N0 192 (91.9) 102 (48.8) 90 (43.1)  299 (48.9) 201 (32.9) 98 (16.0)  
 N1 16 (7.7) 7 (3.3) 9 (4.3) P = 0.47a 9 (1.5) 5 (0.8) 4 (0.7) P = 0.46a 
Androgen receptor 
 Unknown 5 (2.4)    67 (11.0)    
 None 5 (2.4) 1 (0.5) 4 (1.9)  40 (6.5) 18 (2.9) 22 (3.6)  
 Weak 61 (29.2) 22 (10.5) 39 (18.7)  144 (23.6) 79 (12.9) 65 (10.6)  
 Moderate 80 (38.3) 45 (21.5) 35 (16.7)  266 (43.5) 179 (29.3) 87 (14.2)  
 Strong 58 (27.5) 39 (18.7) 19 (9.1) P < 0.001b 94 (15.4) 69 (11.3) 25 (4.1) P < 0.001b 
Ki-67 
 Unknown 5 (2.4)    17 (2.8)    
 Negative (≤median) 118 (56.5) 49 (23.4) 69 (33.0) P < 0.001a 301 (49.3) 153 (25.0) 148 (24.2) P < 0.001a 
 Positive (>median) 86 (41.1) 58 (27.8) 28 (13.4)  293 (48.0) 214 (35.0) 79 (12.9)  
Training cohortTest cohort
All patients (%)PD-L1 high (%)PD-L1 low (%)PAll patients (%)PD-L1 high (%)PD-L1 low (%)P
Patient number 209 (100.0) 109 (52.2) 100 (47.8)  611 (100.0) 377 (61.7) 234 (38.3)  
Mean follow-up (mo) 62.8    49.5    
Median follow-up (mo) 61.0    49.6    
Range follow-up (mo) 0–140    0–129    
Age 
 Range 45–83    43–74    
 Mean 64    62    
 Median 65    62    
 ≤Median 108 (51.7) 47 (22.5) 61 (29.2)  307 (50.2) 190 (31.1) 117 (19.1)  
 >Median 100 (47.8) 61 (29.2) 39 (18.7) P = 0.010a 304 (49.8) 187 (30.6) 117 (19.1) P = 0.92a 
 Unknown 1 (0.5)    0 (0.0)    
 <50 5 (2.4) 0 (0.0) 5 (2.4)  19 (3.1) 11 (1.8) 8 (1.3)  
 50–54 5 (2.4) 3 (1.4) 2 (1.0)  55 (9.0) 31 (5.1) 24 (3.9)  
 55–59 28 (13.4) 14 (6.7) 14 (6.7)  105 (17.2) 62 (10.1) 43 (7.0)  
 60–64 58 (27.8) 24 (11.5) 34 (16.3)  222 (36.3) 149 (24.4) 73 (11.9)  
 65–69 74 (35.4) 49 (23.4) 25 (12.0)  175 (28.6) 100 (16.4) 75 (12.3)  
 ≥70 38 (18.2) 18 (8.6) 20 (9.6) P = 0.077b 35 (5.7) 24 (3.9) 11 (1.8) P = 0.58b 
Pathologic stage 
 pT2 124 (59.3) 63 (30.1) 61 (29.2)  418 (68.4) 251 (41.1) 167 (27.3)  
 pT3/pT4 85(40.7) 46 (22.0) 39 (18.7) P = 0.64a 193 (31.6) 126 (20.6) 67 (11.0) P = 0.22a 
Preoperative PSA (ng/mL) 
 Range 0.7–163    0.8–39.0    
 Mean 11.6    8.5    
 Median 7.5    7.1    
 <Median 98 (46.9) 54 (25.8) 44 (21.1)  301 (49.3) 170 (27.8) 131 (21.4)  
 ≥Median 101 (48.3) 51 (24.4) 50 (23.9) P = 0.571a 302 (49.4) 201 (32.9) 101 (16.5) P = 0.011a 
 Unknown 10 (4.8)    8 (1.3)    
 ≤4.0 16 (7.7) 9 (4.3) 7 (3.3)  46 (7.5) 29 (4.7) 17 (2.8)  
 4.1–10 119 (56.9) 66 (31.6) 53 (25.4)  395 (64.6) 234 (38.3) 161 (26.4)  
 10.1–20 43 (20.6) 18 (8.6) 25 (12.0)  142 (23.2) 101 (16.5) 41 (6.7)  
 >20 21 (10.0) 12 (5.7) 9 (4.3) P = 0.53b 20 (3.3) 7 (1.1) 13 (2.1) P = 0.76b 
Surgical margin 
 Unknown 2 (1.0)    3 (0.5)    
 R1 83 (39.7) 50 (23.9) 33 (15.8)  169 (27.7) 107 (17.5) 62 (10.1)  
 R0 124 (59.3) 57 (27.3) 67 (32.1) P = 0.044a 439 (71.8) 268 (43.9) 171 (28.0) P = 0.61a 
Gleason grade 
 Unknown 5 (2.4)        
 G1 (<7) 99 (47.4) 42 (20.1) 57 (27.3)  216 (35.4) 126 (20.6) 90 (14.7)  
 G2 (7) 61 (29.2) 37 (17.7) 24 (11.5)  286 (46.8) 180 (29.5) 106 (17.3)  
 G3 (>7) 44 (21.1) 29 (13.9) 15 (7.2) P = 0.004b 109 (17.8) 71 (11.6) 38 (6.2) P = 0.20b 
Gleason grading group 
 Unknown 5 (2.4)        
 I (<7) 99 (47.4) 42 (20.1) 57 (27.3)  216 (35.4) 126 (20.6) 90 (14.7)  
 2 (3+4) 41 (19.6) 24 (11.5) 17 (8.1)  228 (37.3) 137 (22.4) 91 (14.9)  
 3 (4+3) 20 (9.6) 13 (6.2) 7 (3.3)  58 (9.5) 43 (7.0) 15 (2.5)  
 4 (8) 30 (14.3) 20 (9.6) 10 (4.8)  69 (11.3) 45 (7.4) 24 (3.9)  
 5 (>8) 14 (6.7) 9 (4.3) 5 (2.4) P = 0.007b 40 (6.5) 26 (4.3) 14 (2.3) P = 0.11b 
Nodal status (pN) 
 Unknown 1 (0.5)    303 (49.6)    
 N0 192 (91.9) 102 (48.8) 90 (43.1)  299 (48.9) 201 (32.9) 98 (16.0)  
 N1 16 (7.7) 7 (3.3) 9 (4.3) P = 0.47a 9 (1.5) 5 (0.8) 4 (0.7) P = 0.46a 
Androgen receptor 
 Unknown 5 (2.4)    67 (11.0)    
 None 5 (2.4) 1 (0.5) 4 (1.9)  40 (6.5) 18 (2.9) 22 (3.6)  
 Weak 61 (29.2) 22 (10.5) 39 (18.7)  144 (23.6) 79 (12.9) 65 (10.6)  
 Moderate 80 (38.3) 45 (21.5) 35 (16.7)  266 (43.5) 179 (29.3) 87 (14.2)  
 Strong 58 (27.5) 39 (18.7) 19 (9.1) P < 0.001b 94 (15.4) 69 (11.3) 25 (4.1) P < 0.001b 
Ki-67 
 Unknown 5 (2.4)    17 (2.8)    
 Negative (≤median) 118 (56.5) 49 (23.4) 69 (33.0) P < 0.001a 301 (49.3) 153 (25.0) 148 (24.2) P < 0.001a 
 Positive (>median) 86 (41.1) 58 (27.8) 28 (13.4)  293 (48.0) 214 (35.0) 79 (12.9)  

NOTE: Clinicopathologic variables and PD-L1 expression dichotomized by median (high = above median, low = below median) of the training (n = 209) and test cohort (n = 611). All patients had a localized prostate cancer (M0) with a postoperative decrease of PSA serum levels below 0.1 ng/mL.

P values in boldface indicate the level of significance.

aχ2 test (Pearson).

bχ2 test (linear-by-linear).

Tissue microarray construction

Tissue microarrays (TMA) were constructed as previously described (32). Briefly, FFPE tissue specimens were selected according to tissue availability and retrieved from the archive of the Charité University Hospital Berlin and the University Hospital Bonn. Each case was represented by one to three tissue cores with a core diameter of 1.2 mm or 1.8 mm.

Cell line controls and FACS analysis

The prostate cancer cell line DU145 was obtained from the ATCC and maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% FBS, 1% l-glutamine, and 1% antibiotics (Life Technologies). Cells were grown at 37°C in a humidified 5% CO2 atmosphere. PD-L1 expression was analyzed using flow cytometric analysis. Briefly, 5 × 105 cells were washed with 5 mL PBS and resuspended in 300 μL of PBS. FACS analysis was performed on a Navios Flow Cytometer (Beckman Coulter) following incubation with FITC-tagged anti-human PD-L1 or mouse IgG isotype control (BD Pharmingen) for 20 minutes at room temperature, a second wash, and resuspension in PBS.

Western blot analysis

Cells were lysed in RIPA buffer (Sigma-Aldrich) in the presence of protease inhibitors. Cleared lysates were separated on NuPAGE 4% to 12% Bis–Tris gels (1.0 mm, 15 well; Life Technologies) and transferred onto an Invitrolon PVDF membrane (Life Technologies). Membranes were stained using anti-PD-L1 rabbit monoclonal antibody (mAb) clone EPR1161(2) (Abcam; 1:500) in 5% nonfat dry milk, and protein concentrations were determined by a colorimetric assay (Bio-Rad Laboratories Inc.). Blots were stripped and reprobed with anti-β-actin mAb (Sigma-Aldrich; 1:500). Western blot analysis with the validated anti-PD-L1 rabbit mAb (E1L3N) XP (Cell Signalling Technology; 1:500) was performed for comparison.

siRNA transfection for transient knockdown of PD-L1

DU145 cells were seeded with 2 × 105 cells in 6-well plates and incubated for 72 hours in DMEM in the presence of 10 to 20 nmol/L siRNA directed against PD-L1 (FlexiTube Gene Solution GS29126, Qiagen) or non-targeting control siRNA (Qiagen) complexed with HiPerFect Transfection Reagent (Qiagen) according to the manufacturer's instructions. Cells treated with HiPerFect Transfection Reagent only served as controls. siRNA-induced knockdown was evaluated using Western blot analysis as described above.

Peptide neutralization

Prior to standard proceedings, the anti-PD-L1 antibody clone EPR1161(2) was incubated with a 10-fold concentration in excess of blocking peptide (Abcam) that corresponds to the epitope recognized by the primary antibody. Western blot analysis was carried out with the neutralized antibody side-by-side with the antibody alone. In analogy to this experiment, immunohistochemical staining was performed on placental and tonsillar tissue, following primary antibody neutralization with a 10-fold concentration in excess of blocking peptide, and compared with standard staining results.

Immunohistochemistry and evaluation

Tissue sections were freshly cut (3 μm), mounted on Super Frost Plus slides (Thermo Fisher), and rehydrated in descending gradient alcohols. Immunohistochemical staining was carried out on the Ventana BenchMark Ultra automated staining system (Ventana) and visualized with the Ventana amplifier detection kit using the following antibodies: PD-L1, clone EPR1161(2) (Abcam; 1:75), Ki-67, clone MIB-1 (Dako; 1:500), and AR, clone AR441 (Dako; 1:400). Omission of the primary antibody was used as a negative control. Immunohistochemical stainings were evaluated independently by two pathologists (G. Kristiansen and H. Gevensleben) who were blinded to patients' clinical outcome. Specific membranous and cytoplasmic staining of epithelial tumor cells was considered positive. Because the staining was uniformly homogeneous, the intensity of PD-L1–positive cells was scored semi-quantitatively as negative (0), weak (1), moderate (2), or strong (3). Discrepant cases were reviewed at a multi-headed microscope and the consensus was reported.

Statistical analysis

Following dichotomization by median, associations between PD-L1 protein expression and clinicopathologic variables, including AR and Ki-67 expression, were analyzed using the χ2 test. The correlation of Ki-67 and AR with PD-L1 expression as continuous variables was further tested using the Kendall τ rank correlation. Biochemical recurrence (BCR)-free survival was calculated for PD-L1 expression dichotomized by median using the Kaplan–Meier method, and survival time differences were compared using the log-rank test. PD-L1 expression as continuous variable was also examined within univariate and multivariate Cox proportional hazards regression models. P values refer to the Wald test. An interrater reliability analysis using the Kappa (κ) statistic was conducted to determine consistency among raters. All statistical tests were two-sided. P < 0.05 were considered to be statistically significant. All analyses were carried out using the SPSS 21 software package (IBM).

Validation of PD-L1 antibody specificity

For immunohistochemical analysis of PD-L1 expression in FFPE tissue, we validated a novel mAb against PD-L1 (clone EPR1161(2)) and compared its performance with the previously validated PD-L1 mAb (E1L3N) XP (14, 24–31). The prostate cancer cell line DU145 served as a positive control. Flow cytometric analysis (FACS) of untreated DU145 cells compared with concentration matched mouse IgG isotype control demonstrated positive PD-L1 membrane expression in 40% of cells (Fig. 1A). For Western blot analysis, cell lysates from untreated DU145 and MCF7 cells were probed with PD-L1 mAbs. Both antibodies detected a band at the expected size of glycosylated PD-L1 (∼55 kDa) for the FACS-positive cell line DU145 (Fig. 1B). As reported previously (36), PD-L1 was not detected in MCF7 cells. Antibody specificity was confirmed by small interfering RNA (siRNA) knockdown of PD-L1 and Western blot analysis (Fig. 1C). Cross-reactivity was excluded using immunizing peptide blocking experiments. Primary antibody neutralization with a specific blocking peptide prior to Western blot and immunohistochemical proceedings abolished immunoreactivity and thus verified specificity of this PD-L1 mAb clone EPR1161(2) (Fig. 1D).

Figure 1.

Validation of PD-L1 antibody specificity. A, flow cytometric analysis of untreated DU145 cells compared with concentration-matched mouse IgG isotype control showed PD-L1 membrane expression in 40% of cells. B, Western blot analysis of untreated DU145 and MCF7 cells probed with the PD-L1 mAb clone EPR1161(2) (left) and the validated PD-L1 mAb (E1L3N) XP (right). Equal loading was shown by probing for β-actin. C, Western blot analysis (left) and quantification of protein levels (right) of siRNA-mediated transient knockdown of PD-L1 in DU145 cells. Cells were treated with siRNA directed against PD-L1 (siPD-L1), nontargeting control siRNA (siCtrl), or transfectant reagent only (HPF) for 72 hours. Lysates were probed with the PD-L1 mAb clone EPR1161(2) (top) and PD-L1 mAb (E1L3N) XP (bottom). Protein levels were normalized against β-actin and quantified using a colorimetric assay. D, Western blot analysis (top) of DU145 cells using PD-L1 mAb clone EPR1161(2) with and without blocking peptide preincubation. Equal loading was shown by probing for β-actin. Immunohistochemical staining (bottom) of tonsillar and placental tissue using PD-L1 mAb clone EPR1161(2) with distinct membranous staining of tonsillar epithelium and placental trophoblasts (left) and primary antibody neutralization using blocking peptide (right).

Figure 1.

Validation of PD-L1 antibody specificity. A, flow cytometric analysis of untreated DU145 cells compared with concentration-matched mouse IgG isotype control showed PD-L1 membrane expression in 40% of cells. B, Western blot analysis of untreated DU145 and MCF7 cells probed with the PD-L1 mAb clone EPR1161(2) (left) and the validated PD-L1 mAb (E1L3N) XP (right). Equal loading was shown by probing for β-actin. C, Western blot analysis (left) and quantification of protein levels (right) of siRNA-mediated transient knockdown of PD-L1 in DU145 cells. Cells were treated with siRNA directed against PD-L1 (siPD-L1), nontargeting control siRNA (siCtrl), or transfectant reagent only (HPF) for 72 hours. Lysates were probed with the PD-L1 mAb clone EPR1161(2) (top) and PD-L1 mAb (E1L3N) XP (bottom). Protein levels were normalized against β-actin and quantified using a colorimetric assay. D, Western blot analysis (top) of DU145 cells using PD-L1 mAb clone EPR1161(2) with and without blocking peptide preincubation. Equal loading was shown by probing for β-actin. Immunohistochemical staining (bottom) of tonsillar and placental tissue using PD-L1 mAb clone EPR1161(2) with distinct membranous staining of tonsillar epithelium and placental trophoblasts (left) and primary antibody neutralization using blocking peptide (right).

Close modal

PD-L1 expression in prostate cancer

Detailed clinicopathologic characteristics of the patient cohorts included in the present study are shown in Table 1. Tumoral immunoreactivity of PD-L1 was uniformly homogenous, and scoring results were highly concordant (κ = 0.75, P < 0.001). PD-L1 expression was detected at the membrane or in the cytoplasm of the tumor cells (Fig. 2). Additionally, stromal and tumor infiltrating lymphocytes displayed a strong PD-L1 immunoreactivity.

Figure 2.

Immunohistochemical analysis of PD-L1 in prostate cancer. Representative immunohistochemical staining showing strong (A), moderate (B), and weak (C) membranous PD-L1 expression in epithelial tumor cells. Negative staining (D) was observed in a minority of cases.

Figure 2.

Immunohistochemical analysis of PD-L1 in prostate cancer. Representative immunohistochemical staining showing strong (A), moderate (B), and weak (C) membranous PD-L1 expression in epithelial tumor cells. Negative staining (D) was observed in a minority of cases.

Close modal

In the training cohort, 52.2% of cases (109/209) expressed moderate to high levels of PD-L1, which positively correlated with proliferation (Ki-67, P < 0.001, τ = 0.26) and AR expression (P < 0.001, τ = 0.21). These results were confirmed by performing the χ2 test (Table 1). Furthermore, an association of PD-L1 with Gleason score was found (P = 0.004; Table 1). The introduction of a cutoff for patients' stratification is required for a Kaplan–Meier analysis. However, data dichotomization based on a cutoff leads to a loss of information on the one hand and a multiple testing problem due to the multitude of possible cutoffs on the other hand. Therefore, PD-L1 expression was initially analyzed as a continuous variable. In univariate proportional hazards model analysis, semi-quantitative PD-L1 expression was strongly prognostic for BCR [P = 0.004; HR, 2.37; 95% confidence interval (CI), 1.32–4.25; Table 2]. Kaplan–Meier survival analysis of PD-L1 expression dichotomized by median confirmed that high PD-L1 expression was associated with significantly reduced BCR-free survival (P = 0.022; Fig. 3A).

Figure 3.

Survival analysis in primary prostate cancer after radical prostatectomy. Kaplan–Meier analysis of BCR-free survival in 209 (training cohort, A) and 611 (test cohort, B) prostate cancer patients stratified by PD-L1 expression.

Figure 3.

Survival analysis in primary prostate cancer after radical prostatectomy. Kaplan–Meier analysis of BCR-free survival in 209 (training cohort, A) and 611 (test cohort, B) prostate cancer patients stratified by PD-L1 expression.

Close modal
Table 2.

Univariate and multivariate Cox analyses on BCR-free survival in the training (n = 209) and validation cohort (n = 611) of prostate cancer cases treated by radical prostatectomy

Univariate Cox analysisMultivariate Cox analysis
Training cohortTest cohortAll patients
HR (95% CI)PHR (95% CI)PHR (95% CI)P
pT category (pT2 reference) 
 pT3 and pT4 1.93 (1.08–3.45) 0.027 4.62 (3.03–7.03) <0.001 2.20 (1.49–3.24) <0.001 
Gleason score (7 reference) 
 <7 0.37 (0.16–0.86)  0.37 (0.20–0.67)  0.43 (0.26–0.71)  
 >7 4.08 (2.02–8.21) <0.001 2.39 (1.55–3.69) <0.001 1.95 (1.32–2.86) <0.001 
Surgical margin (R0 reference) 
 R1 1.94 (1.07–3.52) 0.030 2.84 (1.90–4.25) <0.001 1.44 (1.00–2.09) 0.053 
PD-L1 (continuous variable) 2.37 (1.32–4.25) 0.004 1.49 (1.10–2.02) 0.011 1.46 (1.11–1.92) 0.007 
Preoperative PSA level 1.01 (0.99–1.02) 0.41 1.05 (1.02–1.09) 0.002 1.00 (0.99–1.01) 0.75 
Univariate Cox analysisMultivariate Cox analysis
Training cohortTest cohortAll patients
HR (95% CI)PHR (95% CI)PHR (95% CI)P
pT category (pT2 reference) 
 pT3 and pT4 1.93 (1.08–3.45) 0.027 4.62 (3.03–7.03) <0.001 2.20 (1.49–3.24) <0.001 
Gleason score (7 reference) 
 <7 0.37 (0.16–0.86)  0.37 (0.20–0.67)  0.43 (0.26–0.71)  
 >7 4.08 (2.02–8.21) <0.001 2.39 (1.55–3.69) <0.001 1.95 (1.32–2.86) <0.001 
Surgical margin (R0 reference) 
 R1 1.94 (1.07–3.52) 0.030 2.84 (1.90–4.25) <0.001 1.44 (1.00–2.09) 0.053 
PD-L1 (continuous variable) 2.37 (1.32–4.25) 0.004 1.49 (1.10–2.02) 0.011 1.46 (1.11–1.92) 0.007 
Preoperative PSA level 1.01 (0.99–1.02) 0.41 1.05 (1.02–1.09) 0.002 1.00 (0.99–1.01) 0.75 

In the independent test cohort, moderate to high PD-L1 expression was detected in 61.7% of cases (377/611). The strong association of PD-L1 expression with AR (P < 0.001, τ = 0.16) and Ki-67 (P < 0.001, τ = 0.22) was substantiated in this cohort, whereas no association with Gleason score was verified. Along with known prognostic factors (pT status, Gleason score, surgical margins, and preoperative PSA levels), PD-L1 expression was shown to be significantly prognostic for BCR in univariate Cox proportional hazards model analysis (P = 0.011; HR = 1.49; 95% CI, 1.10–2.02; Table 2). After dichotomization, PD-L1 expression conferred a significantly shorter PSA relapse-free survival in Kaplan–Meier survival analysis (P = 0.009; Fig. 3B). An association of age and presurgical PSA with PD-L1 could only be shown in either the training or test cohort, respectively (Table 1).

In multivariate Cox proportional hazards model analysis of all patients, PD-L1 furthermore remained an independent prognostic factor of BCR (P = 0.007; HR, 1.46; 95% CI, 1.11–1.92; Table 2) when tested together with pT status, Gleason score, preoperative PSA, and surgical margins.

This study is the first to demonstrate that PD-L1 expression is not only highly prevalent in primary prostate cancer but is also an independent prognostic factor of disease progression in cohorts of primary tumors following RP. Consistent with previous studies, our results suggest an association of PD-L1 with aggressive tumor behavior in primary prostate cancer, indicating that PD-1/PD-L1 pathway activation assists the evasion of antitumor immune response, driving tumor proliferation and progression. Further studies in watchful waiting cohorts need to ascertain whether PD-L1 might also contribute to the identification of lethal prostate cancer.

We here comprehensively validated a novel monoclonal rabbit antibody against PD-L1 (clone EPR1161(2)) amenable to FFPE tissue and analyzed two large, well-characterized cohorts of primary prostate cancer after RP for PD-L1 expression. Our study shows that the immunoreactivity of clone EPR1161(2) for the detection of PD-L1 on FFPE tissue is specific and robust. In preliminary experiments (not shown), which we routinely conduct to establish a new antibody in our laboratory, we used different immunohistochemical platforms and found this antibody's performance to be independent of the detection system. It necessitates conventional antigen retrieval, but yields a crisp and clean signal with no background issues at all.

Increased expression of PD-L1 on tumor cells has previously been described for several malignancies, including glioblastoma, pancreatic, ovarian, breast, renal, head and neck, esophageal, and non–small cell lung cancer (37–42). Moreover, PD-L1 expression has been associated with poor prognosis and adverse clinicopathological characteristics (28, 31, 43–46). In line with these findings, we have found moderate to high PD-L1 expression in 52.2% to 61.7% of primary prostate cancers after RP. Multivariate analysis further revealed that high PD-L1 expression was significantly associated with reduced BCR-free survival independently of other clinicopathological factors, suggesting that expression of PD-L1 on tumor cells promotes tumor recurrence by interrupting antitumor immunity (29). In addition, we found a strong correlation of PD-L1 with tumor cell proliferation as estimated by Ki-67, which has been associated with a highly adverse prognosis and primary therapy failure in prostate cancer (47–50).

Preliminary data from a phase I study on an anti-PD1 mAb treatment (nivolumab) also indicated that immunohistochemically detected PD-L1 is a potential predictive biomarker for therapeutic blockade of the PD1/PD-L1 pathway (17). In the landmark study by Topalian and colleagues immunohistochemical assessment of PD-L1 in pretreatment cancer specimens from 42 patients revealed that objective response to treatment was seen exclusively in PD-L1–positive tumors (9/25, 36%; P = 0.006). More recent data suggest that PD-L1–positive tumors have higher response rates to agents targeting the PD1/PD-L1 pathway, although response to treatment was also recorded for PD-L1–negative cases (18–23). Notably, none of the 17 patients with metastatic CRPC included in the original study by Topalian and colleagues responded to anti-PD-1 treatment. For two cancers that were eligible for immunohistochemical analysis, no PD-L1 expression was detected (17), all of which suggesting that PD-1/PD-L1–targeted treatment was not particularly promising for prostate cancer patients. Although the sample size with only two prostate cancer specimens eligible for immunohistochemical analysis is clearly limited, the absence of PD-L1 expression might as well be due to the fact that only heavily pretreated CRPC were enrolled in this study. So far, it is speculative if the strong correlation of AR and PD-L1 observed in our study provides a viable explanation for low expression rates of PD-L1 and lack of response to anti-PD-1 treatment in CRPC (17). However, the high rate of PD-L1 positivity found in primary, and hence hormone-naïve prostate cancers, indicates that PD-1/PD-L1 pathway–targeted therapy might potentially be a novel treatment option, and immunohistochemical assessment of PD-L1 might accordingly represent a biomarker for the identification of patients eligible for this therapy.

Strikingly, Bishop and colleagues recently reported that CRPC patients resistant to enzalutamide showed elevated levels of PD-L1–expressing dendritic cells in blood (51). Additional cell line and xenograft experiments suggested that enzalutamide-resistant tumors might suppress immune response not only through intrinsic PD-L1 expression, but also via induction of PD-L1 expression on circulating dendritic cells. The authors concluded that PD-L1 expression on tumor cells might be a mechanism of non-AR–driven resistance to enzalutamide. In addition to patients with hormone-sensitive prostate cancer, these patients might therefore also potentially respond to anti-PD-L1 checkpoint blockade immunotherapy.

In order to elucidate the potential of PD-1/PD-L1–targeted therapy in prostate cancer, further studies are needed to clarify (i) if PD-L1 expression can contribute to identifying insignificant prostate cancer cases that may be spared immediate definitive therapy, (ii) if PD-L1 expression in primary tumors is predictive for response to anti-PD-L1 therapy, (iii) if castration-sensitive prostate cancers have low PD-L1 expression levels due to androgen suppression and subsequently, (iv) how much ADT itself downregulates PD-L1, and (v) if anti-PD-L1 therapy is effective at all in prostate cancer.

No potential conflicts of interest were disclosed.

Conception and design: H. Gevensleben, D. Dietrich, P. Brossart, G. Kristiansen

Development of methodology: H. Gevensleben, C. Golletz, M. Jung, M. Majores, B. Uhl, P. Brossart, G. Kristiansen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Gevensleben, C. Golletz, S. Steiner, M. Jung, M. Majores, J. Stein, B. Uhl, S. Müller, C. Stephan, K. Jung

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Gevensleben, D. Dietrich, T. Thiesler, K. Jung, P. Brossart, G. Kristiansen

Writing, review, and/or revision of the manuscript: H. Gevensleben, D. Dietrich, J. Stein, J. Ellinger, P. Brossart, G. Kristiansen

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Gevensleben, C. Golletz, S. Steiner, M. Majores, B. Uhl, S. Müller, J. Ellinger, C. Stephan, K. Jung, G. Kristiansen

Study supervision: D. Dietrich, P. Brossart

Other (tissue microarray—construction and quality assurance): M. Majores

Other (collection and database organization for TMA-related follow-up analyses): M. Majores

Other [providing data for clinicopathologic correlations (SPSS-database construction and collecting follow-up data): J. Stein

Other (construction of the tissue microarrays): J. Stein

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ferlay
J
,
Soerjomataram
I
,
Dikshit
R
,
Eser
S
,
Mathers
C
,
Rebelo
M
, et al
Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012
.
Int J Cancer
2015
;
136
:
E359
86
.
2.
Siegel
R
,
DeSantis
C
,
Virgo
K
,
Stein
K
,
Mariotto
A
,
Smith
T
, et al
Cancer treatment and survivorship statistics, 2012
.
CA Cancer J Clin
2012
;
62
:
220
41
.
3.
Heidenreich
A
,
Bastian
PJ
,
Bellmunt
J
,
Bolla
M
,
Joniau
S
,
van der Kwast
T
, et al
EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer
.
Eur Urol
2014
;
65
:
467
79
.
4.
Horwich
A
,
Parker
C
,
de Reijke
T
,
Kataja
V
. 
Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
2013
;
24
Suppl 6
:
vi106
14
.
5.
Laufer
M
,
Denmeade
SR
,
Sinibaldi
VJ
,
Carducci
MA
,
Eisenberger
MA
. 
Complete androgen blockade for prostate cancer: what went wrong?
J Urol
2000
;
164
:
3
9
.
6.
Beer
TM
,
Armstrong
AJ
,
Rathkopf
DE
,
Loriot
Y
,
Sternberg
CN
,
Higano
CS
, et al
Enzalutamide in metastatic prostate cancer before chemotherapy
.
N Engl J Med
2014
;
371
:
424
33
.
7.
Scher
HI
,
Fizazi
K
,
Saad
F
,
Taplin
ME
,
Sternberg
CN
,
Miller
K
, et al
Increased survival with enzalutamide in prostate cancer after chemotherapy
.
N Engl J Med
2012
;
367
:
1187
97
.
8.
de Bono
JS
,
Logothetis
CJ
,
Molina
A
,
Fizazi
K
,
North
S
,
Chu
L
, et al
Abiraterone and increased survival in metastatic prostate cancer
.
N Engl J Med
2011
;
364
:
1995
2005
.
9.
Fizazi
K
,
Scher
HI
,
Molina
A
,
Logothetis
CJ
,
Chi
KN
,
Jones
RJ
, et al
Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study
.
Lancet Oncol
2012
;
13
:
983
92
.
10.
Sweeney
CJ
,
Chen
YH
,
Carducci
M
,
Liu
G
,
Jarrard
DF
,
Eisenberger
M
, et al
Chemohormonal therapy in metastatic hormone-sensitive prostate cancer
.
N Engl J Med
2015
;
373
:
737
46
.
11.
Kwek
SS
,
Cha
E
,
Fong
L
. 
Unmasking the immune recognition of prostate cancer with CTLA4 blockade
.
Nat Rev Cancer
2012
;
12
:
289
97
.
12.
Schweizer
MT
,
Drake
CG
. 
Immunotherapy for prostate cancer: recent developments and future challenges
.
Cancer Metastasis Rev
2014
;
33
:
641
55
.
13.
Zou
W
,
Chen
L
. 
Inhibitory B7-family molecules in the tumour microenvironment
.
Nat Rev Immunol
2008
;
8
:
467
77
.
14.
Freeman
GJ
,
Long
AJ
,
Iwai
Y
,
Bourque
K
,
Chernova
T
,
Nishimura
H
, et al
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation
.
J Exp Med
2000
;
192
:
1027
34
.
15.
Keir
ME
,
Butte
MJ
,
Freeman
GJ
,
Sharpe
AH
. 
PD-1 and its ligands in tolerance and immunity
.
Annu Rev Immunol
2008
;
26
:
677
704
.
16.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQ
,
Hwu
WJ
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
17.
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
.
18.
Cho
DC
,
Sosman
JA
,
Sznol
M
,
Gordon
MS
. 
Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC)
.
J Clin Oncol
2013
;
31
(
suppl; abstr 4505
).
19.
Choueiri
TK
,
Fishman
MN
,
Escudier
BJ
,
Kim
JJ
. 
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
:
5s
:(
suppl; abstr 5012
).
20.
Powles
T
,
Eder
JP
,
Fine
GD
,
Braiteh
FS
,
Loriot
Y
,
Cruz
C
, et al
MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer
.
Nature
2014
;
515
:
558
62
.
21.
Seiwert
T
,
Seiwert
T
,
Seiwert
T
. 
A phase Ib study of MK-3475 in patients with human papillomavirus (HPV)-associated and non-HPV-associated head and neck (H/N) cancer
.
J Clin Oncol
2014
;
32
:
5s
,
2014 (suppl; abstr 6011)
22.
Ribas
A
,
Hodi
FS
,
Kefford
R
. 
Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL)
.
J Clin Oncol
2014
;
2014
;
32 (Suppl)
:
5S
.
23.
Rizvi
N
,
Rizvi
N
,
Rizvi
N
. 
Clinical trials of MPDL3280A (anti-PDL1) in patients (pts) with non-small cell lung cancer (NSCLC)
.
J Clin Oncol
2014
;
32
:
5s
,
2014 (suppl; abstr TPS8123)
.
24.
Chen
BJ
,
Chapuy
B
,
Ouyang
J
,
Sun
HH
,
Roemer
MG
,
Xu
ML
, et al
PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies
.
Clin Cancer Res
2013
;
19
:
3462
73
.
25.
Dong
H
,
Strome
SE
,
Salomao
DR
,
Tamura
H
,
Hirano
F
,
Flies
DB
, et al
Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion
.
Nat Med
2002
;
8
:
793
800
.
26.
Dong
H
,
Zhu
G
,
Tamada
K
,
Chen
L
. 
B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion
.
Nat Med
1999
;
5
:
1365
9
.
27.
Liang
SC
,
Latchman
YE
,
Buhlmann
JE
,
Tomczak
MF
,
Horwitz
BH
,
Freeman
GJ
, et al
Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses
.
Eur J Immunol
2003
;
33
:
2706
16
.
28.
Lyford-Pike
S
,
Peng
S
,
Young
GD
,
Taube
JM
,
Westra
WH
,
Akpeng
B
, et al
Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma
.
Cancer Res
2013
;
73
:
1733
41
.
29.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
30.
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
.
31.
Thompson
RH
,
Kuntz
SM
,
Leibovich
BC
,
Dong
H
,
Lohse
CM
,
Webster
WS
, et al
Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up
.
Cancer Res
2006
;
66
:
3381
5
.
32.
Kristiansen
G
,
Fritzsche
FR
,
Wassermann
K
,
Jager
C
,
Tolls
A
,
Lein
M
, et al
GOLPH2 protein expression as a novel tissue biomarker for prostate cancer: implications for tissue-based diagnostics
.
Br J Cancer
2008
;
99
:
939
48
.
33.
Meller
S
,
Bicker
A
,
Montani
M
,
Ikenberg
K
,
Rostamzadeh
B
,
Sailer
V
, et al
Myoglobin expression in prostate cancer is correlated to androgen receptor expression and markers of tumor hypoxia
.
Virchows Arch
2014
;
465
:
419
27
.
34.
Stein
J
,
Majores
M
,
Rohde
M
,
Lim
S
,
Schneider
S
,
Krappe
E
, et al
KDM5C is overexpressed in prostate cancer and is a prognostic marker for prostate-specific antigen-relapse following radical prostatectomy
.
Am J Pathol
2014
;
184
:
2430
7
.
35.
Epstein
JI
,
Egevad
L
,
Amin
MB
,
Delahunt
B
,
Srigley
JR
,
Humphrey
PA
, et al
The 2014 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma: Definition of Grading Patterns and Proposal for a New Grading System.
Am J Surg Pathol
2015
[Epub ahead of print]
.
36.
Soliman
H
,
Khalil
F
,
Antonia
S
. 
PD-L1 expression is increased in a subset of basal type breast cancer cells
.
PLoS One
2014
;
9
:
e88557
.
37.
Hamanishi
J
,
Mandai
M
,
Iwasaki
M
,
Okazaki
T
,
Tanaka
Y
,
Yamaguchi
K
, et al
Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer
.
Proc Natl Acad Sci U S A
2007
;
104
:
3360
5
.
38.
Karim
R
,
Jordanova
ES
,
Piersma
SJ
,
Kenter
GG
,
Chen
L
,
Boer
JM
, et al
Tumor-expressed B7-H1 and B7-DC in relation to PD-1+ T-cell infiltration and survival of patients with cervical carcinoma
.
Clin Cancer Res
2009
;
15
:
6341
7
.
39.
Konishi
J
,
Yamazaki
K
,
Azuma
M
,
Kinoshita
I
,
Dosaka-Akita
H
,
Nishimura
M
. 
B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression
.
Clin Cancer Res
2004
;
10
:
5094
100
.
40.
Ohigashi
Y
,
Sho
M
,
Yamada
Y
,
Tsurui
Y
,
Hamada
K
,
Ikeda
N
, et al
Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer
.
Clin Cancer Res
2005
;
11
:
2947
53
.
41.
Strome
SE
,
Dong
H
,
Tamura
H
,
Voss
SG
,
Flies
DB
,
Tamada
K
, et al
B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma
.
Cancer Res
2003
;
63
:
6501
5
.
42.
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
.
43.
Bigelow
E
,
Bever
KM
,
Xu
HY
,
Yager
A
,
Wu
A
,
Taube
J
, et al
Immunohistochemical staining of B7-H1 (PD-L1) on paraffin-embedded slides of pancreatic adenocarcinoma tissue
.
Jove-J Vis Exp
2013 Jan 3(71)
:
pii:4059
.
44.
Boland
JM
,
Kwon
ED
,
Harrington
SM
,
Wampfler
JA
,
Tang
H
,
Yang
P
, et al
Tumor B7-H1 and B7-H3 expression in squamous cell carcinoma of the lung
.
Clin Lung Cancer
2013
;
14
:
157
63
.
45.
Nakanishi
J
,
Wada
Y
,
Matsumoto
K
,
Azuma
M
,
Kikuchi
K
,
Ueda
S
. 
Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers
.
Cancer Immunol Immunother
2007
;
56
:
1173
82
.
46.
Zhang
Y
,
Huang
S
,
Gong
D
,
Qin
Y
,
Shen
Q
. 
Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer
.
Cell Mol Immunol
2010
;
7
:
389
95
.
47.
Botticelli
AR
,
Casali
AM
,
Botticelli
L
,
Zaffe
D
. 
Immunohistochemical detection of cell-cycle associated markers on paraffin embedded and formalin fixed needle biopsies of prostate cancer: correlation of p120 protein expression with AgNOR, PCNA/cyclin, Ki-67/MIB1 proliferation-scores and Gleason gradings
.
Eur J Histochem
1998
;
42
:
41
8
.
48.
Kaibuchi
T
,
Furuya
Y
,
Akakura
K
,
Masai
M
,
Ito
H
. 
Changes in cell proliferation and apoptosis during local progression of prostate cancer
.
Anticancer Res
2000
;
20
:
1135
9
.
49.
Scalzo
DA
,
Kallakury
BV
,
Gaddipati
RV
,
Sheehan
CE
,
Keys
HM
,
Savage
D
, et al
Cell proliferation rate by MIB-1 immunohistochemistry predicts postradiation recurrence in prostatic adenocarcinomas
.
Am J Clin Pathol
1998
;
109
:
163
8
.
50.
Visakorpi
T
. 
Proliferative activity determined by DNA flow cytometry and proliferating cell nuclear antigen (PCNA) immunohistochemistry as a prognostic factor in prostatic carcinoma
.
J Pathol
1992
;
168
:
7
13
.
51.
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
.