• PD-L1 expression in neoplastic cells or stromal cells is associated with poor or good prognosis in ATLL, respectively.

  • Distinction of expression pattern of PD-L1 might be important on the point of prognostic and therapeutic markers in ATLL.

Programmed cell death ligand 1 (PD-L1) is expressed on both tumor and tumor-infiltrating nonmalignant cells in lymphoid malignancies. The programmed cell death 1 (PD-1)/PD-L1 pathway suppresses host antitumor responses, although little is known about the significance of PD-1/PD-L1 expression in the tumor microenvironment. To investigate the clinicopathological impact of PD-L1 expression in adult T-cell leukemia/lymphoma (ATLL), we performed PD-L1 immunostaining in 135 ATLL biopsy samples. We observed 2 main groups: 1 had clear PD-L1 expression in lymphoma cells (nPD-L1(+), 7.4% of patients), and the other showed minimal expression in lymphoma cells (nPD-L1(−), 92.6%). Within the nPD-L1(−) group, 2 subsets emerged: the first displayed abundant PD-L1 expression in nonmalignant stromal cells of the tumor microenvironment (miPD-L1(+), 58.5%) and the second group did not express PD-L1 in any cell (PD-L1(−), 34.1%). nPD-L1(+) ATLL (median survival time [MST] 7.5 months, 95% CI [0.4-22.3]) had inferior overall survival (OS) compared with nPD-L1(−) ATLL (MST 14.5 months, 95% CI [10.1-20.0]) (P = .0085). Among nPD-L1(−) ATLL, miPD-L1(+) ATLL (MST 18.6 months, 95% CI [11.0-38.5]) showed superior OS compared with PD-L1(−) ATLL (MST 10.2 months, 95% CI [8.0-14.7]) (P = .0029). The expression of nPD-L1 and miPD-L1 maintained prognostic value for OS in multivariate analysis (P = .0322 and P = .0014, respectively). This is the first report describing the clinicopathological features and outcomes of PD-L1 expression in ATLL. More detailed studies will disclose clinical and biological significance of PD-L1 expression in ATLL.

Adult T-cell leukemia/lymphoma (ATLL) is defined as a peripheral T-cell neoplasm caused by human T-cell leukemia virus type 1 (HTLV-1).1  Of the 4 clinical subtypes of ATLL, acute type and lymphoma type are considered to follow an aggressive course compared with chronic type and smoldering type.2,3  Especially in aggressive ATLL and chronic type with poor prognostic factors, an effective therapy has not been established. Chemotherapies, zidovudine/interferon-α (IFN-α) treatment, and allogenic hematopoietic stem cell transplantation deliver disappointing outcomes.4-11  Thus, new treatment strategies associated with molecular oncogenic mechanisms are being explored.

Programmed cell death ligand 1 (PD-L1) is an immunomodulatory glycoprotein expressed on antigen-presenting cells.12  Binding of PD-L1 to the programmed cell death 1 (PD-1) receptor, which is present on the surface of T cells, plays a significant role in the regulation of effector T-cell responses by suppressing cytokine production, T-cell proliferation, and T-cell adhesion.13  In various types of solid tumors, PD-L1 expression on tumor cells has been reported to associate with tumor progression and poor prognosis14-20  through inhibition of local antitumor T-cell responses.14,15  Blockade of the PD-1/PD-L1 pathway by anti-PD-1 or anti-PD-L1 monoclonal antibodies may make antitumor responses effective.21-23 

In several types of lymphoid malignancies, PD-L1 is expressed on tumor cells and tumor-infiltrating nonmalignant cells, including macrophages.23,24  The presence of high levels of plasma-soluble PD-L1 and PD-L1 expression on lymphoma cells is associated with poor overall survival (OS) and is considered an important biomarker in diffuse large B-cell lymphoma (DLBCL).25,26  Moreover, blockade therapy of the PD-1/PD-L1 pathway showed a remarkable effect for Hodgkin lymphoma, in which PD-L1 is expressed on neoplastic cells.27  These results suggest that the PD-1/PD-L1 pathway might support tumor cell survival and that blockade of this pathway could be an effective therapeutic method in lymphoid malignancies other than DLBCL and Hodgkin lymphoma. Although a previous study reported that some proportion of ATLL patients showed PD-L1 expression,28  a detailed study of the clinicopathological characteristics of PD-L1 expression in ATLL is required to determine the effectiveness of anti-PD-1/PD-L1 therapies. In this study, we retrospectively investigated PD-L1 expression in ATLL and then evaluated the clinical significance between expression and clinicopathological features.

Patients and samples

We reviewed 135 biopsy samples from patients newly diagnosed as ATLL. Sixty-two patients were from the international Peripheral T-cell Lymphoma Project29  and 73 patients were admitted to the Department of Pathology, Kurume University, Japan from 2006 to 2012. A tissue microarray (TMA) was created, which contained each of the 135 samples in a formalin-fixed paraffin block, where each sample was placed in a 2-mm-diameter space. The same TMA specimen slides were used in the authors’ previous study.30  Some of the morphological and immunohistochemical results in this study are consistent with the previous study.30  This study includes patients diagnosed by biopsy sample and does not include patients diagnosed by peripheral blood sample.

Cases were reclassified according to the World Health Organization classification1  by experienced hematopathologists (H.M. and K.O.). The use of patient materials and clinical information was approved by the Research Ethics Committee of Kurume University and was in accordance with the Declaration of Helsinki.

Immunohistochemical staining other than PD-L1

The antibodies used for immunohistochemistry (IHC) were anti-CD3 (Clone F7.2.38; Dako Japan), anti-CD30 (Clone Ber-H2; Dako Japan), anti-CCR4 (POTELIGEO TEST; Kyowa Medex, Tokyo, Japan), anti-FoxP3 (Clone SP97; Abcam, Tokyo, Japan), and anti-PD-1 (Clone NAT105; Abcam). In IHC analyses, except for PD-L1, samples were considered positive if more than ∼20% of the neoplastic cells were stained.

Classification of ATLL according to PD-L1 expression

Immunohistochemical staining of PD-L1 was carried out by using 2.5-μm-thick, formalin-fixed, paraffin-embedded tissue sections from the TMA for all cases. The slides were deparaffinized with xylene, followed by ethanol. After rehydration with water, antigen retrieval was performed with EDTA buffer (pH 8.0) in a microwave oven at 95°C for 20 minutes. After cooling and rinsing with buffer, the slides were placed in a Dako autostainer (Dakocytomation, Kyoto, Japan). Endogenous peroxidase activity was blocked by incubating in 3% hydrogen peroxide for 5 minutes. Slides were incubated with anti-PD-L1 rabbit monoclonal antibodies (EPR1161(2); 1:200 dilution; ab174838, Abcam) overnight at 4°C. The slides were incubated with an EnVision+ System horseradish peroxide–labeled anti-rabbit polymer (K4003; Dakocytomation) for 30 minutes. Visualization of PD-L1 was performed using diaminobenzidine for 10 minutes. Slides were counterstained with hematoxylin, dehydrated with ethanol, and mounted under coverslips.

ATLL patients were categorized according to the patterns of PD-L1 expression by 2 experienced hematopathologists (H.M. and K.O.) as previously reported.25  The groups are defined as follows: (1) neoplastic PD-L1 (nPD-L1)-positive ATLL: patients with 50% or more of the lymphoma cells presenting membranous staining of PD-L1; (2) nPD-L1-negative ATLL: patients that had less than 50% PD-L1 staining of lymphoma cells; (3) microenvironmental PD-L1 (miPD-L1) -positive ATLL: a subpopulation of nPD-L1-negative ATLL patients where 10 or more PD-L1-positive nonmalignant stromal cells were observed per high power field (HPF); (4) PD-L1-negative ATLL: patients without PD-L1 expression in neoplastic cells or stromal cells (Figure 1; supplemental Figure 1, available on the Blood Web site).

Figure 1

Representative immunohistochemical analysis of PD-L1 expression in ATLL. (A) nPD-L1-positive ATLL. The plasma membranes of neoplastic cells were positive for PD-L1 (brown staining). (B) miPD-L1-positive ATLL. Neoplastic cells were negative for PD-L1. PD-L1-positive nonmalignant stromal cells were observed. Those stromal cells with PD-L1 expression were morphologically consistent with monocyte-derived cells, including macrophage or dendritic cells. (C) PD-L1-negative ATLL. PD-L1 was negative for both neoplastic cells and nonmalignant stromal cells. Original magnification ×600 for all panels.

Figure 1

Representative immunohistochemical analysis of PD-L1 expression in ATLL. (A) nPD-L1-positive ATLL. The plasma membranes of neoplastic cells were positive for PD-L1 (brown staining). (B) miPD-L1-positive ATLL. Neoplastic cells were negative for PD-L1. PD-L1-positive nonmalignant stromal cells were observed. Those stromal cells with PD-L1 expression were morphologically consistent with monocyte-derived cells, including macrophage or dendritic cells. (C) PD-L1-negative ATLL. PD-L1 was negative for both neoplastic cells and nonmalignant stromal cells. Original magnification ×600 for all panels.

Close modal

Quantitative analysis of PD-1-positive TILs

We performed immunohistochemical staining for detection of PD-1 (Clone NAT105; Abcam) and quantitative analysis of PD-1-positive tumor-infiltrating lymphocytes (TILs). For evaluation of PD-1-positive TILs in each patient, up to 5 representative HPFs were selected from the specimen. The total number of PD-1-positive TILs was counted in the representative fields, and the average PD-1-positive TILs per HPF was calculated for each patient.

Statistical analysis

Clinicopathological characteristics of the patients were compared by using the χ2 test or Fisher’s 2-sided exact test. Wilcoxon rank sum test was performed when comparing PD-1-positive TIL counts between different groups. OS was defined as the time from the day of diagnosis to the day of death or the date of last follow-up. The Kaplan-Meier method was used to estimate the OS distributions, and the log-rank test was performed to determine significant differences. Univariate and multivariate Cox proportional regression models were used to evaluate the proposed prognostic factors. P < .05 were considered statistically significant. JMP version 11.0 was used in all analyses.

Clinicopathological characteristics of ATLL patients

Table 1 summarizes the characteristics of the 135 patients. Median age was 67 years old with a range from 35 to 90. Seventy-six men and 59 women were included. Follow-up period ranged from 0.03 to 114.8 months with a median value of 10.9 months. With regard to Shimoyama classification, 1.8% (2/111) of the patients were categorized as smoldering type; 50.5% (56/111) were categorized as acute type, and 47.7% (53/111) were categorized as lymphoma type. A high score of the Japan Clinical Oncology Group prognostic index (JCOG-PI) was observed in 39.4% (52/132) of the patients. Regarding PD-L1 expression, 7.4% (10/135) of patients were categorized as nPD-L1 positive and 92.6% of the patients (125/135) were nPD-L1 negative. Among nPD-L1-negative patients, 58.5% (79/135) of the patients were miPD-L1 positive, and 34.1% (46/135) of the patients were PD-L1 negative. The average value of PD-1-positive TILs was 2.4 counts/HPF ranging from 0 to 38.4.

Table 1

Characteristics of ATLL

Clinical characteristicNo. (n = 135)%
Age, y, median (range) 67 (35-90) — 
Sex, male/female 76/59 56/44 
ECOG PS, 2-4 43/132 33 
Shimoyama classification   
 Smoldering type 2/111 
 Acute type 56/111 51 
 Lymphoma type 53/111 48 
B symptom 42/130 32 
Elevated LDH 77 57 
Hypercalcemia 21 16 
Elevated CRP 80/132 61 
Ann Arbor stage, III or IV 113/134 84 
Peripheral blood involvement 70/132 53 
Bone marrow involvement 30/106 28 
Skin lesion 23/134 17 
WBC, ×109/L, median (range) 6.6 (1.2-97.1) — 
Hemoglobin, g/dL, median (range) 12.9 (6.3-18.6) — 
Platelets, ×109/L, median (range) 213 (19-840) — 
IPI, high-intermediate or more 78/129 61 
JCOG-PI, high 52/132 39 
Treatment   
 Chemotherapy 117/133 88 
 Radiation 14/131 11 
 Transplantation 19/131 15 
 No chemotherapy or radiotherapy 13/133 10 
CR or CR(u) 35/116 30 
Clinical characteristicNo. (n = 135)%
Age, y, median (range) 67 (35-90) — 
Sex, male/female 76/59 56/44 
ECOG PS, 2-4 43/132 33 
Shimoyama classification   
 Smoldering type 2/111 
 Acute type 56/111 51 
 Lymphoma type 53/111 48 
B symptom 42/130 32 
Elevated LDH 77 57 
Hypercalcemia 21 16 
Elevated CRP 80/132 61 
Ann Arbor stage, III or IV 113/134 84 
Peripheral blood involvement 70/132 53 
Bone marrow involvement 30/106 28 
Skin lesion 23/134 17 
WBC, ×109/L, median (range) 6.6 (1.2-97.1) — 
Hemoglobin, g/dL, median (range) 12.9 (6.3-18.6) — 
Platelets, ×109/L, median (range) 213 (19-840) — 
IPI, high-intermediate or more 78/129 61 
JCOG-PI, high 52/132 39 
Treatment   
 Chemotherapy 117/133 88 
 Radiation 14/131 11 
 Transplantation 19/131 15 
 No chemotherapy or radiotherapy 13/133 10 
CR or CR(u) 35/116 30 
Pathological characteristicsNo. (n = 135)%
Morphological classification   
 Small cell variant 
 Medium cell variant 28 21 
 Large cell variant 57 42 
 Hodgkin like variant 
 Anaplastic variant 
 Pleomorphic variant 36 27 
IHC   
 nPD-L1 positive 10 
 nPD-L1 negative 125 93 
 mPD-L1 positive 79 59 
 PD-L1 positive 46 34 
Neoplastic cells   
 PD-1, positive 25 19 
 CD30, positive 36 27 
 FoxP3, positive 35 26 
 CCR4, positive 126 93 
PD-1-positive TIL, counts/HPF, average (range) 2.4 (0-38.4) — 
Pathological characteristicsNo. (n = 135)%
Morphological classification   
 Small cell variant 
 Medium cell variant 28 21 
 Large cell variant 57 42 
 Hodgkin like variant 
 Anaplastic variant 
 Pleomorphic variant 36 27 
IHC   
 nPD-L1 positive 10 
 nPD-L1 negative 125 93 
 mPD-L1 positive 79 59 
 PD-L1 positive 46 34 
Neoplastic cells   
 PD-1, positive 25 19 
 CD30, positive 36 27 
 FoxP3, positive 35 26 
 CCR4, positive 126 93 
PD-1-positive TIL, counts/HPF, average (range) 2.4 (0-38.4) — 

CRP, C-reactive protein; CR, complete response/remission; CR(u), uncertain complete response/remission; ECOG PS, Eastern Cooperative Oncology Group performance status; IPI, international prognostic index; LDH, lactate dehydrogenase; WBC, white blood cell.

Clinicopathological comparison between nPD-L1-positive and -negative ATLL

Table 2 shows statistical comparisons between PD-L1 expression patterns for different clinical parameters of ATLL patients. As for Shimoyama classification, 70% of acute-type patients (7/10) and 30% of lymphoma-type patients (3/10) were nPD-L1 positive. Among nPD-L1-negative patients, 2.0% (2/101) were categorized as smoldering-type ATLL; 45.5% (46/101) were acute type ATLL, and 52.5% (53/101) were lymphoma-type ATLL. Statistical significance was not observed between nPD-L1-positive and these subtypes (P = .39). Even when smoldering type was removed from the statistical analysis, statistical significance was not achieved (P = .20). Of the miPD-L1-positive patients, 3.0% (2/67) were smoldering type, 44.8% (30/67) were acute type, and 52.2% (35/67) were lymphoma type. Among PD-L1-negative patients, 47.1% (16/34) were classified as acute type and 52.9% of patients (18/34) were lymphoma type. There was no statistical significance between miPD-L1-positive and these Shimoyama classifications (P = .43). Even in statistical analysis where smoldering type was removed, significance was not observed (P = .93).

Table 2

Clinicopathological comparison among PD-L1 expression patterns in ATLL

nPD-L1 negative (n = 125)P
nPD-L1 positive (n = 10)miPD-L1 positive (n = 79)PD-L1 negative (n = 46)nPD-L1+ vs nPD-L1−
miPD-L1 + vs PD-L1−
CharacteristicNo.%No.%No.%P*P
Sex (male/female) 7/3 70/30 40/39 51/49 29/17 63/73 .51 .18 
Age, y         
Median (range) 66 (47–82) 66 (35–90) 64 (40–86) .78 .34 
ECOG PS, 2-4 50 26/77 34 12/45 27 .29 .41 
Shimoyama classification       .39 .43 
 Smoldering type 2/67 0/34   
 Acute type 70 30/67 45 16/34 47 [.20[.93] 
 Lymphoma type 30 35/67 52 18/34 53 
B symptoms 50 26/74 35 11 24 .29 .91 
Elevated serum LDH 60 43 54 28 61 1.00 .48 
Hypercalcemia 20 10 11 24 .65 .04 
Elevated CRP 80 44/76 58 28 61 .31 .75 
Ann Arbor stage, III or IV 10 100 64/78 82 39 85 .36 .69 
Peripheral blood involvement 60 41/76 54 23 50 .75 .67 
Bone marrow involvement 4/7 57 19/65 29 7/34 21 .098 .35 
Skin lesion 10 19/78 24 1.00 .01 
WBC, ×109/L, median (range) 14.8 (2.9-48.3) 8.6 (1.2-97.1) 9.4 (1.6-45.9) .17 .55 
Hemoglobin, g/dL, median (range) 12.0 (7.7-14.7) 12.4 (6.3-16.1) 13.2 (8.0-18.6) .49 .052 
Platelets, ×109/L, median (range) 193 (65-370) 222 (19-538) 240 (22-840) .24 .46 
IPI risk group, high (3-5) 90 42/74 57 27/45 60 .09* .73 
JCOG-PI, high 5/10 50 28/77 36 19/45 42 .51 .52 
Treatment         
 Chemotherapy 80 69/78 89 40/45 89 .34 .94 
 Radiotherapy 9/77 12 5/44 11 .60 1.00 
 Transplantation 10 10/77 13 8/44 18 1.00 .45 
 No therapy 20 9/78 12 2/45 .25 .33 
CR or CR(u) 2/8 25 22/67 33 11/41 27 1.00 .51 
Morphological classification       .51 .54 
Small-cell variant   
Medium-cell variant 20 19 24 15   
Large-cell variant 20 31 39 24 52   
Hodgkin-like variant   
Anaplastic variant 10   
Pleomorphic variant 50 20 25 11 24   
IHC         
 Neoplastic cells         
  PD-1, positive 16 20 20 .21 .93 
  CD30, positive 50 20 25 11 24 .13 .86 
  CCR4, positive 10 100 72 91 44 96 1.00 .48 
  FoxP3, positive 20 21 27 12 26 1.00 .95 
 PD-1 positive TIL, counts/HPF, average (range) 3.0 (0-24.4) 3.6 (0-38.4) 0.3 (0-5.2) .16 .0009 
nPD-L1 negative (n = 125)P
nPD-L1 positive (n = 10)miPD-L1 positive (n = 79)PD-L1 negative (n = 46)nPD-L1+ vs nPD-L1−
miPD-L1 + vs PD-L1−
CharacteristicNo.%No.%No.%P*P
Sex (male/female) 7/3 70/30 40/39 51/49 29/17 63/73 .51 .18 
Age, y         
Median (range) 66 (47–82) 66 (35–90) 64 (40–86) .78 .34 
ECOG PS, 2-4 50 26/77 34 12/45 27 .29 .41 
Shimoyama classification       .39 .43 
 Smoldering type 2/67 0/34   
 Acute type 70 30/67 45 16/34 47 [.20[.93] 
 Lymphoma type 30 35/67 52 18/34 53 
B symptoms 50 26/74 35 11 24 .29 .91 
Elevated serum LDH 60 43 54 28 61 1.00 .48 
Hypercalcemia 20 10 11 24 .65 .04 
Elevated CRP 80 44/76 58 28 61 .31 .75 
Ann Arbor stage, III or IV 10 100 64/78 82 39 85 .36 .69 
Peripheral blood involvement 60 41/76 54 23 50 .75 .67 
Bone marrow involvement 4/7 57 19/65 29 7/34 21 .098 .35 
Skin lesion 10 19/78 24 1.00 .01 
WBC, ×109/L, median (range) 14.8 (2.9-48.3) 8.6 (1.2-97.1) 9.4 (1.6-45.9) .17 .55 
Hemoglobin, g/dL, median (range) 12.0 (7.7-14.7) 12.4 (6.3-16.1) 13.2 (8.0-18.6) .49 .052 
Platelets, ×109/L, median (range) 193 (65-370) 222 (19-538) 240 (22-840) .24 .46 
IPI risk group, high (3-5) 90 42/74 57 27/45 60 .09* .73 
JCOG-PI, high 5/10 50 28/77 36 19/45 42 .51 .52 
Treatment         
 Chemotherapy 80 69/78 89 40/45 89 .34 .94 
 Radiotherapy 9/77 12 5/44 11 .60 1.00 
 Transplantation 10 10/77 13 8/44 18 1.00 .45 
 No therapy 20 9/78 12 2/45 .25 .33 
CR or CR(u) 2/8 25 22/67 33 11/41 27 1.00 .51 
Morphological classification       .51 .54 
Small-cell variant   
Medium-cell variant 20 19 24 15   
Large-cell variant 20 31 39 24 52   
Hodgkin-like variant   
Anaplastic variant 10   
Pleomorphic variant 50 20 25 11 24   
IHC         
 Neoplastic cells         
  PD-1, positive 16 20 20 .21 .93 
  CD30, positive 50 20 25 11 24 .13 .86 
  CCR4, positive 10 100 72 91 44 96 1.00 .48 
  FoxP3, positive 20 21 27 12 26 1.00 .95 
 PD-1 positive TIL, counts/HPF, average (range) 3.0 (0-24.4) 3.6 (0-38.4) 0.3 (0-5.2) .16 .0009 

Abbreviations are explained in Table 1.

*

nPD-L1 positive versus nPD-L1 negative.

miPD-L1 positive versus PD-L1 negative.

Fisher’s exact test; values in brackets indicate P value between acute type and lymphoma type.

Clinicopathological comparison between miPD-L1-positive and PD-L1-negative ATLL

Compared with the PD-L1-negative group, the miPD-L1-positive group was significantly associated with a low proportion of hypercalcemia (P = .042) and a high proportion of skin lesions (P = .014).

Quantitative analysis of PD-1-positive TILs

In regards to PD-1-positive TILs, the nPD-L1-positive group showed an average of 3.0 counts/HPF (median 0.3; range 0-24.4) and the nPD-L1-negative group averaged 2.3 counts/HPF (median 0; range 0-38.4). Among nPD-L1-negative patients, the miPD-L1-positive group averaged 3.6 counts/HPF (median 0; range, 0-38.4) and the PD-L1-negative group had 0.3 counts/HPF (median 0; range 0-5.2). The nPD-L1-positive group did not show a significant difference in PD-1-positive TILs compared with the nPD-L1-negative group (P = .162); however, there was a significantly higher number of PD-1-positive TILs in the nPD-L1-positive group compared with the PD-L1-negative group (P = .0041). miPD-L1-positive patients had significantly more PD-1-positive TILs than the PD-L1-negative group (P = .0009).

OS in ATLL patients according to PD-L1 expression

nPD-L1-positive patients (median survival time [MST] 7.5 months, 95% confidence interval [CI] [0.4-22.3]) had inferior OS compared with nPD-L1-negative patients (MST 14.5 months, 95% CI [10.1-20.0]) (P = .0085; Figure 2A). Among nPD-L1-negative patients, miPD-L1-positive patients (MST 18.6 months, 95% CI [11.0-38.5]) showed superior OS than PD-L1-negative patients (MST 10.2 months, 95% CI [8.0-14.7]) (P = .0029; Figure 2B).

Figure 2

OS of ATLL among the 4 PD-L1 expression groups. (A) nPD-L1 positive ATLL shows worse OS compared with nPD-L1 negative ATLL (P = .0085). (B) miPD-L1 positive ATLL shows better OS compared with PD-L1 negative ATLL (P = .0029).

Figure 2

OS of ATLL among the 4 PD-L1 expression groups. (A) nPD-L1 positive ATLL shows worse OS compared with nPD-L1 negative ATLL (P = .0085). (B) miPD-L1 positive ATLL shows better OS compared with PD-L1 negative ATLL (P = .0029).

Close modal

Univariate analyses identified the following variables as prognostic factors: nPD-L1 expression (hazard ratio [HR], 2.455; 95% CI, 1.144-4.642; P = .0234), miPD-L1 expression (HR, 0.525; 95% CI, 0.341-0.811; P = .0039), high JCOG-PI (HR, 1.777; 95% CI, 1.149-2.710; P = .0103), and age over 70 (HR, 1.647; 95% CI, 1.060-2.515; P = .0269) (Table 3). Multivariate analysis with nPD-L1 expression and other factors showed that nPD-L1 expression remained a significant prognostic factor (HR, 2.373; 95% CI, 1.084-4.625; P = .0322) (Table 3, Multivariate analysis 1). A similar multivariate analysis with miPD-L1 expression indicated that miPD-L1 expression persisted as a significant prognostic factor (HR, 0.481; 95% CI, 0.309-0.750; P = .0014) (Table 3, Multivariate analysis 2).

Table 3

Prognostic factors affecting the OS of patients with ATLL

CharacteristicsUnivariate analysisMultivariate analysis 1*Multivariate analysis 2
HR (95% CI)PHR (95% CI)PHR (95% CI)P
nPD-L1, positive (vs nPD-L1 negative) 2.455 (1.144-4.642) .0234 2.373 (1.084-4.625) .0322   
miPD-L1, positive (vs PD-L1 negative) 0.525 (0.341-0.811) .0039   0.48 1(0.309-0.750) .0014 
JCOG-PI, high (vs low) 1.777 (1.149-2.710) .0103 1.750 (1.114-2.721) .0157 1.797 (1.112-2.863) .0172 
Age, >70 (vs ≤70) 1.647 (1.060-2.515) .0269 1.694 (1.080-2.612) .0224 1.656 (1.027-2.615) .0389 
Ann Arbor stage, III/IV (vs I/II) 1.776 (0.988-3.530) .0551 1.640 (0.903-3.283) .1081 1.829 (0.999-3.687) .0503 
LDH, elevated (vs normal) 1.000 (0.664-1.513) .9986 0.850 (0.554-1.309) .4575 0.945 (0.599-1.671) .8089 
CharacteristicsUnivariate analysisMultivariate analysis 1*Multivariate analysis 2
HR (95% CI)PHR (95% CI)PHR (95% CI)P
nPD-L1, positive (vs nPD-L1 negative) 2.455 (1.144-4.642) .0234 2.373 (1.084-4.625) .0322   
miPD-L1, positive (vs PD-L1 negative) 0.525 (0.341-0.811) .0039   0.48 1(0.309-0.750) .0014 
JCOG-PI, high (vs low) 1.777 (1.149-2.710) .0103 1.750 (1.114-2.721) .0157 1.797 (1.112-2.863) .0172 
Age, >70 (vs ≤70) 1.647 (1.060-2.515) .0269 1.694 (1.080-2.612) .0224 1.656 (1.027-2.615) .0389 
Ann Arbor stage, III/IV (vs I/II) 1.776 (0.988-3.530) .0551 1.640 (0.903-3.283) .1081 1.829 (0.999-3.687) .0503 
LDH, elevated (vs normal) 1.000 (0.664-1.513) .9986 0.850 (0.554-1.309) .4575 0.945 (0.599-1.671) .8089 
*

The variables included in multivariate analysis 1 for OS were nPD-L1, JCOG-PI, age, Ann Arbor stage, and LDH.

The variables included in multivariate analysis 2 for OS were miPD-L1, JCOG-PI, age, Ann Arbor stage, and LDH.

In the present study, we demonstrated for the first time that PD-L1 expression on ATLL tumor cells is an independent poor prognostic factor for OS and that PD-L1 expression on nonmalignant stromal cells was an independent good prognostic factor for OS in ATLL.

Many previous reports have showed that high PD-L1 expression in tumor cells is associated with clinical progression and worse OS in other neoplasms, including DLBCL.8,25,31  Although PD-L1-expressing cells have various mechanisms to escape T-cell immunity,15  the bad prognoses associated with PD-L1 expression in neoplastic cells is thought to be caused by the activity of the PD-1/PD-L1 pathway. This pathway induces T-cell-mediated immune suppression in the tumor microenvironment.14  Some in vitro studies demonstrated that the binding of PD-L1 in neoplastic cells to PD-1 on T cells negatively regulates synthesis of interleukin-2 and IFN-γ. Suppressed secretion of interleukin-2 and IFN-γ from PD-1-positive T cells consequently leads to apoptosis of CD8+ cytotoxic T lymphocytes.14,31  DLBCL cell lines with PD-L1 expression also exhibited inhibition of T-cell proliferation and IFN-γ secretion by tumor-associated T cells.23  Although these results are not enough to explain the precise underlying mechanisms, they do suggest that the PD-1/PD-L1 pathway may contribute to a worse clinical outcome in ATLL patients with PD-L1 expression on neoplastic cells.

This study found that PD-L1 expression on stromal cells in the tumor microenvironment correlated with better prognosis. Stromal cells with PD-L1 expression showed roundlike nuclei with abundant and poorly defined cytoplasms. These cells may be monocyte-derived cells, including macrophage and dendritic cells. Some of the stromal cells with PD-L1 expression were classified as macrophages based on IHC (supplemental Figure 1). Previous reports suggest that PD-L1 may be upregulated in activated macrophages and dendritic cells.12,32  As for lymphoid malignancies, macrophages in the microenvironment have been shown to exhibit PD-L1 expression in Hodgkin lymphoma and DLBCL.24,33  Some kinds of dendritic cells are also known to express PD-L1 in reactive lymphoid tissue.34  However, the biological significance of PD-L1 expression on macrophages and dendritic cells in the tumor microenvironment remains unclear. More detailed research of stromal cells with PD-L1 expression is needed to understand the biological effect in lymphoid malignancies.

Concerning the association between better prognosis and PD-L1 expression, few studies have demonstrated the association between PD-L1 expression in stromal cells and good prognosis.35,36  In contrast, PD-L1 expression in neoplastic cells is reported to be a good prognostic factor in several malignancies.35-37  In those malignancies, PD-L1 expression in neoplastic and stromal cells was associated with increased IFN-γ and TILs, including CD8-positive and regulatory T cells.35-37  An in vitro study reported that CD8-positive T cells and IFN-γ induced PD-L1 expression of stromal cells in the microenvironment.38  These results suggest the possibility that the better prognosis of miPD-L1 expression in ATLL is not the result of an upregulated PD-1/PD-L1 pathway; miPD-L1 expression may simply be an indicator of a different immune condition in which antitumor immunity is much more activated.

In our quantitative analysis of PD-1-positive TILs, the numbers of PD-1-positive TILs were higher in nPD-L1- and miPD-L1-positive patients than those in PD-L1-negative patients. In various other cancers such as melanoma and sarcoma, PD-L1 expression on neoplastic cells and increased PD-1-positive TILs cause worse prognosis.39-41  These results suggest that, in ATLL, neoplastic cells with PD-L1 expression and PD-1-positive TILs might activate the PD-L1/PD-1 pathway that suppresses the T-cell immune response against neoplastic cells. On the other hand, the significance of a high number of PD-1-positive TILs in miPD-L1-positive patients associated with better prognosis is unknown. In follicular lymphoma and DLBCL, a high number of PD-1-positive TILs are associated with better prognosis regardless of PD-L1 expression.25,42,43  Nevertheless, most malignancies with a high number of PD-1-positive TILs show worse OS probably due to activation of the PD-L1/PD-1 pathway.39-41  The association between better prognosis and high levels of PD-1-positive TILs in FL and DLBCL is presumably caused not by the characteristics of PD-1-positive T cells associated with tumor-immunity, but by those of bystander follicular helper T cells. These cells coexist with neoplastic B cells in the germinal center and might not play a specific role in tumor immunity.25,42,43  A high level of PD-1-positive TILs and PD-L1 expression in stromal cells in ATLL could similarly indicate other pathological or immune conditions. More detailed study of PD-1-positive TILs will elucidate the mechanism underlying these associations.

Some studies have shown an association with PD-L1 expression and HTLV-1 infection. A previous report25  showed that PD-L1 expression was not observed in lymphocytes of asymptomatic HTLV-1 carriers, but PD-L1 expression did occur in some ATLL cases. Furthermore, when PD-L1-positive neoplastic cells from ATLL patients were treated with anti-PD-L1 antagonistic antibody, the HTLV-1-specific CD8+ T-cell response was upregulated. These results suggest that the PD-1/PD-L1 pathway may play a significant role in maintenance of persistent HTLV-1 infection25 ; specifically, this pathway might promote immune escape of neoplastic cells and encourage progression of ATLL. In regards to PD-L1 expression on nonmalignant cells in the tumor microenvironment, we could not show the association between other markers of infection (CCR4) and PD-L1 expression in microenvironment cells. More detailed studies could possibly reveal an association.

Immunotherapy against the PD1/PD-L1 pathway is reported as an effective treatment in several malignancies.22  In DLBCL and Hodgkin lymphoma, anti-PD-1 monoclonal antibody treatment (pidilizumab, nivolumab, and so forth) achieved marked improvement of clinical outcome.27,44  This study suggests that immunotherapy targeting the PD1/PD-L1 pathway for nPD-L1-positive ATLL could result in a better clinical outcome. On the other hand, administration of immunotherapy against the PD-1/PD-L1 pathway for miPD-L1-positive ATLL should be carefully considered because stromal cells with PD-L1 expression and PD-1-positive TILs possibly could lead to better clinical outcome. In any case, careful clinical studies are needed to determine whether blockade of the PD-1/PD-L1 pathway can be an effective treatment strategy for patients of ATLL according to PD-L1 expression patterns.

There are some limitations in this study. First, the number of cases is relatively small. Regardless, statistical significance was partially observed. Further studies with larger populations are warranted to validate our results. Second, this study includes only patients diagnosed by biopsy specimen and does not contain diagnoses by peripheral blood sample. The selection of only 1 sampling method might lend bias to these findings. Further studies that include patients diagnosed by peripheral blood sample are required to confirm our results. Third, this study could not evaluate all subtypes of Shimoyama classification partially because of shortages of clinical information for some patients. More studies, including all Shimoyama classifications, are needed to examine whether these findings are true for chronic-type and smoldering-type ATLL. Fourth, PD-L1 expression in this study was evaluated only with IHC. Additional analyses for genomic abnormalities and transcriptional levels of PD-L1 will be required to understand the biological and clinical significance of PD-L1 expression. Last, it is still unknown whether PD-1/PD-L1 blockade should actually be administered to ATLL with PD-L1 expression on neoplastic cells or stromal cells. Future clinical analyses are needed to determine if PD-1/PD-L1 blockade will lead to better clinical outcomes for ATLL patients.

In conclusion, we have demonstrated that PD-L1 expression on neoplastic cells and stromal cells in ATLL induces worse or better prognosis, respectively. These results suggest that the association between clinical outcome and PD-L1 expression in neoplastic cells and stromal cells could be caused by tumor microenvironment, including antitumor immunity. The administration of PD-1/PD-L1 blockade should be selected carefully according to expression patterns of PD-L1 in ATLL because PD-L1 expression in stromal cells may be associated with better clinical outcome. More detailed studies will disclose the clinical and biological significance of PD-L1 expression in ATLL.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Kazutaka Nakashima, Mayumi Miura, Kanoko Miyazaki, Yuki Morotomi, Chie Kuroki, and Kaoruko Nagatomo for their technical assistance.

This research was supported by a Grant-in-Aid for Scientific Research by the Ministry of Education, Culture, Sports, Science, and Technology, Japan, by Japan Society for the Promotion of Science (KAKENHI) grant number JP16K19093, and by a grant from Practical Research for Innovative Cancer Control Agency for Medical Research and Development (15ck0106015h0002) (Y.I., M.S., and K.O.). This study was performed using tissue samples obtained from the many hospitals and/or institutes included in the Kyushu Lymphoma Study Group: Department of Medicine and Biosystemic Science, Kyushu University Faculty of Medicine; Department of Hematology, Karatsu Red Cross Hospital; Department of Hematology, Sasebo City General Hospital; Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University; and Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University. The authors express their appreciation for the donated samples.

Contribution: H.M. and K.O. were responsible for conception and design; T.K., K. Kato, H.T., and Y.I. provided study materials or patients; H.M., J.K., T.K., J.S., Y.S., D.K., and K. Kawamoto performed collection and assembly of data; H.M., J.K., T.K., N.Y., S.Y., M.S., and K.O. performed data analysis and interpretation; H.M. wrote the manuscript; and all authors approved the final manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Hiroaki Miyoshi, Department of Pathology, School of Medicine, Kurume University, Asahimachi 67, Kurume-city, Fukuoka 830-0011, Japan; e-mail: miyoshi_hiroaki@med.kurume-u.ac.jp.

1
Ohshima
 
K
Jaffe
 
ES
Kikuchi
 
M
Adult T-cell leukemia/lymphoma.
World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
Lyon
IARC Press
(pg. 
281
-
284
)
2
Shimoyama
 
M
Diagnostic criteria and classification of clinical subtypes of adult T-cell leukaemia-lymphoma. A report from the Lymphoma Study Group (1984-87).
Br J Haematol
1991
, vol. 
79
 
3
(pg. 
428
-
437
)
3
Fukushima
 
T
Nomura
 
S
Shimoyama
 
M
, et al. 
Japan Clinical Oncology Group (JCOG) prognostic index and characterization of long-term survivors of aggressive adult T-cell leukaemia-lymphoma (JCOG0902A).
Br J Haematol
2014
, vol. 
166
 
5
(pg. 
739
-
748
)
4
Tsukasaki
 
K
Tobinai
 
K
Human T-cell lymphotropic virus type I-associated adult T-cell leukemia-lymphoma: new directions in clinical research.
Clin Cancer Res
2014
, vol. 
20
 
20
(pg. 
5217
-
5225
)
5
Ishitsuka
 
K
Tamura
 
K
Human T-cell leukaemia virus type I and adult T-cell leukaemia-lymphoma.
Lancet Oncol
2014
, vol. 
15
 
11
(pg. 
e517
-
e526
)
6
Utsunomiya
 
A
Choi
 
I
Chihara
 
D
Seto
 
M
Recent advances in the treatment of adult T-cell leukemia-lymphomas.
Cancer Sci
2015
, vol. 
106
 
4
(pg. 
344
-
351
)
7
Katsuya
 
H
Ishitsuka
 
K
Utsunomiya
 
A
, et al. 
ATL-Prognostic Index Project
Treatment and survival among 1594 patients with ATL.
Blood
2015
, vol. 
126
 
24
(pg. 
2570
-
2577
)
8
Bazarbachi
 
A
Plumelle
 
Y
Carlos Ramos
 
J
, et al. 
Meta-analysis on the use of zidovudine and interferon-alfa in adult T-cell leukemia/lymphoma showing improved survival in the leukemic subtypes.
J Clin Oncol
2010
, vol. 
28
 
27
(pg. 
4177
-
4183
)
9
Hodson
 
A
Crichton
 
S
Montoto
 
S
, et al. 
Use of zidovudine and interferon alfa with chemotherapy improves survival in both acute and lymphoma subtypes of adult T-cell leukemia/lymphoma.
J Clin Oncol
2011
, vol. 
29
 
35
(pg. 
4696
-
4701
)
10
Chihara
 
D
Ito
 
H
Matsuda
 
T
, et al. 
Association between decreasing trend in the mortality of adult T-cell leukemia/lymphoma and allogeneic hematopoietic stem cell transplants in Japan: analysis of Japanese vital statistics and Japan Society for Hematopoietic Cell Transplantation (JSHCT).
Blood Cancer J
2013
, vol. 
3
 
11
pg. 
e159
 
11
Ishida
 
T
Hishizawa
 
M
Kato
 
K
, et al. 
Allogeneic hematopoietic stem cell transplantation for adult T-cell leukemia-lymphoma with special emphasis on preconditioning regimen: a nationwide retrospective study.
Blood
2012
, vol. 
120
 
8
(pg. 
1734
-
1741
)
12
Keir
 
ME
Francisco
 
LM
Sharpe
 
AH
PD-1 and its ligands in T-cell immunity.
Curr Opin Immunol
2007
, vol. 
19
 
3
(pg. 
309
-
314
)
13
Pedoeem
 
A
Azoulay-Alfaguter
 
I
Strazza
 
M
Silverman
 
GJ
Mor
 
A
Programmed death-1 pathway in cancer and autoimmunity.
Clin Immunol
2014
, vol. 
153
 
1
(pg. 
145
-
152
)
14
Dong
 
H
Strome
 
SE
Salomao
 
DR
, et al. 
Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion.
Nat Med
2002
, vol. 
8
 
8
(pg. 
793
-
800
)
15
Zou
 
W
Chen
 
L
Inhibitory B7-family molecules in the tumour microenvironment.
Nat Rev Immunol
2008
, vol. 
8
 
6
(pg. 
467
-
477
)
16
Thompson
 
RH
Dong
 
H
Kwon
 
ED
Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy.
Clin Cancer Res
2007
, vol. 
13
 
2 Pt 2
(pg. 
709s
-
715s
)
17
Routh
 
JC
Ashley
 
RA
Sebo
 
TJ
, et al. 
B7-H1 expression in Wilms tumor: correlation with tumor biology and disease recurrence.
 
J Urol. 2008;179(5):1954-1959; discussion 1959-1960
18
Gao
 
Q
Wang
 
XY
Qiu
 
SJ
, et al. 
Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma.
Clin Cancer Res
2009
, vol. 
15
 
3
(pg. 
971
-
979
)
19
Nomi
 
T
Sho
 
M
Akahori
 
T
, et al. 
Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer.
Clin Cancer Res
2007
, vol. 
13
 
7
(pg. 
2151
-
2157
)
20
Ohigashi
 
Y
Sho
 
M
Yamada
 
Y
, 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
, vol. 
11
 
8
(pg. 
2947
-
2953
)
21
Armand
 
P
Nagler
 
A
Weller
 
EA
, et al. 
Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial.
J Clin Oncol
2013
, vol. 
31
 
33
(pg. 
4199
-
4206
)
22
Brahmer
 
JR
Tykodi
 
SS
Chow
 
LQ
, et al. 
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer.
N Engl J Med
2012
, vol. 
366
 
26
(pg. 
2455
-
2465
)
23
Andorsky
 
DJ
Yamada
 
RE
Said
 
J
Pinkus
 
GS
Betting
 
DJ
Timmerman
 
JM
Programmed death ligand 1 is expressed by non-hodgkin lymphomas and inhibits the activity of tumor-associated T cells.
Clin Cancer Res
2011
, vol. 
17
 
13
(pg. 
4232
-
4244
)
24
Chen
 
BJ
Chapuy
 
B
Ouyang
 
J
, et al. 
PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies.
Clin Cancer Res
2013
, vol. 
19
 
13
(pg. 
3462
-
3473
)
25
Kiyasu
 
J
Miyoshi
 
H
Hirata
 
A
, et al. 
Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma.
Blood
2015
, vol. 
126
 
19
(pg. 
2193
-
2201
)
26
Rossille
 
D
Gressier
 
M
Damotte
 
D
, et al. 
Groupe Ouest-Est des Leucémies et Autres Maladies du Sang; Groupe Ouest-Est des Leucémies et Autres Maladies du Sang
High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial.
Leukemia
2014
, vol. 
28
 
12
(pg. 
2367
-
2375
)
27
Ansell
 
SM
Lesokhin
 
AM
Borrello
 
I
, et al. 
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma.
N Engl J Med
2015
, vol. 
372
 
4
(pg. 
311
-
319
)
28
Kozako
 
T
Yoshimitsu
 
M
Fujiwara
 
H
, et al. 
PD-1/PD-L1 expression in human T-cell leukemia virus type 1 carriers and adult T-cell leukemia/lymphoma patients.
Leukemia
2009
, vol. 
23
 
2
(pg. 
375
-
382
)
29
Weisenburger
 
DDSK
Savage
 
KJ
Harris
 
NL
, et al. 
International Peripheral T-cell Lymphoma Project
Peripheral T-cell lymphoma, not otherwise specified: a report of 340 cases from the International Peripheral T-cell Lymphoma Project.
Blood
2011
, vol. 
117
 
12
(pg. 
3402
-
3408
)
30
Yoshida
 
N
Miyoshi
 
H
Kato
 
T
, et al. 
CCR4 frameshift mutation identifies a distinct group of adult T cell leukaemia/lymphoma with poor prognosis.
J Pathol
2016
, vol. 
238
 
5
(pg. 
621
-
626
)
31
Mazanet
 
MM
Hughes
 
CCW
B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis.
J Immunol
2002
, vol. 
169
 
7
(pg. 
3581
-
3588
)
32
Keir
 
ME
Liang
 
SC
Guleria
 
I
, et al. 
Tissue expression of PD-L1 mediates peripheral T cell tolerance.
J Exp Med
2006
, vol. 
203
 
4
(pg. 
883
-
895
)
33
Kwon
 
D
Kim
 
S
Kim
 
PJ
, et al. 
Clinicopathological analysis of programmed cell death 1 and programmed cell death ligand 1 expression in the tumour microenvironments of diffuse large B cell lymphomas.
Histopathology
 
2016;68(7):1079-89
34
Xu
 
J
Sun
 
HH
Fletcher
 
CD
, et al. 
Expression of programmed cell death 1 ligands (PD-L1 and PD-L2) in histiocytic and dendritic cell disorders.
Am J Surg Pathol
2016
, vol. 
40
 
4
(pg. 
443
-
453
)
35
Droeser
 
RA
Hirt
 
C
Viehl
 
CT
, et al. 
Clinical impact of programmed cell death ligand 1 expression in colorectal cancer.
Eur J Cancer
2013
, vol. 
49
 
9
(pg. 
2233
-
2242
)
36
Taube
 
JM
Anders
 
RA
Young
 
GD
, 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
, vol. 
4
 
127
pg. 
127ra37
 
37
Schmidt
 
LH
Kümmel
 
A
Görlich
 
D
, et al. 
PD-1 and PD-L1 Expression in NSCLC Indicate a Favorable Prognosis in Defined Subgroups.
PLoS One
2015
, vol. 
10
 
8
pg. 
e0136023
 
38
Spranger
 
S
Spaapen
 
RM
Zha
 
Y
, et al. 
Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells.
Sci Transl Med
2013
, vol. 
5
 
200
pg. 
200ra116
 
39
Kim
 
JR
Moon
 
YJ
Kwon
 
KS
, et al. 
Tumor infiltrating PD1-positive lymphocytes and the expression of PD-L1 predict poor prognosis of soft tissue sarcomas.
PLoS One
2013
, vol. 
8
 
12
pg. 
e82870
 
40
Taube
 
JM
Klein
 
A
Brahmer
 
JR
, et al. 
Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy.
Clin Cancer Res
2014
, vol. 
20
 
19
(pg. 
5064
-
5074
)
41
Thompson
 
RH
Dong
 
H
Lohse
 
CM
, et al. 
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma.
Clin Cancer Res
2007
, vol. 
13
 
6
(pg. 
1757
-
1761
)
42
Carreras
 
J
Lopez-Guillermo
 
A
Roncador
 
G
, et al. 
High numbers of tumor-infiltrating programmed cell death 1-positive regulatory lymphocytes are associated with improved overall survival in follicular lymphoma.
J Clin Oncol
2009
, vol. 
27
 
9
(pg. 
1470
-
1476
)
43
Wahlin
 
BE
Aggarwal
 
M
Montes-Moreno
 
S
, et al. 
A unifying microenvironment model in follicular lymphoma: outcome is predicted by programmed death-1--positive, regulatory, cytotoxic, and helper T cells and macrophages.
Clin Cancer Res
2010
, vol. 
16
 
2
(pg. 
637
-
650
)
44
Camicia
 
R
Winkler
 
HC
Hassa
 
PO
Novel drug targets for personalized precision medicine in relapsed/refractory diffuse large B-cell lymphoma: a comprehensive review.
Mol Cancer
2015
, vol. 
14
 
1
pg. 
207
 
Sign in via your Institution