Skip to main content
Erschienen in: BMC Cancer 1/2020

Open Access 01.12.2020 | Research article

PD-L1 and CD4 are independent prognostic factors for overall survival in endometrial carcinomas

verfasst von: Shuang Zhang, Takeo Minaguchi, Chenyang Xu, Nan Qi, Hiroya Itagaki, Ayumi Shikama, Nobutaka Tasaka, Azusa Akiyama, Manabu Sakurai, Hiroyuki Ochi, Toyomi Satoh

Erschienen in: BMC Cancer | Ausgabe 1/2020

Abstract

Background

Tumor microenvironment (TME) including the immune checkpoint system impacts prognosis in some types of malignancy. The aim of our study was to investigate the precise prognostic significance of the TME profile in endometrial carcinoma.

Methods

We performed immunohistochemistry of the TME proteins, PD-L1, PD-1, CD4, CD8, CD68, and VEGF in endometrial carcinomas from 221 patients.

Results

High PD-L1 in tumor cells (TCs) was associated with better OS (p = 0.004), whereas high PD-L1 in tumor-infiltrating immune cells (TICs) was associated with worse OS (p = 0.02). High PD-L1 in TICs correlated with high densities of CD8+ TICs and CD68+ TICs, as well as microsatellite instability (p = 0.00000064, 0.00078, and 0.0056), while high PD-L1 in TCs correlated with longer treatment-free interval (TFI) after primary chemotherapy in recurrent cases (p = 0.000043). High density of CD4+ TICs correlated with better OS and longer TFI (p = 0.0008 and 0.014). Univariate and multivariate analyses of prognostic factors revealed that high PD-L1 in TCs and high density of CD4+ TICs were significant and independent for favorable OS (p = 0.014 and 0.0025).

Conclusion

The current findings indicate that PD-L1 and CD4+ helper T cells may be reasonable targets for improving survival through manipulating chemosensitivity, providing significant implications for combining immunotherapies into the therapeutic strategy for endometrial carcinoma.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
FIGO
International Federation of Gynecology and Obstetrics
IHC
Immunohistochemistry
LVI
Lymphovascular space invasion
MSI
Microsatellite instability
MSS
Microsatellite stable
OS
Overall survival
PD-L1
Programmed cell death-ligand 1
PD-1
Programmed cell death-1
PFS
Progression-free survival
TAM
Tumor-associated macrophage
TCs
Tumor cells
TFI
Treatment-free interval
TICs
Tumor-infiltrating immune cells
TILs
Tumor infiltrating lymphocytes
TME
Tumor microenvironment

Background

Endometrial cancer is the most common malignancy of female reproductive organs in developed countries, and the incidence is recently increasing [1]. Primary treatment comprises surgery in combination with adjuvant chemotherapy and/or radiotherapy based on the risk stratification for recurrence. The majority of cases are diagnosed at an early stage, and the 5-year survival rate for those with localized disease is 95% [2]. Yet 15–20% of these tumors recur after primary treatment [3]. The 5-year survival rate for those with advanced/recurrent measurable disease is < 10%, and the efficacy of second-line chemotherapy after primary regimens with taxane plus platinum is not more than 15% [4]. Thus, development of novel treatment strategy for those diseases is urgently required.
Programmed cell death-1 (PD-1), immune inhibiting receptor, is expressed on the surface of activated T cells and B cells, and the PD-1 pathway plays critical roles in maintaining immunological self-tolerance [5]. There are two ligands for this receptor, programmed cell death-ligand 1 (PD-L1) and PD-L2. PD-L2 is expressed on activated dendritic cells and macrophages predominantly as well as on tumor cells and B cells, while PD-L1 is expressed on many cell types including immune cells and tumor cells [6]. Tumor cells escape host antitumor immune response through the PD-1/PD-L1 pathway. Recently, therapeutics targeting this immune checkpoint system have shown unprecedented durable clinical responses in various kinds of tumor [7].
A study by Teng et al. on advanced malignant melanomas showed that tumor microenvironment (TME) can be classified based on tumor infiltrating lymphocytes (TILs) and PD-L1 expression: PD-L1+ TIL+ group of tumors favorably responded to immune checkpoint blockade [8]. Another study on melanomas by Tumeh et al. showed that pre-existing CD8+ T cells located at the invasive tumor margin were associated with the expression of PD-1/PD-L1 immune inhibitory system and may predict response to anti-PD-L1 therapy [7]. Regarding ovarian cancer, a study by Webb et al. on high-grade serous ovarian cancer showed that PD-L1 expressed by tumor-associated macrophages (TAM) was significantly associated with favorable disease-specific survival after anti-PD-1 antibody therapy [9]. Darb-Esfahani et al. have shown that PD-1/PD-L1 expressions in high-grade serous ovarian cancer were significantly associated with favorable progression-free survival (PFS) and overall survival (OS) [10]. Another study on ovarian cancer by Hamanishi et al. has shown that high PD-L1 expression on tumor cells and low CD8+ T lymphocyte count are independent prognostic factors for poor PFS and OS [11]. Colorectal cancers with microsatellite instability (MSI) were reported to lead to higher mutation burden, with a greater density of CD8+ lymphocytes, and to benefit more from pembrolizumab, a kind of anti-PD-1 antibody [12]. Frequency of MSI in endometrial cancer is reportedly 22–33%, higher than cervical (8%) and ovarian (10%) cancers, being highest among gynecologic malignancies [13]. As regards endometrial cancer, the significance of the PD-1/PD-L1 pathway has just begun to be investigated including a number of ongoing clinical trials [14].
There exist varieties of factors in the TME of endometrial carcinoma. The purpose of the current study is to find out the relationships between the TME profile including PD-1/PD-L1 expressions and clinicopathologic features, and to identify predictive biomarkers for the outcome by treatments. Our findings provide significant implications for formulating novel therapeutic strategy for the disease.

Methods

Patients and specimens

All patients diagnosed with endometrial carcinoma, who received surgery in the Department of Obstetrics and Gynecology at the University of Tsukuba Hospital between 1999 and 2009, were identified through our database. A total of consecutive 221 patients were included in the present study, and their medical records were retrospectively reviewed. All samples were obtained with opt-out procedure in accordance with the study protocol approved by the Ethics Committee University of Tsukuba Hospital. The study was performed in accordance with the Declaration of Helsinki. A median follow-up duration was 132 months (range, 3–209 months). Follow-up data were retrieved until 2018-7-20. Staging was performed based on the criteria of International Federation of Gynecology and Obstetrics (FIGO, 2008). Endometrioid carcinomas were subclassified into three grades (G1, G2, and G3) according to the FIGO criteria. Treatment of patients was described previously [15]. Table 1 summarizes the patient demographics.
Table 1
Patient demographics
Characteristic
Number (n = 221)
%
Median age (range)
57 (26–84)
FIGO stage
 I
144
 
65
  IA
 
110
50
  IB
 
34
15
 II
17
 
8
 III
36
 
16
  IIIA
 
13
6
  IIIC
 
23
10
 IV
24
 
11
  IVA
 
2
1
  IVB
 
22
10
Histotype
 Endometrioid
196
 
89
  G1
 
115
52
  G2
 
56
25
  G3
 
25
11
 Serous
12
 
5
 Adenosquamous
4
 
2
 Clear cell
4
 
2
 Poorly differentiated
1
 
0
 Undifferentiated
1
 
0
 Mixed epithelial
3
 
1
Myometrial invasion> 1/2
81
 
37
Lymphovascular space invasion
84
 
38
Primary treatment
   
 Surgery
221
 
100
  Lymphadenectomy
 
171
77
  Lymphnode sampling
 
21
10
  Lymphnode not removed
 
29
13
 Adjuvant chemotherapy
60
 
27
  TC
 
55
25
  CAP
 
4
2
 Adjuvant radiotherapy
58
 
26
Abbreviations: FIGO International Federation of Gynecology and Obstetrics, TC paclitaxel and carboplatin combination, CAP cyclophosphamide, doxorubicin, and cisplatin combination

Immunohistochemistry

Immunohistochemical (IHC) procedures were conducted as described previously [15]. Antibodies used are PD-L1 (SP142, rabbit monoclonal, Spring Bioscience, Pleasanton, CA, USA), PD-1 (NAT105, mouse monoclonal, GeneTex, Irvine, CA, USA), CD4 (clone SP35, rabbit monoclonal, Spring Bioscience, Pleasanton, CA, USA), CD8 (clone C8/144B, mouse monoclonal, Nichirei Biosciences, Tokyo, Japan), CD68 (PG-M1, mouse monoclonal, DAKO, Tokyo, Japan), and VEGF (A-20, rabbit polyclonal, Santa Cruz, Dallas, TX, USA). For PD-L1 staining, antigen retrieval was done by autoclaving at 121 °C for 10 min in Tris/EDTA buffer (pH 9.0), and 1st antibody incubation (1:100) was conducted at 4 °C overnight. The corresponding normal endometria or stroma provided an internal positive control, and negative controls without addition of primary antibody showed low background staining.

IHC scoring

Blinded for clinical and pathologic parameters, immunoreaction was assigned by two investigators (SZ and TM), and any discrepancies were resolved by conferring over a multiviewer microscope. For semiquantitative analyses for PD-L1 and VEGF, the IHC staining was scored by multiplying the percentages of positive tumor cells (PP: 0, no positive cell; 1, < 10%; 2, 10–50%; and 3, > 50% positive tumor cells) by their prevalent degree of staining (SI: 0, no staining; 1, weak; 2, moderate; and 3, strong). The IHC scores (IHS=PP × SI) range from 0 to 9. For PD-L1, we evaluated membrane staining of tumor cells (TCs) and tumor-infiltrating immune cells (TICs) separately. For CD4, CD8, CD68, and PD-1, we counted positive TICs by magnification of × 200 in most abundant 3 locations of the slide and calculated the average. The representative images for immunostaining are shown in Fig. 1.

MSI analysis

MSI status was analyzed with the five fluorescence-labeled microsatellite markers, BAT25, BAT26, D2S123, D5S346 and D17S250 [16]. Tumors showing allelic shift at one or more markers were classified as MSI, and tumors with no allelic shift at any marker as microsatellite stable (MSS).

Statistical analyses

Differences in proportions were evaluated by the Fisher’s exact test. Differences in continuous variables were evaluated by the Mann-Whitney U test. The optimal cut-off values of IHC scores for the relationship with OS were determined by the K-Adaptive partitioning method (Table 2) [17]. Kaplan–Meier survival curves were calculated and compared statistically using the log-rank test. The Cox proportional hazard model was used for the univariate and multivariate analyses. OS was measured from the start of primary treatment to death from any cause. Treatment-free interval (TFI) was measured from the end of primary adjuvant chemotherapy to the diagnosis of recurrence. Statistical analyses were performed using R version 3.5.3.
Table 2
Optimal cut-off values of IHC scores for the relationship with OS
 
Mean ± SD
Min
Max
Cut-off
Category
N (%)
PD-L1 in TCs
1.36 ± 1.47
0
9
0<
High expression
155 (70)
Low expression
66 (30)
PD-L1 in TICs
2.70 ± 1.94
0
9
4<
High expression
36 (16)
Low expression
185 (84)
PD-1+ TICs
16.94 ± 23.55
0
129.33
6.67<
High density
131 (59)
Low density
90 (41)
VEGF in TCs
3.64 ± 1.69
1
9
2<
High expression
151 (68)
Low expression
70 (32)
CD4+ TICs
137.99 ± 65.18
31
391
126.33<
High density
117 (53)
Low density
104 (47)
CD8+ TICs
196.52 ± 121.71
18.33
582.67
296.33<
High density
43 (19)
Low density
178 (81)
CD68+ TICs
161.61 ± 88.00
16.33
527.67
126<
High density
129 (58)
Low density
92 (42)
Abbreviations: IHC immunohistochemical, OS overall survival, SD standard deviation, Min minimum, Max maximum, PD-L1 programmed cell death-ligand 1, TCs tumor cells, TICs tumor-infiltrating immune cells, PD-1 programmed cell death-1, VEGF vascular endothelial growth factor

Results

We performed IHC evaluation of the TME proteins in 221 primary endometrial carcinomas (Table 2). First, we examined mutual relationships among the IHC results. High PD-L1 expression in TCs showed an inverse correlation with high PD-L1 expression in TICs (p = 0.0054; Table 3). High PD-L1 expression in TICs correlated with high density of PD-1+, CD8+, and CD68+ TICs (p = 0.00032, 6.4E-07, and 0.00078; Table 3). High density of PD-1+ TICs correlated with high density of CD8+, and CD68+ TICs (p = 0.0097 and 0.00028; Table 3). High density of CD4+, CD8+, and CD68+ TICs showed mutual correlations (Table 3).
Table 3
Mutual relationships among TME protein expressions
 
PD-L1 in TICs
 
PD-1+ TICs
 
VEGF in TCs
 
CD4+ TICs
 
CD8+ TICs
 
CD68+ TICs
 
 
High expression (n = 36)
Low expression (n = 185)
P-value
High density (n = 131)
Low density (n = 90)
P-value
High expression (n = 151)
Low expression (n = 70)
P-value
High density (n = 117)
Low density (n = 104)
P-value
High density (n = 43)
Low density (n = 178)
P-value
High density (n = 129)
Low density (n = 92)
P-value
High PD-L1 expression in TCs
18 (50%)
137 (74%)
0.0054
88 (67%)
67 (74%)
0.30
102 (68%)
53 (76%)
0.27
86 (74%)
69 (66%)
0.30
33 (77%)
122 (69%)
0.36
88 (68%)
67 (73%)
0.55
High PD-L1 expression in TICs
31 (24%)
5 (6%)
0.00032
29 (19%)
7 (10%)
0.12
24 (21%)
12 (12%)
0.10
19 (44%)
17 (10%)
6.4E-07
30 (23%)
6 (7%)
0.00078
High density of PD-1+ TICs
86 (57%)
45 (64%)
0.38
76 (65%)
55 (53%)
0.076
33 (77%)
98 (55%)
0.0097
90 (70%)
41 (45%)
0.00028
High VEGF expression in TCs
76 (65%)
75 (72%)
0.31
33 (77%)
118 (66%)
0.21
88 (68%)
63 (68%)
1.0
High density of CD4+ TICs
32 (74%)
85 (48%)
0.0020
77 (60%)
40 (43%)
0.020
High density of CD8+ TICs
32 (25%)
11 (12%)
0.024
Abbreviations: TME tumor microenvironment, PD-L1 programmed cell death-ligand 1, TICs tumor-infiltrating immune cells, PD-1 programmed cell death-1, VEGF vascular endothelial growth factor, TCs tumor cells
Secondly, we examined the relationships between the IHC evaluations and clinicopathologic parameters (Table 4). High PD-L1 expression in TCs was associated with G1, non-G3, superficial myometrial invasion, and negative lymphovascular space invasion (LVI) (p = 3.2E-05, 0.00026, 0.0037, and 0.049; Table 4), while high PD-L1 expression in TICs was associated with non-endometrioid histology, non-G1, deep myometrial invasion, positive LVI, and advanced FIGO stage (p = 0.0089, 0.018, 0.0044, 0.00026, and 0.014; Table 4). High density of PD-1+ TICs was associated with non-endometrioid histology, non-G1, positive LVI, and MSI (p = 0.0086, 1.1E-05, 0.0047, and 0.0015; Table 4). High VEGF expression in TCs was associated with deep myometrial invasion, non-stage I, and advanced stage (p = 0.00051, 0.0015, and 0.024; Table 4). High density of CD4+ TICs was significantly associated with endometrioid histology and superficial myometrial invasion (p = 0.033 and 0.00044; Table 4), while high density of CD8+ TICs was associated with MSI (p = 0.012; Table 4). High density of CD68+ TICs showed no significant association with clinicopathologic parameters (Table 4).
Table 4
Relationships between TME protein expressions and clinicopathologic parameters
 
PD-L1 in TCs
 
PD-L1 in TICs
 
PD-1+ TICs
 
VEGF in TCs
 
CD4+ TICs
 
CD8+ TICs
 
CD68+ TICs
 
 
High expression (n = 155)
Low expression (n = 66)
P-value
High expression (n = 36)
Low expression (n = 185)
P-value
High density (n = 131)
Low density (n = 90)
P-value
High expression (n = 151)
Low expression (n = 70)
P-value
High density (n = 117)
Low density (n = 104)
P-value
High density (n = 43)
Low density (n = 178)
P-value
High density (n = 129)
Low density (n = 92)
P-value
Age > 60
57 (37%)
23 (35%)
0.88
12 (33%)
68 (37%)
0.85
51 (39%)
29 (32%)
0.32
56 (37%)
24 (34%)
0.76
35 (30%)
45 (43%)
0.050
11 (26%)
69 (39%)
0.12
52 (40%)
28 (30%)
0.16
Endometrioid (vs. Non-endometrioid)
141 (91%)
55 (83%)
0.11
27 (75%)
169 (91%)
0.0089
110 (84%)
86 (96%)
0.0086
132 (87%)
64 (91%)
0.50
109 (93%)
87 (84%)
0.033
40 (93%)
156 (88%)
0.43
112 (87%)
84 (91%)
0.39
G1
95 (61%)
20 (30%)
3.2E-05
12 (33%)
103 (56%)
0.018
52 (40%)
63 (70%)
1.1E-05
76 (50%)
39 (56%)
0.47
65 (56%)
50 (48%)
0.28
25 (58%)
90 (51%)
0.40
65 (50%)
50 (54%)
0.59
G3
9 (6%)
16 (24%)
0.00026
7 (19%)
18 (10%)
0.14
17 (13%)
8 (9%)
0.39
21 (14%)
4 (6%)
0.11
12 (10%)
13 (13%)
0.67
7 (16%)
18 (10%)
0.28
17 (13%)
8 (9%)
0.39
MI > 1/2
47 (30%)
34 (52%)
0.0037
21 (58%)
60 (32%)
0.0044
50 (38%)
31 (34%)
0.67
67 (44%)
14 (20%)
0.00051
30 (26%)
51 (49%)
0.00044
13 (30%)
68 (38%)
0.38
53 (41%)
28 (30%)
0.12
LVI
52 (34%)
32 (48%)
0.049
24 (67%)
60 (32%)
0.00026
60 (46%)
24 (27%)
0.0047
64 (42%)
20 (29%)
0.054
46 (39%)
38 (37%)
0.68
18 (42%)
66 (37%)
0.60
55 (43%)
29 (32%)
0.12
FIGO stage I
104 (67%)
40 (61%)
0.36
19 (53%)
125 (68%)
0.125
81 (62%)
63 (70%)
0.25
88 (58%)
56 (80%)
0.0015
81 (69%)
63 (61%)
0.20
32 (74%)
112 (63%)
0.21
82 (64%)
62 (67%)
0.57
FIGO stage III-IV
37 (24%)
23 (35%)
0.10
16 (44%)
44 (24%)
0.014
42 (32%)
18 (20%)
0.064
48 (32%)
12 (17%)
0.024
30 (26%)
30 (29%)
0.65
10 (23%)
50 (28%)
0.57
40 (31%)
20 (22%)
0.17
MSI
29 (19%)
19 (29%)
0.11
12 (33%)
36 (19%)
0.078
38 (29%)
10 (11%)
0.0015
36 (24%)
12 (17%)
0.30
29 (25%)
19 (18%)
0.26
16 (37%)
32 (18%)
0.012
31 (24%)
17 (18%)
0.41
Abbreviations: TME tumor microenvironment, PD-L1 programmed cell death-ligand 1, TCs tumor cells, TICs tumor-infiltrating immune cells, PD-1 programmed cell death-1, VEGF vascular endothelial growth factor, MI myometrial invasion, LVI lymphovascular space invasion, FIGO International Federation of Gynecology and Obstetrics, MSI microsatellite instability
Thirdly, the patient OS was compared according to the IHC evaluations. Patients with TCs expressing high PD-L1 showed better OS than those with low PD-L1 expression (p = 0.004; Fig. 2a), while conversely patients with TICs expressing high PD-L1 showed worse OS than those with low PD-L1 expression (p = 0.02; Fig. 2b). High densities of CD4+ TICs and CD8+ TICs both correlated with better OS (p = 0.0008 and 0.04; Fig. 2e and f). As for PD-1+ TICs, VEGF in TCs, and CD68+ TICs, the OS showed no significant difference (p = 0.1, 0.06, and 0.2; Fig. 2c, d, and g). The OS according to MSI/MSS showed no difference (p = 0.9; Fig. 2h).
Next, the associations between TFI after primary adjuvant chemotherapy and the TME protein expressions were examined. High PD-L1 expression in TCs and high density of CD4+ TICs were both associated with longer TFI (p = 0.000043 and 0.014; Fig. 3a). We further examined the relationships between MSI status and the TME protein expressions. High PD-L1 expression in TICs and high densities of PD-1+ TICs and CD8+ TICs were associated with MSI (p = 0.0056, 0.00040, and 0.00086; Fig. 3b).
Lastly, we conducted univariate and multivariate analyses of prognostic factors for OS. In the univariate analysis, high PD-L1 expression in TICs, older age (> 60), advanced FIGO stage, non-endometrioid histology, deep myometrial invasion (> 1/2), and positive LVI were found to be significant for worse OS (p = 0.023, 0.0017, 2.0E-09, 1.9E-07, 6.4E-06, and 0.00011; Table 5), while high PD-L1 expression in TCs and high density of CD4+ TICs were significant for better OS (p = 0.0050 and 0.0015; Table 5). Subsequent multivariate analysis revealed that high PD-L1 expression in TCs, high density of CD4+ TICs, advanced stage, non-endometrioid histology, and positive LVI were significant and independent for OS (p = 0.014, 0.0025, 0.000042, 0.0031, and 0.028; Table 5).
Table 5
Univariate and multivariate analyses of prognostic factors for OS
 
Univariate
Multivariate
HR
95% CI
P-value
HR
95% CI
P-value
High PD-L1 expression in TCs
0.40
0.21–0.76
0.0050
0.43
0.22–0.85
0.014
High PD-L1 expression in TICs
2.25
1.12–4.54
0.023
0.76
0.31–1.82
0.53
High density of PD-1+ TICs
1.71
0.85–3.46
0.13
High VEGF expression in TCs
2.14
0.94–4.85
0.07
High density of CD4+ TICs
0.32
0.16–0.65
0.0015
0.31
0.15–0.67
0.0025
High density of CD8+ TICs
0.31
0.096–1.01
0.053
High density of CD68+ TICs
1.58
0.79–3.12
0.19
Age > 60
2.81
1.47–5.36
0.0017
1.36
0.68–2.72
0.39
FIGO stage III/IV (vs. I/II)
8.62
4.26–17.4
2.0E-09
5.50
2.43–12.5
0.000042
Non-endometrioid (vs. Endometrioid)
5.78
2.99–11.2
1.9E-07
3.31
1.50–7.32
0.0031
MI > 1/2
5.04
2.50–10.2
6.4E-06
1.46
0.66–3.19
0.35
LVI (+)
3.75
1.92–7.34
0.00011
2.27
1.09–4.73
0.028
Abbreviations: OS overall survival, HR hazard ratio, CI confidence interval, PD-L1 programmed cell death-ligand 1, TCs tumor cells, TICs tumor-infiltrating immune cells, PD-1 programmed cell death-1, VEGF vascular endothelial growth factor, FIGO International Federation of Gynecology and Obstetrics, MI myometrial invasion, LVI lymphovascular space invasion

Discussion

Our survival analyses exhibited that high PD-L1 expression in TCs was associated with better OS, while conversely high PD-L1 expression in TICs was associated with worse OS (Fig. 2a, b, Table 5). Besides, high PD-L1 expression in TICs showed an inverse correlation with high PD-L1 expression in TCs (Table 3). These findings indicate that PD-L1 expression in TCs and that in TICs seem contrary to each other. PD-L1 expressed on the surface of TCs is supposed to bind to PD-1 receptor on immune cells and to induce adaptive immune resistance. Our above observations may be explicable if some proportion of expressed PD-L1 could move between the surface of TCs and the surface of TICs so that the PD-L1 bound to PD-1 on the surface of TICs may induce adaptive immune resistance leading to poor survival, while the PD-L1 remaining on the surface of TCs may not. This hypothesis may be supported by the published findings that, in addition to tissue PD-L1, there also exist circulating PD-L1 such as exosomal PD-L1 [18, 19] and soluble PD-L1 [20, 21]. However, further molecular and clinical investigations are essential to verify our observation and to elucidate the mechanism underlying them.
High PD-L1 expression in TICs was associated with MSI (Fig. 3b), and with high density of CD8+ TICs and CD68+ TICs (Table 3), suggesting that PD-L1-induced adaptive immune resistance may involve MSI, killer T cells, and TAMs, as CD8 and CD68 are markers for killer T cells and TAMs, respectively. MSI is known to cause hypermutation leading to increased burden of tumor antigens, which induces increased immune response [13]. Increased immune response may induce PD-1/PD-L1-mediated adaptive immune resistance, which will lead to aggressive tumor phenotype and poor prognosis. Indeed, our analyses of the relationships between the TME protein expressions and clinicopathologic parameters exhibited that high PD-L1 expression in TICs was associated with non-endometrioid histology, non-G1, deep myometrial invasion, positive LVI, and advanced FIGO stage (Table 4), and our survival analysis demonstrated that high PD-L1 expression in TICs was associated with unfavorable OS (Fig. 2b). Taken together, these findings suggest that PD-L1 expression of TICs may be a biomarker for the T cell-inflamed tumor phenotype [22]. Clinical response to anti-PD-1 monoclonal antibody was reported to occur almost exclusively in patients with pre-existing T cell infiltrates in the region of PD-L1 upregulation [7, 23]. Following anti-PD-1 administration, these CD8+ T cells seemed to proliferate and expand to penetrate throughout the tumor, which correlated with tumor regression [7]. Altogether, our findings implicate that anti-PD-1/PD-L1 therapy may improve the unfavorable survival of the subset of endometrial cancers with TICs expressing high PD-L1.
Moreover, in the analysis of the associations between the TME protein expressions and TFI after primary adjuvant chemotherapy, high PD-L1 expression in TCs indicated a longer TFI (Fig. 3a), suggesting that prognostic impact of PD-L1 expression may be mediated by affected chemosensitivity, as TFI reportedly correlates with response to chemotherapy for recurrence and/or survival after recurrence in endometrial cancer [2426]. This hypothesis may be supported by the published findings where upregulation of the PD-1/PD-L1 axis confers chemoresistance in some types of tumor [2729]. Accordingly, our findings further suggest that anti-PD-1/PD-L1 therapy may attenuate chemoresistance in the patients with TICs expressing high PD-L1.
In the univariate and multivariate analyses of prognostic factors, besides high PD-L1 expression in TCs, high density of CD4+ TICs was found to be significant and independent for favorable OS (Table 5), being consistent with previous publications where high infiltration of CD4+ TILs was reported to be a favorable prognostic factor for some types of malignancy [3032]. Besides, high density of CD4+ TICs was found to be associated with longer TFI (Fig. 3a), suggesting that helper T cells also may affect prognosis through involving chemosensitivity. The proliferation and differentiation into regulatory T cells of CD4+ T cells is reported to be manipulated by retinoic acid [33], STAT3 silencing [34], and DNGR-1 targeting [35], raising their therapeutic possibility. Further basic and clinical studies are warranted to verify our proposal.
The KEYNOTE-028 phase I study evaluated the safety and efficacy of pembrolizumab, an anti-PD-1 monoclonal antibody, in patients with PD-L1-positive advanced solid tumors [36]. Pembrolizumab demonstrated a favorable safety profile and durable antitumor activity in a subgroup of patients with heavily pretreated advanced PD-L1-positive endometrial cancer [36]. Currently, many phase II/III clinical trials of anti-PD-1/PD-L1 therapy in endometrial cancers are ongoing. Our above findings indicate that anti-PD1/PD-L1 therapies combined with conventional chemotherapeutics may be beneficial for the patients with poor prognosis due to high PD-L1 expression in TICs through improving chemosensitivity.
There exist only few reports on prognostic significances of the TME proteins in endometrial cancer so far. Regarding PD-L1 expression and survival, Kim et al. have recently reported on 183 primary endometrial cancers that high PD-L1 expression on immune cells was an independent prognostic factor for poor PFS [37]. Ikeda et al. have also reported on 32 endometrioid endometrial cancers that the cases with high PD-L1 mRNA expression in cancer tissues showed significantly longer PFS [38]. Yamashita et al. have recently reported on 149 endometrioid endometrial cancers that high PD-L1 expression in tumor cells was significantly associated with better PFS [39]. These findings are in line with our results that high PD-L1 expression in TCs was associated with better OS (Fig. 2a), while high PD-L1 on TICs was associated with worse OS (Fig. 2b). As for CD8 expression and survival, Yamashita et al. have reported that CD8+ TILs was significantly associated with better PFS [39]. Ikeda et al. also reported that high CD8 mRNA expression in tumor tissues was significantly associated with longer PFS [38]. These findings are consistent with our result that high density of CD8+ TICs correlated with better OS (Fig. 2f). Bellone et al. have recently reported on 131 endometrial cancers that POLE-mutated tumors were associated with improved PFS and displayed increased numbers of CD4+ and CD8+ TILs as compared to wild-type POLE tumors, and that PD-1 was overexpressed in TILs from POLE-mutated vs. wild-type-tumors [40]. In our study, MSI was associated with high PD-L1 expression in TICs (Fig. 3b), which was significantly associated with worse OS (Fig. 2b). POLE-mutated endometrial cancers have been reported to be MSS in a couple of studies including this article [4042]. Therefore, it is plausible that POLE-mutated tumors and MSI tumors may have the opposite prognostic features. As regards the relationship between PD-L1 expression and clinicopathologic features, Mo et al. reported on 75 endometrial cancers that PD-L1 expression in TICs was more frequently found in the moderately and poorly-differentiated tumors and type II than in the type I tumors [43], being in line with our finding that high PD-L1 expression in TICs was associated with non-endometrioid histology and non-G1 (Table 4). Further studies are warranted to clarify the clinical and prognostic significance of the TME status in endometrial cancer.
The present study still contains some limitations. The retrospective study design potentially causes selection biases. The number of studied samples is relatively small. The evaluation method for the TME protein expression is mainly based on semi-quantitative analyses. Nevertheless, the treatment strategy was almost consistent throughout the study period, and most importantly the follow-up duration was much longer than the former studies (median, 132 vs. 30.3–38 months [37, 39]), supporting the validity of our survival data.

Conclusions

We have demonstrated here that high PD-L1 in TCs was associated with better OS, while high PD-L1 in TICs was associated with worse OS. High PD-L1 in TICs exhibited associations with high densities of CD8+ TILs and CD68+ TAMs, and MSI, while high PD-L1 in TCs correlated with longer TFI. High density of CD4+ TICs correlated with better OS and longer TFI. Univariate and multivariate analyses exhibited that high PD-L1 in TCs and high density of CD4+ TICs were significant and independent prognostic factors for favorable OS. The current findings indicate that PD-L1 and CD4+ helper T cells may be reasonable targets for improving survival via enhancing chemosensitivity, providing useful information for combining immunotherapies into the therapeutic strategy for endometrial carcinoma.

Acknowledgements

Not applicable.
The study protocol was approved by the Ethics Committee University of Tsukuba Hospital (H26–118). The committee waived the requirement for informed consent because of the opt-out approach in accordance with national regulations.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34.CrossRef
3.
Zurück zum Zitat Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012;13(8):e353–61.CrossRef Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012;13(8):e353–61.CrossRef
4.
Zurück zum Zitat Moxley KM, McMeekin DS. Endometrial carcinoma: a review of chemotherapy, drug resistance, and the search for new agents. Oncologist. 2010;15(10):1026–33.CrossRef Moxley KM, McMeekin DS. Endometrial carcinoma: a review of chemotherapy, drug resistance, and the search for new agents. Oncologist. 2010;15(10):1026–33.CrossRef
5.
Zurück zum Zitat Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19(7):813–24.CrossRef Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19(7):813–24.CrossRef
6.
Zurück zum Zitat Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 2018;11(1):31.CrossRef Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 2018;11(1):31.CrossRef
7.
Zurück zum Zitat Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.CrossRef Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.CrossRef
8.
Zurück zum Zitat Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res. 2015;75(11):2139–45.CrossRef Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res. 2015;75(11):2139–45.CrossRef
9.
Zurück zum Zitat Webb JR, Milne K, Kroeger DR, Nelson BH. PD-L1 expression is associated with tumor-infiltrating T cells and favorable prognosis in high-grade serous ovarian cancer. Gynecol Oncol. 2016;141(2):293–302.CrossRef Webb JR, Milne K, Kroeger DR, Nelson BH. PD-L1 expression is associated with tumor-infiltrating T cells and favorable prognosis in high-grade serous ovarian cancer. Gynecol Oncol. 2016;141(2):293–302.CrossRef
10.
Zurück zum Zitat Darb-Esfahani S, Kunze CA, Kulbe H, Sehouli J, Wienert S, Lindner J, Budczies J, Bockmayr M, Dietel M, Denkert C, et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget. 2016;7(2):1486–99.CrossRef Darb-Esfahani S, Kunze CA, Kulbe H, Sehouli J, Wienert S, Lindner J, Budczies J, Bockmayr M, Dietel M, Denkert C, et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget. 2016;7(2):1486–99.CrossRef
11.
Zurück zum Zitat Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, 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(9):3360–5.CrossRef Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, 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(9):3360–5.CrossRef
12.
Zurück zum Zitat Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.CrossRef Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.CrossRef
13.
Zurück zum Zitat Dudley JC, Lin MT, Le DT, Eshleman JR. Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res. 2016;22(4):813–20.CrossRef Dudley JC, Lin MT, Le DT, Eshleman JR. Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res. 2016;22(4):813–20.CrossRef
14.
Zurück zum Zitat Di Tucci C, Capone C, Galati G, Iacobelli V, Schiavi MC, Di Donato V, Muzii L, Panici PB. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol. 2019;30(3):e46.CrossRef Di Tucci C, Capone C, Galati G, Iacobelli V, Schiavi MC, Di Donato V, Muzii L, Panici PB. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol. 2019;30(3):e46.CrossRef
15.
Zurück zum Zitat Abe A, Minaguchi T, Ochi H, Onuki M, Okada S, Matsumoto K, Satoh T, Oki A, Yoshikawa H. PIK3CA overexpression is a possible prognostic factor for favorable survival in ovarian clear cell carcinoma. Hum Pathol. 2013;44(2):199–207.CrossRef Abe A, Minaguchi T, Ochi H, Onuki M, Okada S, Matsumoto K, Satoh T, Oki A, Yoshikawa H. PIK3CA overexpression is a possible prognostic factor for favorable survival in ovarian clear cell carcinoma. Hum Pathol. 2013;44(2):199–207.CrossRef
16.
Zurück zum Zitat Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN. A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. In: AACR. 1998. Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN. A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. In: AACR. 1998.
17.
Zurück zum Zitat Eo S-H, Hong S-M, Cho H. K-adaptive partitioning for survival data: the Kaps add-on package for R. arXiv preprint arXiv. 2013;13064615. Eo S-H, Hong S-M, Cho H. K-adaptive partitioning for survival data: the Kaps add-on package for R. arXiv preprint arXiv. 2013;13064615.
18.
Zurück zum Zitat Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28(8):862–4.CrossRef Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28(8):862–4.CrossRef
19.
Zurück zum Zitat Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, Whiteside TL. Clinical significance of PD-L1(+) Exosomes in plasma of head and neck Cancer patients. Clin Cancer Res. 2018;24(4):896–905.CrossRef Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE, Whiteside TL. Clinical significance of PD-L1(+) Exosomes in plasma of head and neck Cancer patients. Clin Cancer Res. 2018;24(4):896–905.CrossRef
20.
Zurück zum Zitat Okuma Y, Hosomi Y, Nakahara Y, Watanabe K, Sagawa Y, Homma S. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer. 2017;104:1–6.CrossRef Okuma Y, Hosomi Y, Nakahara Y, Watanabe K, Sagawa Y, Homma S. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer. 2017;104:1–6.CrossRef
21.
Zurück zum Zitat Okuma Y, Wakui H, Utsumi H, Sagawa Y, Hosomi Y, Kuwano K, Homma S. Soluble programmed cell death ligand 1 as a novel biomarker for Nivolumab therapy for non-small-cell lung Cancer. Clin Lung Cancer. 2018;19(5):410–7 e411.CrossRef Okuma Y, Wakui H, Utsumi H, Sagawa Y, Hosomi Y, Kuwano K, Homma S. Soluble programmed cell death ligand 1 as a novel biomarker for Nivolumab therapy for non-small-cell lung Cancer. Clin Lung Cancer. 2018;19(5):410–7 e411.CrossRef
22.
Zurück zum Zitat Gajewski TF. The next hurdle in Cancer immunotherapy: overcoming the non-T-cell-inflamed tumor microenvironment. Semin Oncol. 2015;42(4):663–71.CrossRef Gajewski TF. The next hurdle in Cancer immunotherapy: overcoming the non-T-cell-inflamed tumor microenvironment. Semin Oncol. 2015;42(4):663–71.CrossRef
23.
Zurück zum Zitat Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–54.CrossRef Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–54.CrossRef
24.
Zurück zum Zitat Shimamoto K, Saito T, Okadome M, Shimokawa M. Prognostic significance of the treatment-free interval in patients with recurrent endometrial cancer. Eur J Obstet Gynecol Reprod Biol. 2014;175:92–6.CrossRef Shimamoto K, Saito T, Okadome M, Shimokawa M. Prognostic significance of the treatment-free interval in patients with recurrent endometrial cancer. Eur J Obstet Gynecol Reprod Biol. 2014;175:92–6.CrossRef
25.
Zurück zum Zitat Miyake T, Ueda Y, Egawa-Takata T, Matsuzaki S, Yokoyama T, Miyoshi Y, Kimura T, Yoshino K, Fujita M, Yamasaki M. Recurrent endometrial carcinoma: prognosis for patients with recurrence within 6 to 12 months is worse relative to those relapsing at 12 months or later. Am J Obstet Gynecol. 2011;204(6):535. e531–5.CrossRef Miyake T, Ueda Y, Egawa-Takata T, Matsuzaki S, Yokoyama T, Miyoshi Y, Kimura T, Yoshino K, Fujita M, Yamasaki M. Recurrent endometrial carcinoma: prognosis for patients with recurrence within 6 to 12 months is worse relative to those relapsing at 12 months or later. Am J Obstet Gynecol. 2011;204(6):535. e531–5.CrossRef
26.
Zurück zum Zitat Moore KN, Tian C, McMeekin DS, Thigpen JT, Randall ME, Gallion HH. Does the progression-free interval after primary chemotherapy predict survival after salvage chemotherapy in advanced and recurrent endometrial cancer? A gynecologic oncology group ancillary data analysis. Cancer. 2010;116(23):5407–14.CrossRef Moore KN, Tian C, McMeekin DS, Thigpen JT, Randall ME, Gallion HH. Does the progression-free interval after primary chemotherapy predict survival after salvage chemotherapy in advanced and recurrent endometrial cancer? A gynecologic oncology group ancillary data analysis. Cancer. 2010;116(23):5407–14.CrossRef
27.
Zurück zum Zitat Black M, Barsoum IB, Truesdell P, Cotechini T, Macdonald-Goodfellow SK, Petroff M, Siemens DR, Koti M, Craig AW, Graham CH. Activation of the PD-1/PD-L1 immune checkpoint confers tumor cell chemoresistance associated with increased metastasis. Oncotarget. 2016;7(9):10557–67.CrossRef Black M, Barsoum IB, Truesdell P, Cotechini T, Macdonald-Goodfellow SK, Petroff M, Siemens DR, Koti M, Craig AW, Graham CH. Activation of the PD-1/PD-L1 immune checkpoint confers tumor cell chemoresistance associated with increased metastasis. Oncotarget. 2016;7(9):10557–67.CrossRef
28.
Zurück zum Zitat Xu S, Tao Z, Hai B, Liang H, Shi Y, Wang T, Song W, Chen Y, OuYang J, Chen J, et al. miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun. 2016;7:11406.CrossRef Xu S, Tao Z, Hai B, Liang H, Shi Y, Wang T, Song W, Chen Y, OuYang J, Chen J, et al. miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun. 2016;7:11406.CrossRef
29.
Zurück zum Zitat Zhang P, Ma Y, Lv C, Huang M, Li M, Dong B, Liu X, An G, Zhang W, Zhang J, et al. Upregulation of programmed cell death ligand 1 promotes resistance response in non-small-cell lung cancer patients treated with neo-adjuvant chemotherapy. Cancer Sci. 2016;107(11):1563–71.CrossRef Zhang P, Ma Y, Lv C, Huang M, Li M, Dong B, Liu X, An G, Zhang W, Zhang J, et al. Upregulation of programmed cell death ligand 1 promotes resistance response in non-small-cell lung cancer patients treated with neo-adjuvant chemotherapy. Cancer Sci. 2016;107(11):1563–71.CrossRef
30.
Zurück zum Zitat Chen K, Zhu Z, Zhang N, Cheng G, Zhang F, Jin J, Wu J, Ying L, Mao W, Su D. Tumor-infiltrating CD4+ lymphocytes predict a favorable survival in patients with operable esophageal squamous cell carcinoma. Med Sci Monit. 2017;23:4619–32.CrossRef Chen K, Zhu Z, Zhang N, Cheng G, Zhang F, Jin J, Wu J, Ying L, Mao W, Su D. Tumor-infiltrating CD4+ lymphocytes predict a favorable survival in patients with operable esophageal squamous cell carcinoma. Med Sci Monit. 2017;23:4619–32.CrossRef
31.
Zurück zum Zitat Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, et al. Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck. 2016;38(7):1074–84.CrossRef Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, et al. Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck. 2016;38(7):1074–84.CrossRef
32.
Zurück zum Zitat Nejati R, Goldstein JB, Halperin DM, Wang H, Hejazi N, Rashid A, Katz MH, Lee JE, Fleming JB, Rodriguez-Canales J, et al. Prognostic significance of tumor-infiltrating lymphocytes in patients with pancreatic ductal adenocarcinoma treated with Neoadjuvant chemotherapy. Pancreas. 2017;46(9):1180–7.CrossRef Nejati R, Goldstein JB, Halperin DM, Wang H, Hejazi N, Rashid A, Katz MH, Lee JE, Fleming JB, Rodriguez-Canales J, et al. Prognostic significance of tumor-infiltrating lymphocytes in patients with pancreatic ductal adenocarcinoma treated with Neoadjuvant chemotherapy. Pancreas. 2017;46(9):1180–7.CrossRef
33.
Zurück zum Zitat Brown CC, Noelle RJ. Seeing through the dark: new insights into the immune regulatory functions of vitamin a. Eur J Immunol. 2015;45(5):1287–95.CrossRef Brown CC, Noelle RJ. Seeing through the dark: new insights into the immune regulatory functions of vitamin a. Eur J Immunol. 2015;45(5):1287–95.CrossRef
34.
Zurück zum Zitat Sanseverino I, Purificato C, Varano B, Conti L, Gessani S, Gauzzi MC. STAT3-silenced human dendritic cells have an enhanced ability to prime IFNgamma production by both alphabeta and gammadelta T lymphocytes. Immunobiology. 2014;219(7):503–11.CrossRef Sanseverino I, Purificato C, Varano B, Conti L, Gessani S, Gauzzi MC. STAT3-silenced human dendritic cells have an enhanced ability to prime IFNgamma production by both alphabeta and gammadelta T lymphocytes. Immunobiology. 2014;219(7):503–11.CrossRef
35.
Zurück zum Zitat Joffre OP, Sancho D, Zelenay S, Keller AM, Reis e Sousa C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A. Eur J Immunol. 2010;40(5):1255–65.CrossRef Joffre OP, Sancho D, Zelenay S, Keller AM, Reis e Sousa C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A. Eur J Immunol. 2010;40(5):1255–65.CrossRef
36.
Zurück zum Zitat Ott PA, Bang YJ, Berton-Rigaud D, Elez E, Pishvaian MJ, Rugo HS, Puzanov I, Mehnert JM, Aung KL, Lopez J, et al. Safety and antitumor activity of Pembrolizumab in advanced programmed death ligand 1-positive endometrial Cancer: results from the KEYNOTE-028 study. J Clin Oncol. 2017;35(22):2535–41.CrossRef Ott PA, Bang YJ, Berton-Rigaud D, Elez E, Pishvaian MJ, Rugo HS, Puzanov I, Mehnert JM, Aung KL, Lopez J, et al. Safety and antitumor activity of Pembrolizumab in advanced programmed death ligand 1-positive endometrial Cancer: results from the KEYNOTE-028 study. J Clin Oncol. 2017;35(22):2535–41.CrossRef
37.
Zurück zum Zitat Kim J, Kim S, Lee HS, Yang W, Cho H, Chay DB, Cho SJ, Hong S, Kim JH. Prognostic implication of programmed cell death 1 protein and its ligand expressions in endometrial cancer. Gynecol Oncol. 2018;149(2):381–7.CrossRef Kim J, Kim S, Lee HS, Yang W, Cho H, Chay DB, Cho SJ, Hong S, Kim JH. Prognostic implication of programmed cell death 1 protein and its ligand expressions in endometrial cancer. Gynecol Oncol. 2018;149(2):381–7.CrossRef
38.
Zurück zum Zitat Ikeda Y, Kiyotani K, Yew PY, Sato S, Imai Y, Yamaguchi R, Miyano S, Fujiwara K, Hasegawa K, Nakamura Y. Clinical significance of T cell clonality and expression levels of immune-related genes in endometrial cancer. Oncol Rep. 2017;37(5):2603–10.CrossRef Ikeda Y, Kiyotani K, Yew PY, Sato S, Imai Y, Yamaguchi R, Miyano S, Fujiwara K, Hasegawa K, Nakamura Y. Clinical significance of T cell clonality and expression levels of immune-related genes in endometrial cancer. Oncol Rep. 2017;37(5):2603–10.CrossRef
39.
Zurück zum Zitat Yamashita H, Nakayama K, Ishikawa M, Nakamura K, Ishibashi T, Sanuki K, Ono R, Sasamori H, Minamoto T, Iida K, et al. Microsatellite instability is a biomarker for immune checkpoint inhibitors in endometrial cancer. Oncotarget. 2018;9(5):5652–64.CrossRef Yamashita H, Nakayama K, Ishikawa M, Nakamura K, Ishibashi T, Sanuki K, Ono R, Sasamori H, Minamoto T, Iida K, et al. Microsatellite instability is a biomarker for immune checkpoint inhibitors in endometrial cancer. Oncotarget. 2018;9(5):5652–64.CrossRef
40.
Zurück zum Zitat Bellone S, Bignotti E, Lonardi S, Ferrari F, Centritto F, Masserdotti A, Pettinella F, Black J, Menderes G, Altwerger G, et al. Polymerase epsilon (POLE) ultra-mutation in uterine tumors correlates with T lymphocyte infiltration and increased resistance to platinum-based chemotherapy in vitro. Gynecol Oncol. 2017;144(1):146–52.CrossRef Bellone S, Bignotti E, Lonardi S, Ferrari F, Centritto F, Masserdotti A, Pettinella F, Black J, Menderes G, Altwerger G, et al. Polymerase epsilon (POLE) ultra-mutation in uterine tumors correlates with T lymphocyte infiltration and increased resistance to platinum-based chemotherapy in vitro. Gynecol Oncol. 2017;144(1):146–52.CrossRef
41.
Zurück zum Zitat Konstantinopoulos PA, Matulonis UA. POLE mutations as an alternative pathway for microsatellite instability in endometrial cancer: implications for lynch syndrome testing. Cancer. 2015;121(3):331–4.CrossRef Konstantinopoulos PA, Matulonis UA. POLE mutations as an alternative pathway for microsatellite instability in endometrial cancer: implications for lynch syndrome testing. Cancer. 2015;121(3):331–4.CrossRef
42.
Zurück zum Zitat Billingsley CC, Cohn DE, Mutch DG, Stephens JA, Suarez AA, Goodfellow PJ. Polymerase varepsilon (POLE) mutations in endometrial cancer: clinical outcomes and implications for lynch syndrome testing. Cancer. 2015;121(3):386–94.CrossRef Billingsley CC, Cohn DE, Mutch DG, Stephens JA, Suarez AA, Goodfellow PJ. Polymerase varepsilon (POLE) mutations in endometrial cancer: clinical outcomes and implications for lynch syndrome testing. Cancer. 2015;121(3):386–94.CrossRef
43.
Zurück zum Zitat Mo Z, Liu J, Zhang Q, Chen Z, Mei J, Liu L, Yang S, Li H, Zhou L, You Z. Expression of PD-1, PD-L1 and PD-L2 is associated with differentiation status and histological type of endometrial cancer. Oncol Lett. 2016;12(2):944–50.CrossRef Mo Z, Liu J, Zhang Q, Chen Z, Mei J, Liu L, Yang S, Li H, Zhou L, You Z. Expression of PD-1, PD-L1 and PD-L2 is associated with differentiation status and histological type of endometrial cancer. Oncol Lett. 2016;12(2):944–50.CrossRef
Metadaten
Titel
PD-L1 and CD4 are independent prognostic factors for overall survival in endometrial carcinomas
verfasst von
Shuang Zhang
Takeo Minaguchi
Chenyang Xu
Nan Qi
Hiroya Itagaki
Ayumi Shikama
Nobutaka Tasaka
Azusa Akiyama
Manabu Sakurai
Hiroyuki Ochi
Toyomi Satoh
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2020
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-6545-9

Weitere Artikel der Ausgabe 1/2020

BMC Cancer 1/2020 Zur Ausgabe

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.