Skip to main content
Erschienen in: BMC Cancer 1/2018

Open Access 01.12.2018 | Research article

βcatenin is a marker of poor clinical characteristics and suppressed immune infiltration in testicular germ cell tumors

verfasst von: Michal Chovanec, Zuzana Cierna, Viera Miskovska, Katarina Machalekova, Katarina Kalavska, Katarina Rejlekova, Daniela Svetlovska, Dusan Macak, Stanislav Spanik, Karol Kajo, Pavel Babal, Michal Mego, Jozef Mardiak

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

WNT/βcatenin (WNTβ) pathway is activated in early stages of embryonic development. We aimed to evaluate the significance of βcatenin in germ cell tumors (GCTs) and explore associations with the inflamed environment.

Methods

Surgical specimens from 247 patients were analyzed. Βcatenin expression was detected in the tumor tissue by immunohistochemistry and correlated with clinical characteristics, outcome, PD-L1 expression and systemic immune-inflammation index (SII). The Ingenuity Pathway Analysis (IPA) was used to investigate the immune-cell related effects of βcatenin and PD-L1 encoding genes.

Results

βcatenin was expressed in 86.2% of GCTs. The expression in seminomas was significantly lower compared to all subtypes of non-seminoma (all P <  0.0001). A high expression (weighted histoscore > 150) was associated with primary mediastinal non-seminoma (P = 0.035), intermediate/poor risk disease (P = 0.033) and high tumor markers (P = 0.035). We observed a positive correlation with the PD-L1 in tumor and an inverse correlation with the SII. IPA uncovered relationships of CTNNB (βcatenin) and CD274 (PD-L1) genes and their effects on differentiation, proliferation and activation of lymphocyte subtypes.

Conclusion

Herein, we showed that βcatenin is associated with male adult GCT characteristics as well as supressed immune environment.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12885-018-4929-x) contains supplementary material, which is available to authorized users.
Michal Chovanec and Zuzana Cierna are co-first authors
Abkürzungen
GCT
Germ cell tumor
H&E
Hematoxylin eosin
HS
Histoscore
IGCCCG
International Germ Cell Cancer Collaborative Group
IPA
Ingenuity pathway analysis
L
Lymphocytes
N
Neutrophils
OS
Overall survival
P
Platelets
PFS
Progression free survival
SII
Systemic immune-inflammation index
TIL
Tumor infiltrating lymphocytes

Background

Testicular-germ cell tumors (GCTs) are chemotherapy sensitive malignancies [1, 2]. Little was known regarding immune mechanisms in this disease until recently. The significance of PD-1/PD-L1 pathway as well as the role of systemic-immune inflammation was previously described in our works [3, 4]. Additionally, changes in cytokine signalling was observed by our team in association with poor prognosis of GCTs [5, 6]. The role of immune check-point inhibition with anti-PD-1/anti-PD-L1/anti-CTLA4 agents has been established in numerous malignancies [710]. Data suggest that a T-cell inflamed microenvironment seems to be a predisposing factor for the efficacy of check-point inhibition covering only a subset of patients who benefit greatly from modern immune therapy [11, 12]. An activation of WNT/βcatenin (WNTβ) signalling pathway has been recently suggested as an intrinsic inhibitory mechanism for tumor T-cell infiltration in malignant melanoma pre-clinical model [13]. WNTβ pathway is known to be activated in early stages of embryonic development and it is involved in the regulation of differentiation of pluripotent cells [14, 15]. It has also been associated with carcinogenesis [16], cell proliferation and migration and the process of epithelial-mesenchymal transition [17]. Mutations in WNTβ have been identified in malignancies, such as hepatocellular, breast, colorectal and other cancer [18], however, the constitutive receptor stimulation can account for hyperactive WNTβ signalling in the absence of activating mutations as well [19]. Potapov et al. described the abundance of βcatenin in embryonal carcinoma identified by immunohistochemistry in 39 cases of testicular embryonal carcinomas [20]. Two studies reported differential expression in seminomas versus non-seminomas, but no correlation to resistance to chemotherapy was found [21, 22]. However, the role of βcatenin and WNTB signalling and its clinical implications in GCTs have not been comprehensively explored. In this retrospective study, we aimed to evaluate the role of βcatenin in GCTs and find correlations with systemic immune-inflammation and PD-L1 expression in tumor and tumor infiltrating lymphocytes (TILs).

Methods

Study patients

This translational study (chair M. Mego) included 247 patients with GCTs identified from the Slovak National Cancer Institute database, treated from January 1999 to December 2013 with available paraffin embedded tumor tissue specimen and sufficient follow-up clinical data. Patients with concurrent malignancy other than non-melanoma skin cancer in the previous 5 years were excluded. In all patients, data regarding age, tumor histology, clinical stage, type and number of metastatic sites, and delivery of systemic therapy were recorded and compared with βcatenin expression. Previously published data regarding PD-L1 expression on tumor/TILs and systemic immune-inflammation index (SII) in this cohort were available from 240 and 181 study patients, respectively [4, 23]. The Institutional Review Board (Ethics committee of Slovak National Cancer Institute in Bratislava approved this retrospective study (version 6.1 from 15 February 2017; ref. IZLO1) and a waiver of consent form patients was granted.

Tumor pathology

Pathology review was conducted at the Department of Pathology, Faculty of Medicine, Comenius University, by two pathologists (ZC and PB) associated with the study.

Diagnosis and tissue samples

Tumor tissue and normal testicular tissue were evaluated in all cases, when available. The study included tumor specimens from 247 patients before administration of systemic therapy. All but 9 of these specimens were obtained from primary testicular tumors. Biopsies of abdominal and mediastinal masses were performed in 7 and in 2 cases of primary extragonadal tumors, respectively. The GCTs were classified according to World Health Organization criteria [24]. Normal testicular tissue from non-cancer patients was not available for analysis, therefore we used normal tissue adjacent to testicular tumor for βcatenin expression evaluation, as described in previous studies [25, 26].

Tissue microarray construction

According to the tumor histology, one or two representative tumor areas from each histological subtype (1–6 cores from each tumor) of germ cell tumor were identified on H&E sections. In case of mixed germ cell tumors, selected GCT histologies were sampled to isolate a specific histological pattern. Samples from normal testicular tissue were also marked, if present. Sections were matched to their corresponding wax blocks (the donor blocks), and 3-mm diameter cores of the tissues were removed from these donor blocks with the multipurpose sampling tool Harris Uni-Core (Sigma-Aldrich, Steinheim, Germany) and inserted into the recipient master block. The recipient block was cut into 5-μm sections and sections were transferred to coated slides. Tissue microarray construction and immunohistochemical staining was described in detail previously [4].

Immunohistochemical staining

Slides were deparaffinized and rehydrated in phosphate buffered saline solution (10 mM, pH 7.2). The tissue epitopes were demasked using the automated water bath heating process in Dako PT Link (Dako, Glostrup, Denmark); the slides were incubated in TRIS-EDTA retrieval solution (10 mM TRIS, 1 mM EDTA pH 9.0) at 98 °C for 20 min. The slides were subsequently incubated for 1 h at room temperature with the primary mouse monoclonal antibody against Beta-Catenin (Dako, β-Catenin-1, IR702, Ready-to-Use) and immunostained using anti-mouse/anti-rabbit immuno-peroxidase polymer (EnVision FLEX/HRP, Dako, Glostrup, Denmark) for 30 min at room temperature, according to the manufacturer’s instructions. For visualization, the slides were reacted with diaminobenzidine substrate-chromogen solution (DAB, Dako, Glostrup, Denmark) for 5 min. Finally, the slides were counterstained with haematoxylin. Βcatenin positivity of epithelial cells in the colon was used as a positive control, same tissue with omitting of the primary antibody served as negative control.

Immunohistochemical stain scoring

Tumor cores were independently assessed by two observers (ZC and PB) who were blinded to the clinicopathological data. In cases of disagreement, the consensus was made. Tumor cells with βcatenin expression were scored by a weighted histoscore (HS) which accounts for both the extent of cell staining and the staining intensity [27]. The portion of positive cells was estimated on a scale from 0 to 100%. The average intensity of positively staining cells was given a score from 0 to 3 (0 = no staining; 1 = weak; 2 = intermediate; and 3 = strong staining). The HS was then calculated by multiplying the percentage score by the intensity score, to yield a minimum value of 0 and a maximum value of 300. Based on the HS, a βcatenin expression was graded as low (0–150) or high (160–300) as we described previously [25]. If multiple histologic subtypes were present in a sample, we chose the highest number among these for a final βcatenin expression of a mixed GCT.

Systemic immune-inflammation index

The SII is an index based on platelets (P), neutrophils (N) and lymphocytes (L) counts from the complete blood count. It was calculated as: SII = P x N/L as defined previously [28]. The median value (1003) obtained and validated in our previous study [23] was used as the cut-off value of SII, which was then dichotomized into low (below median) and high (above median) categories.

Ingenuity pathway analysis (IPA)

We used IPA to further explore the immune related effects and interactions of CTNNB1 (the gene encoding βcatenin) and CD274 (the gene encoding PD-L1). CTNNB1 and CD274 genes were entered into IPA search engine. Immune related effects were subsequently selected and interactions between CTNNB1, CD274 and the selected effects were assessed to create the network of interactions.

Statistical analysis

Patients’ characteristics were tabulated. A distribution of βcatenin HS was significantly different from the normal distribution (Shapiro–Wilk test), therefore we used non-parametric tests for analyses. Analyses of differences in distributions of βcatenin expression between the two groups of patients were performed using the Mann–Whitney U test, while Fisher’s exact test or the χ2 test when appropriate were used, when βcatenin expression was categorized as ‘low’ or ‘high’. A one-way analysis of variance and a Chi-square test were used for analyses of associations between the βcatenin and SII or PD-L1 expression.
A median follow-up period was calculated as a median observation time among all patients and among those still alive at the time of their last follow-up. PFS was calculated from the date of orchiectomy or the date of tumor biopsy to the date of progression or death or the date of the last adequate follow-up. OS was calculated from the date of orchiectomy or the date of tumor biopsy to the date of death or last the follow-up. PFS and OS were estimated using the Kaplan–Meier product limit method and compared by the log-rank test. All reported P values were two sided. A P value < 0.05 was considered as significant. Statistical analyses were performed using NCSS 2007 software (Hintze J, 2007, Kaysville, Utah, USA).

Results

Patients’ characteristics are shown in Table 1. Majority of patients had a non-seminoma histology. A testicular tumor was the most common primary and more than two thirds of patients were within good risk according to the IGCCCG (International Germ Cell Cancer Collaborative Group). Tumor specimens from 247 patients before administration of systemic therapy included 50 pure seminomas, 128 non-seminomas (86 embryonal carcinomas, 19 yolk sac tumors, 1 choriocarcinoma, 22 teratomas) and 69 mixed GCTs. (Additional file 1: Table S1). Eight cases of seminomas were clinically considered as non-seminomas based on the positivity of alpha-fetoprotein.
Table 1
Patients’ characteristics
 
N = 247
%
Age (years)
 Median (range)
30 (17–67)
 
Histology
 Pure seminoma
50
20.2
 Non-seminoma or mixed GCT
197
79.8
Primary tumor
 Gonadal
238
96.4
Primary retroperitoneal
7
2.8
Primary mediastinal
2
0.9
IGCCCG risk group
 Good risk
184
74.5
 Intermediate risk
29
11.7
 Poor risk
34
13.8
Sites of metastases
 Retroperitoneum
175
70.9
 Mediastinum
32
13.0
 Lungs
60
24.3
 Liver
18
7.3
 Other
34
13.8
 Non-pulmonary visceral metastases
22
8.9
No. of metastatic sites
 0
63
25.5
 1–2
141
57.1
  > 3
43
17.4
Mean (range) of pre-treatment markers
 AFP mIU/ml
1274 (0–60,570)
 β-HCG IU/ml
10,412 (0–423,338)
 LDH (mkat/l)
12 (2–89)

Distribution of the βcatenin expression among histological subtypes

βcatenin expression was found in specimens from 213 of 247 patients (86.2%) in this cohort. We observed membranous staining only, cytoplasmic or nuclear staining was not present. Significantly more non-seminomas/mixed GCTs (97.3%) showed positive staining for βcatenin compared to seminomas (51.8%), (P <  0.0001, Fisher’s exact test). The expression in all seminomas (including pure seminomas and components in mixed tumors) was low or none (median = 5; interquartile range 50) while non-seminomas have generally shown a stronger expression (median = 200; interquartile range 100), (P <  0.0001). Detailed expression in all subtypes is shown in Table 2 and Additional file 2: Figure S1.
Table 2
βcatenin expression in different histologic subtypes of germ cell tumors
Histologic subtype
 
Expression of βcatenin
N
Median
Interquartile range
p-value
Low
High
p-value
    
N
%
N
%
 
Seminoma
83
5
50
NA
81
97.6
2
2.4
NA
GCNIS
73
200
100
< 0.001
35
48.0
38
52.9
< 0.001
Embryonal carcinoma
134
200
100
< 0.001
63
47.0
71
53.0
< 0.001
Yolk sac tumor
31
200
0
< 0.001
7
22.6
24
77.4
< 0.001
Choriocarcinoma
13
100
165
< 0.001
7
53.8
6
46.2
< 0.001
Teratoma
58
100
173
< 0.001
32
55.2
26
44.8
< 0.001
All histological subtypes of non-seminoma showed any intensity in staining more frequently than seminomas (embryonal carcinoma 98.5%, teratoma 81.1%, yolk sac tumor 96.8%, choriocarcinoma 76.9%), (P <  0.0001 for EC, T, YST and P = 0.074 for CHC), (Fig. 1). Proportion of patients with high βcatenin expression (HS > 150) was significantly higher in GCNIS, YST and EC and lower in seminoma compared to adjacent normal testicular tissue (Table 2).

βcatenin and patients’ characteristics

βcatenin has shown significant associations with several clinical characteristics of patients in this cohort. Patients with testicular and retroperitoneal primary GCT had lower expression of βcatenin compared to patients with primary mediastinal non-seminoma (PMNSGCT) (P = 0.035) (Table 3). βcatenin was higher among intermediate and poor IGCCCG risk groups as opposed to good risk patients (P = 0.033). Similarly, the higher expression was seen in patients with highly elevated tumor markers (S2–3) compared to patients with no or mild marker elevation (S0–1) (P = 0.035). Metastatic sites were generally not associated with differences in the βcatenin expression (Table 3).
Table 3
Patients’ characteristics according to the expression of βcatenin
Variable
  
βcatenin
N
Median
interquartile range
p-value
Low
High
p-value
    
N
%
N
%
 
All patients
247
100
150
NA
131
53
116
47
 
Clinical subtype
 Pure seminoma
50
50
100
< 0.0001
42
84
8
16
< 0.0001
 Non-seminoma
197
200
115
 
89
45
108
55
 
Tumor primary
 Testicular/retroperitoneal
245
100
150
0.035
131
53
114
47
0.131
 Mediastinal
2
300
N/A
 
0
0
2
100
 
IGCCCG risk group
 Good
184
100
170
0.033
105
57
79
43
0.030
 Intermediate/Poor
63
200
130
 
26
42
37
58
 
Number of metastatic sites
 0
63
100
100
0.408
32
51
31
49
0.813
 1 to 2
142
100
175
 
75
53
67
47
 
  ≥ 3
42
100
142.5
 
24
57
18
43
 
Retroperitoneal LN metastases
 Absent
72
130
100
0.075
36
50
36
50
0.540
 Present
175
100
175
 
95
54
80
46
 
Mediastinal LN metastases
 Absent
215
100
150
0.598
117
54
98
46
0.260
 Present
32
200
168.75
 
14
44
18
56
 
Lung metastases
 Absent
187
100
150
0.967
98
52
89
48
0.726
 Present
60
100
145
 
33
55
27
45
 
Liver
 Absent
229
100
150
0.243
124
54
105
46
0.212
 Present
18
200
107.5
 
7
39
11
61
 
Non-pulmonary visceral metastases
 Absent
225
100
150
0.891
120
53
105
47
0.765
 Present
22
150
150
 
11
50
11
50
 
S – stage
 0–1
187
100
170
0.035
107
57
80
43
0.020
 2–3
60
200
130
 
24
40
36
60
 

βcatenin and patient outcome

A survival analysis did not show differences among patients expressing high versus low amounts of βcatenin prior to treatment for PFS and OS; (HR 0.70, 95% CI 0.41–1.19, P = 0.185) and (HR 0.89, 95% CI 0.45–1.74, P = 0.727), respectively (Additional file 3: Figure S2). Sub-analyses for survival in patients with seminoma and non-seminoma also did not show statistical differences for PFS and OS (data not shown). In the subsequent analyses, we assessed the βcatenin and its’ correlations with PD-L1 expression and the SII, which were previously assessed in the same patient cohort [3, 4, 23].

The association with PD-L1 on tumor cells, TILs and SII

The expression of PD-L1 on tumor cells have shown a significant correlation with the expression of βcatenin. The expression of PD-L1 was originally reported in our previous studies [3, 4]. Samples from 240 patients were previously examined for PD-L1 and currently for the expression of βcatenin. Patients with low βcatenin had lower expression of PD-L1 on tumor cells compared to patients with the high βcatenin (median = 40 vs 67.5, respectively; P = 0.010). Spearman’s correlation analysis confirmed the significant correlation between PD-L1 on tumor cells and βcatenin (r = 0.316; P = 0.0002). The PD-L1 expression on TILs did not significantly differ among patients with low (median = 100) vs high (median = 60) expression of βcatenin (P = 0.220).
βcatenin was significantly higher in patients with low SII (median = 200) vs high SII (median = 100) (P = 0.011).

βcatenin and the infiltration with TILs

We have performed an analysis to explore the association of βcatenin and the presence of TILs in tumor specimen of patients from our previous study [4]. Samples from 240 patients were previously examined for TILs and currently for the expression of βcatenin. Of 240 evaluated patients, 225 (93.7%) had their tumor infiltrated with TILs. In our cohort, patients with no TILs did not have significantly higher expression of βcatenin compared to patients with TILs present in the specimen (median = 200 vs 100, respectively; P = 0.083).

IPA

We have discovered several effects of aberrant CTNNB1 and CD274 (upregulations, mutations or interactions) related to differentiation, proliferation and activation of CD4+ and CD8+ lymphocytes. The results of the IPA are summarized in Fig. 2.

Discussion

In this translational study, we assessed the significance of the βcatenin expression in patients with GCT prior to administration of chemotherapy. We performed a survival analysis and correlated its’ expression to clinical and pathological characteristics. We identified a significant difference in the expression of βcatenin between seminomas and non-seminomas. A study by Fritsch et al. reported activated WNTβ and differential βcatenin expression in female paediatric GCT patients [29]. All non-seminoma subtypes in our study have shown strong staining for βcatenin with the highest expressions seen in embryonal carcinoma and yolk sac tumor. This finding can be attributed to the pluripotency of embryonal cancer cells that are known to be malignant counterparts of embryonic stem cells and act as stem cells in the development of GCTs. However, the implication for yolk sac tumor is unclear [30, 31]. Raggioli et al. demonstrated that βcatenin was vital for the maintenance of pluripotency associated genes in mouse embryonic stem cell lines. In addition, removal of βcatenin led to a strong increase of cell death [32]. Our study suggests the loss of βcatenin during the development from the progenitor cell, as only a weak or no expression was seen in seminomas. We hypothesize it can be responsible for different biology and treatment sensitivity compared to non-seminoma GCTs. The loss of other markers of pluripotency, such as SOX2 and CD30 was well described in the past [33]. Our further analysis uncovered differences in βcatenin expression in association with poor-risk clinical features. Primary testicular and retroperitoneal tumors had lower expression of βcatenin in comparison with PMNSGCTs which represent the worst prognostic group of GCT patients [34]. However, this sub-analysis is underpowered due to small number of PMNSGCTs in the cohort. The underlying connection to βcatenin and WTNβ signalling suggest that patients with tumors that have higher abilities for pluripotency belong to poorer risk category and produce more tumor markers. Although βcatenin assessed in our cohort of chemotherapy naïve patients did not show a prognostic significance in terms of PFS and OS, further studies assessing the βcatenin in relapsed or platinum refractory patients are needed to explore its’ clinical significance.
Our subsequent analysis evaluated βcatenin and its possible associations with the tumor immune microenvironment. Our previous works have shown prognostic significance of PD-L1 expressed on tumor cells and TILs and systemic inflammation in GCTs [3, 4, 23]. Herein we identified the positive correlation of βcatenin to PD-L1 expression on tumor cells, but not on TILs. High PD-L1 expression on TILs was linked to a better outcome, whereas high PD-L1 expression on tumor cells was predictive of poor outcome in GCTs. The role of βcatenin and its’ interaction with PD-1/PD-L1 pathway is not clear. Its’ association with PD-L1 on tumor cells but not on TILs may suggest the independence of PD-L1 positive TILs from WNTβ signalling. Moreover, the association with immune inflamed microenvironment was confirmed in inverse correlation with SII suggesting that patients with low inflammatory activity had activated WNTβ signalling. Spranger et al. observed a lack of T-cell infiltrate in autochtonous mouse models, which led to resistance to anti-PD-L1/anti-CTLA4 monoclonal antibody therapy. Subsequently, authors were able to restore T-cell infiltration in mice by introduction of injected dendritic cells harvested from transgenic mice [13]. We did not observe a higher expression of βcatenin in patients without TILs in tumor specimens, perhaps due to insufficient size of our cohort. Our subsequent ingenuity pathway analysis uncovered important roles of CTNNB1 and CD274 genes in the process of differentiation, proliferation and function of CD8+ and CD4+ T cells. The increase in number of CD8+ positive cells resulting from aberrant mutated CTNNB1 is seemingly conflicting with other described effects of WNT signalling, however, Cohen et al. described that these cells failed to be activated due to a lack of cross-presentation resulting from increased splenic CD8α dendritic cell activity [35]. Furthermore, animal model experiments performed by Augustin et al. in mouse embryonic stem cells with overexpressed WNT secretion factor Evi/WIs, showed an increased tumor growth and impaired immune cell recruitment in the presence of enhanced Evi expression. Authors observed reduced T-cell and B-cell infiltration especially in Evi overexpressing teratomas, thus, showing the negative effect of WNT signalling on immune surveillance [36].
Our study has some strengths and limitations. The large patient population creates a relatively balanced population for the biomarker assessment. Additionally, the availability of the PD-L1 expression and the SII data from the same set of patients creates a unique cohort with the possibility to uncover associations among these. Limitations include the retrospective nature of the analysis and underrepresentation of extragonadal germ cell tumors. The membranous expression of βcatenin does not provide clear evidence of WNT pathway signalling, thus the expression of a target gene would provide more information. Our selection of βcatenin to explore as a biomarker in this hypothesis generating study may be biased, therefore we plan a more comprehensive proteomic analysis in the future.

Conclusion

In conclusion, this is the first translational study to assess the biological, prognostic and clinical significance of βcatenin in GCTs of adult men. We uncovered the differences between seminomas and non-seminomas. We have shown associations with several clinical characteristics and systemic immune-inflammation and PD-L1 on tumor cells, thus providing the insights into immunobiology of GCTs. More studies are needed to validate this exploratory trial and to explore the possible therapeutic implications.

Acknowledgements

We would like to acknowledge our collaborators from departments of pathology in Slovakia: Antol M, Benko J, Danis D, Durcansky D, Fiala P, Galbavy S, Gogora M, Hudcovsky P, Macuch J, Martanovic P, Ondrias F, Plank L, Svajdler M. We would like to acknowledge Daniela Jantekova, from the Population Registry of Slovak Republic for updating patients’ database, Zlatica Pekova for administration support, Alzbeta Jancikova for tissue blocks and informed consent collection, Andrea Krieschova and Simona Turnova for informed consent collection, Emilia Klincova and Ludovit Gaspar for excellent technical assistance.

Funding

This study was supported by the Slovak Research and Development Agency under contract No. APVV-15-0086 and Scientific Grant Agency under contract number VEGA 1/0043/18.

Availability of data and materials

The datasets and materials used and analysed during the reported study are available at 2nd Department of Oncology and the Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.

Competing interest

The authors declare that they have no competing interests.
All procedures performed in this study involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent was obtained from all individual participants included in the study. The Institutional Review Board (Ethics committee of Slovak National Cancer Institute in Bratislava approved this retrospective study (version 6.1 from 15 February 2017; ref. IZLO1).
Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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.
Literatur
1.
Zurück zum Zitat Einhorn LH. Treatment of testicular cancer: a new and improved model. J Clin Oncol. 1990;8(11):1777–81.CrossRef Einhorn LH. Treatment of testicular cancer: a new and improved model. J Clin Oncol. 1990;8(11):1777–81.CrossRef
2.
Zurück zum Zitat Mardiak J, Salek T, Sycova-Mila Z, Obertova J, Reckova M, Mego M, Hlavata Z, Brozmanova K, Risnyovzska Z, Svetlovska D, et al. Paclitaxel, bleomycin, etoposide, and cisplatin (T-BEP) as initial treatment in patients with poor-prognosis germ cell tumors (GCT): a phase II study. Neoplasma. 2007;54(3):240–5.PubMed Mardiak J, Salek T, Sycova-Mila Z, Obertova J, Reckova M, Mego M, Hlavata Z, Brozmanova K, Risnyovzska Z, Svetlovska D, et al. Paclitaxel, bleomycin, etoposide, and cisplatin (T-BEP) as initial treatment in patients with poor-prognosis germ cell tumors (GCT): a phase II study. Neoplasma. 2007;54(3):240–5.PubMed
3.
Zurück zum Zitat Cierna Z, Mego M, Miskovska V, Machalekova K, Chovanec M, Svetlovska D, Hainova K, Rejlekova K, Macak D, Spanik S, et al. Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. Ann Oncol. 2016;27(2):300–5.CrossRef Cierna Z, Mego M, Miskovska V, Machalekova K, Chovanec M, Svetlovska D, Hainova K, Rejlekova K, Macak D, Spanik S, et al. Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. Ann Oncol. 2016;27(2):300–5.CrossRef
4.
Zurück zum Zitat Chovanec M, Cierna Z, Miskovska V, Machalekova K, Svetlovska D, Kalavska K, Rejlekova K, Spanik S, Kajo K, Babal P, et al. Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors. Oncotarget. 2017;8(13):21794–805.CrossRef Chovanec M, Cierna Z, Miskovska V, Machalekova K, Svetlovska D, Kalavska K, Rejlekova K, Spanik S, Kajo K, Babal P, et al. Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors. Oncotarget. 2017;8(13):21794–805.CrossRef
5.
Zurück zum Zitat Chovanec M, Mego M, Cholujova D, Gronesova P, Miskovska V, Sycova-Mila Z, Usakova V, Svetlovska D, Bujdak P, Spanik S, et al. A cytokine and angiogenic factor (CAF) analysis in plasma in testicular germ cell tumor patients (TGCTs). J Clin Oncol. 2015;33(15):e15552. Chovanec M, Mego M, Cholujova D, Gronesova P, Miskovska V, Sycova-Mila Z, Usakova V, Svetlovska D, Bujdak P, Spanik S, et al. A cytokine and angiogenic factor (CAF) analysis in plasma in testicular germ cell tumor patients (TGCTs). J Clin Oncol. 2015;33(15):e15552.
6.
Zurück zum Zitat Svetlovska D, Miskovska V, Cholujova D, Gronesova P, Cingelova S, Chovanec M, Sycova-Mila Z, Obertova J, Palacka P, Rajec J, et al. Plasma cytokines correlated with disease characteristic, progression free survival and overall survival in testicular germ cell tumour patients. Clin Genitourin Cancer. 2017; Article In Press. Svetlovska D, Miskovska V, Cholujova D, Gronesova P, Cingelova S, Chovanec M, Sycova-Mila Z, Obertova J, Palacka P, Rajec J, et al. Plasma cytokines correlated with disease characteristic, progression free survival and overall survival in testicular germ cell tumour patients. Clin Genitourin Cancer. 2017; Article In Press.
7.
Zurück zum Zitat Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 2015;372(4):311–9.CrossRef Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 2015;372(4):311–9.CrossRef
8.
Zurück zum Zitat Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.CrossRef Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.CrossRef
9.
Zurück zum Zitat Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–7.CrossRef Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–7.CrossRef
10.
Zurück zum Zitat Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, Bellmunt J, Burris HA, Petrylak DP, Teng SL, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515(7528):558–62.CrossRef Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, Bellmunt J, Burris HA, Petrylak DP, Teng SL, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515(7528):558–62.CrossRef
11.
Zurück zum Zitat Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69(7):3077–85.CrossRef Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69(7):3077–85.CrossRef
12.
Zurück zum Zitat Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, Alaparthy S, Berman D, Jure-Kunkel M, Siemers NO, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer immunology, immunotherapy : CII. 2012;61(7):1019–31.CrossRef Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, Alaparthy S, Berman D, Jure-Kunkel M, Siemers NO, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer immunology, immunotherapy : CII. 2012;61(7):1019–31.CrossRef
13.
Zurück zum Zitat Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–5.CrossRef Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–5.CrossRef
14.
Zurück zum Zitat ten Berge D, Kurek D, Blauwkamp T, Koole W, Maas A, Eroglu E, Siu RK, Nusse R. Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat Cell Biol. 2011;13(9):1070–5.CrossRef ten Berge D, Kurek D, Blauwkamp T, Koole W, Maas A, Eroglu E, Siu RK, Nusse R. Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat Cell Biol. 2011;13(9):1070–5.CrossRef
15.
Zurück zum Zitat Ying QL, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453(7194):519–23.CrossRef Ying QL, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453(7194):519–23.CrossRef
16.
Zurück zum Zitat Bhatia N, Spiegelman VS. Activation of Wnt/beta-catenin/Tcf signaling in mouse skin carcinogenesis. Mol Carcinog. 2005;42(4):213–21.CrossRef Bhatia N, Spiegelman VS. Activation of Wnt/beta-catenin/Tcf signaling in mouse skin carcinogenesis. Mol Carcinog. 2005;42(4):213–21.CrossRef
17.
Zurück zum Zitat Eger A, Stockinger A, Schaffhauser B, Beug H, Foisner R. Epithelial mesenchymal transition by c-Fos estrogen receptor activation involves nuclear translocation of beta-catenin and upregulation of beta-catenin/lymphoid enhancer binding factor-1 transcriptional activity. J Cell Biol. 2000;148(1):173–88.CrossRef Eger A, Stockinger A, Schaffhauser B, Beug H, Foisner R. Epithelial mesenchymal transition by c-Fos estrogen receptor activation involves nuclear translocation of beta-catenin and upregulation of beta-catenin/lymphoid enhancer binding factor-1 transcriptional activity. J Cell Biol. 2000;148(1):173–88.CrossRef
18.
Zurück zum Zitat Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al. COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in Cancer. Nucleic Acids Res. 2011;39(Database issue):D945–50.CrossRef Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al. COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in Cancer. Nucleic Acids Res. 2011;39(Database issue):D945–50.CrossRef
19.
Zurück zum Zitat Blagodatski A, Poteryaev D, Katanaev VL. Targeting the Wnt pathways for therapies. Mol Cell Ther. 2014;2:28.CrossRef Blagodatski A, Poteryaev D, Katanaev VL. Targeting the Wnt pathways for therapies. Mol Cell Ther. 2014;2:28.CrossRef
20.
Zurück zum Zitat Potapov S, Sidorenko R, Galata D, Stratiy N, Gargin V. Peculiarities of catenin activity in the embryonal testicular carcinoma. Georgian Med News. 2016;261:68–73. Potapov S, Sidorenko R, Galata D, Stratiy N, Gargin V. Peculiarities of catenin activity in the embryonal testicular carcinoma. Georgian Med News. 2016;261:68–73.
21.
Zurück zum Zitat Honecker F, Kersemaekers AM, Molier M, Van Weeren PC, Stoop H, De Krijger RR, Wolffenbuttel KP, Oosterhuis W, Bokemeyer C, Looijenga LH. Involvement of E-cadherin and beta-catenin in germ cell tumours and in normal male fetal germ cell development. J Pathol. 2004;204(2):167–74.CrossRef Honecker F, Kersemaekers AM, Molier M, Van Weeren PC, Stoop H, De Krijger RR, Wolffenbuttel KP, Oosterhuis W, Bokemeyer C, Looijenga LH. Involvement of E-cadherin and beta-catenin in germ cell tumours and in normal male fetal germ cell development. J Pathol. 2004;204(2):167–74.CrossRef
22.
Zurück zum Zitat Fernando G, Paul F, Laura J, Alejandra AM, Gabriela M, Alberto PL. Is the Wnt/beta catenin signalling pathway activated in seminoma?: An immunohistochemical study. J Cancer Res Ther. 2016;12(2):1075–9.CrossRef Fernando G, Paul F, Laura J, Alejandra AM, Gabriela M, Alberto PL. Is the Wnt/beta catenin signalling pathway activated in seminoma?: An immunohistochemical study. J Cancer Res Ther. 2016;12(2):1075–9.CrossRef
23.
Zurück zum Zitat Chovanec M, Cierna Z, Miskovska V, Machalekova K, Kalavska K, Rejlekova K, Svetlovska D, Macak D, Spanik S, Kajo K, et al. Systemic immune-inflammation index in germ-cell tumours. Br J Cancer. 2018;118(6):831–8.CrossRef Chovanec M, Cierna Z, Miskovska V, Machalekova K, Kalavska K, Rejlekova K, Svetlovska D, Macak D, Spanik S, Kajo K, et al. Systemic immune-inflammation index in germ-cell tumours. Br J Cancer. 2018;118(6):831–8.CrossRef
24.
Zurück zum Zitat Ulbright TM, Amin MB, Balzer B, Berney DM, Epstein JI, Gu C, Indrees MT, Looijenga LHJ, Paner G, Rajpert-De Meyts E, et al. Tumours of the testis and paratesticular tissue. In: Moch H, Humphrey PA, Ulbright TM, Reuter VE, editors. WHO Classification of Tumours of the Urinary System and Male Genital Organs. 4th ed. Lyon: International Agency for Research on Cancer (IARC); 2016. p. 185–258. Ulbright TM, Amin MB, Balzer B, Berney DM, Epstein JI, Gu C, Indrees MT, Looijenga LHJ, Paner G, Rajpert-De Meyts E, et al. Tumours of the testis and paratesticular tissue. In: Moch H, Humphrey PA, Ulbright TM, Reuter VE, editors. WHO Classification of Tumours of the Urinary System and Male Genital Organs. 4th ed. Lyon: International Agency for Research on Cancer (IARC); 2016. p. 185–258.
25.
Zurück zum Zitat Mego M, Cierna Z, Svetlovska D, Macak D, Machalekova K, Miskovska V, Chovanec M, Usakova V, Obertova J, Babal P, et al. PARP expression in germ cell tumours. J Clin Pathol. 2013;66(7):607–12.CrossRef Mego M, Cierna Z, Svetlovska D, Macak D, Machalekova K, Miskovska V, Chovanec M, Usakova V, Obertova J, Babal P, et al. PARP expression in germ cell tumours. J Clin Pathol. 2013;66(7):607–12.CrossRef
26.
Zurück zum Zitat Ulisse S, Baldini E, Mottolese M, Sentinelli S, Gargiulo P, Valentina B, Sorrenti S, Di Benedetto A, De Antoni E, D'Armiento M. Increased expression of urokinase plasminogen activator and its cognate receptor in human seminomas. BMC Cancer. 2010;10:151.CrossRef Ulisse S, Baldini E, Mottolese M, Sentinelli S, Gargiulo P, Valentina B, Sorrenti S, Di Benedetto A, De Antoni E, D'Armiento M. Increased expression of urokinase plasminogen activator and its cognate receptor in human seminomas. BMC Cancer. 2010;10:151.CrossRef
27.
Zurück zum Zitat Kirkegaard T, Edwards J, Tovey S, McGlynn LM, Krishna SN, Mukherjee R, Tam L, Munro AF, Dunne B, Bartlett JM. Observer variation in immunohistochemical analysis of protein expression, time for a change? Histopathology. 2006;48(7):787–94.CrossRef Kirkegaard T, Edwards J, Tovey S, McGlynn LM, Krishna SN, Mukherjee R, Tam L, Munro AF, Dunne B, Bartlett JM. Observer variation in immunohistochemical analysis of protein expression, time for a change? Histopathology. 2006;48(7):787–94.CrossRef
28.
Zurück zum Zitat Hu B, Yang XR, Xu Y, Sun YF, Sun C, Guo W, Zhang X, Wang WM, Qiu SJ, Zhou J, et al. Systemic immune-inflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clinical Cancer Res. 2014;20(23):6212–22.CrossRef Hu B, Yang XR, Xu Y, Sun YF, Sun C, Guo W, Zhang X, Wang WM, Qiu SJ, Zhou J, et al. Systemic immune-inflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clinical Cancer Res. 2014;20(23):6212–22.CrossRef
29.
Zurück zum Zitat Fritsch MK, Schneider DT, Schuster AE, Murdoch FE, Perlman EJ. Activation of Wnt/beta-catenin signaling in distinct histologic subtypes of human germ cell tumors. Pediatr Dev Pathol. 2006;9(2):115–31.CrossRef Fritsch MK, Schneider DT, Schuster AE, Murdoch FE, Perlman EJ. Activation of Wnt/beta-catenin signaling in distinct histologic subtypes of human germ cell tumors. Pediatr Dev Pathol. 2006;9(2):115–31.CrossRef
30.
Zurück zum Zitat Andrews PW, Matin MM, Bahrami AR, Damjanov I, Gokhale P, Draper JS. Embryonic stem (ES) cells and embryonal carcinoma (EC) cells: opposite sides of the same coin. Biochem Soc Trans. 2005;33(Pt 6):1526–30.CrossRef Andrews PW, Matin MM, Bahrami AR, Damjanov I, Gokhale P, Draper JS. Embryonic stem (ES) cells and embryonal carcinoma (EC) cells: opposite sides of the same coin. Biochem Soc Trans. 2005;33(Pt 6):1526–30.CrossRef
31.
Zurück zum Zitat Abu Dawud R, Schreiber K, Schomburg D, Adjaye J. Human embryonic stem cells and embryonal carcinoma cells have overlapping and distinct metabolic signatures. PLoS One. 2012;7(6):e39896.CrossRef Abu Dawud R, Schreiber K, Schomburg D, Adjaye J. Human embryonic stem cells and embryonal carcinoma cells have overlapping and distinct metabolic signatures. PLoS One. 2012;7(6):e39896.CrossRef
32.
Zurück zum Zitat Raggioli A, Junghans D, Rudloff S, Kemler R. Beta-catenin is vital for the integrity of mouse embryonic stem cells. PLoS One. 2014;9(1):e86691.CrossRef Raggioli A, Junghans D, Rudloff S, Kemler R. Beta-catenin is vital for the integrity of mouse embryonic stem cells. PLoS One. 2014;9(1):e86691.CrossRef
33.
Zurück zum Zitat de Jong J, Stoop H, Gillis AJ, Hersmus R, van Gurp RJ, van de Geijn GJ, van Drunen E, Beverloo HB, Schneider DT, Sherlock JK, et al. Further characterization of the first seminoma cell line TCam-2. Genes Chromosomes Cancer. 2008;47(3):185–96.CrossRef de Jong J, Stoop H, Gillis AJ, Hersmus R, van Gurp RJ, van de Geijn GJ, van Drunen E, Beverloo HB, Schneider DT, Sherlock JK, et al. Further characterization of the first seminoma cell line TCam-2. Genes Chromosomes Cancer. 2008;47(3):185–96.CrossRef
34.
Zurück zum Zitat Adra N, Althouse SK, Liu H, Brames MJ, Hanna NH, Einhorn LH, Albany C. Prognostic factors in patients with poor-risk germ-cell tumors: a retrospective analysis of the Indiana University experience from 1990 to 2014. Ann Oncol. 2016;27(5):875–9.CrossRef Adra N, Althouse SK, Liu H, Brames MJ, Hanna NH, Einhorn LH, Albany C. Prognostic factors in patients with poor-risk germ-cell tumors: a retrospective analysis of the Indiana University experience from 1990 to 2014. Ann Oncol. 2016;27(5):875–9.CrossRef
35.
Zurück zum Zitat Cohen SB, Smith NL, McDougal C, Pepper M, Shah S, Yap GS, Acha-Orbea H, Jiang A, Clausen BE, Rudd BD, et al. Beta-catenin signaling drives differentiation and proinflammatory function of IRF8-dependent dendritic cells. J Immunol. 2015;194(1):210–22.CrossRef Cohen SB, Smith NL, McDougal C, Pepper M, Shah S, Yap GS, Acha-Orbea H, Jiang A, Clausen BE, Rudd BD, et al. Beta-catenin signaling drives differentiation and proinflammatory function of IRF8-dependent dendritic cells. J Immunol. 2015;194(1):210–22.CrossRef
36.
Zurück zum Zitat Augustin I, Dewi DL, Hundshammer J, Rempel E, Brunk F, Boutros M. Immune cell recruitment in teratomas is impaired by increased Wnt secretion. Stem Cell Res. 2016;17(3):607–15.CrossRef Augustin I, Dewi DL, Hundshammer J, Rempel E, Brunk F, Boutros M. Immune cell recruitment in teratomas is impaired by increased Wnt secretion. Stem Cell Res. 2016;17(3):607–15.CrossRef
Metadaten
Titel
βcatenin is a marker of poor clinical characteristics and suppressed immune infiltration in testicular germ cell tumors
verfasst von
Michal Chovanec
Zuzana Cierna
Viera Miskovska
Katarina Machalekova
Katarina Kalavska
Katarina Rejlekova
Daniela Svetlovska
Dusan Macak
Stanislav Spanik
Karol Kajo
Pavel Babal
Michal Mego
Jozef Mardiak
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4929-x

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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