Skip to main content
Erschienen in: Surgery Today 1/2018

Open Access 12.07.2017 | Original Article

Expression of podoplanin in stromal fibroblasts plays a pivotal role in the prognosis of patients with pancreatic cancer

verfasst von: Kazuyoshi Hirayama, Hiroshi Kono, Yuuki Nakata, Yoshihiro Akazawa, Hiroyuki Wakana, Hisataka Fukushima, Hideki Fujii

Erschienen in: Surgery Today | Ausgabe 1/2018

Abstract

Purpose

To investigate the role of podoplanin (PDPN) expression in invasive ductal carcinoma of the pancreas (IDCP) in humans.

Methods

Tumor samples were obtained from 95 patients with IDCP. Immunohistochemical staining was done to evaluate the expression of PDPN in cancer tissues.

Results

PDPN was detected predominantly in stromal fibroblasts, stained with α-smooth muscle actin. The cutoff value of PDPN-positive areas was calculated according to a histogram. There was no significant difference in clinicopathologic factors between patients with high vs. those with low PDPN expression. The high PDPN group showed significantly poorer disease-free and disease-specific survival rates than the low PDPN group. Among patients from the high PDPN group, those with lymph node metastases and those with a tumor larger than 20 cm in diameter had significantly poorer prognoses than similar patients from the low PDPN group. Multivariate Cox proportional hazards analysis indicated that a high expression of PDPN was an independent risk factor for disease-specific survival.

Conclusions

PDPN expression in cancer-related fibrotic tissues is associated with a poor prognosis, especially in patients with large tumors or lymph node metastases.

Introduction

Pancreatic cancer is associated with one of the poorest prognoses of any cancer because early detection is difficult and it progresses rapidly [13]. The number of deaths from pancreatic cancer is increasing in Japan. In fact, more than 30,000 deaths from pancreatic cancer in 2013 slightly exceeded the number of deaths from liver cancer in the same year [4].
Pancreatic fibrosis is one of the histopathologic findings at the time of the desmoplastic reaction associated with chronic pancreatitis or pancreatic cancer. Pancreatic stellate cells (PSCs) were first isolated and identified in the pancreas in 1998 [5, 6]. It was found that PSCs are similar to liver stellate cells and play a pivotal role in pancreatic fibrosis. In the normal pancreas, PSCs are quiescent and store cytoplasmic vitamin A-containing lipid droplets [7]. Inflammatory stimulation or signals from cancer cells activate PSCs, which develop a myofibroblast-like morphology and produce an extracellular matrix [7]. Activated PSCs secrete various growth factors and cytokines, such as fibroblast growth factor, transforming growth factor-β, stromal cell-derived factor 1, and interleukin-6 [8].
Podoplanin (PDPN), a 38-kDa type I transmembrane glycoprotein, is known as a marker of lymphatic endothelial cells [9]. In normal tissues, PDPN is expressed in kidney podocytes [10], alveolar type I cells [11], osteocytes [12], basal keratinocytes [13], and mesothelial cells [13]. Recently, PDPN was found in several other cancers, such as brain tumors [14], squamous cell carcinomas [15], germ cell tumors [16], and mesotheliomas [17]. It has been reported that PDPN expression is associated with malignancy in malignant astrocytic tumors [14]. PDPN is also found in some stromal fibroblasts, and an abundance of PDPN-positive stromal fibroblasts is associated with poor prognosis in lung adenocarcinoma, invasive breast cancer, and esophageal squamous cell carcinoma patients [1820]. Since pancreatic cancer is rich in fibrous tissues, we investigated the correlation between PDPN expression in stromal fibroblasts in invasive ductal carcinoma of the pancreas (IDCP) and prognosis in humans.

Materials and methods

Patients and pancreatic cancer samples

Pancreatic cancer samples were obtained from 95 patients with IDCP, who underwent surgery at the University of Yamanashi Hospital between 1995 and 2013. Table 1 summarizes the clinicopathological characteristics of the patients. The histological diagnosis of the specimens was confirmed based on the criteria of the uploaded World Health Organization classification [21]. The stage was graded according to the Union for International Cancer Control (UICC) classification, 7th edition [22]. There were 57 men and 38 women, ranging in age from 46 to 83 years (median 70.0). One patient had stage 0 disease, five had stage IA, three had stage IB, 29 had stage IIA, 56 had stage IIB, and one had stage III. This study was approved by the Ethics Committee of Yamanashi University (approved no. 1565) and was performed in accordance with the ethical standards of the Declaration of Helsinki and its later amendments. Serum carcinoembryonic antigen, carbohydrate antigen 19-9, Duke pancreatic monoclonal antigen type 2, and s-pancreas antigen-1 levels were measured at least every 3 months. Computed tomography from the chest to pelvis was performed at least every 6 months. Survival was measured from the time of pancreatic resection until death or censor. The follow-up duration ranged from 3 to 191 months.
Table 1
Clinicopathological characteristics of the 95 patients with invasive ductal carcinoma of the pancreas
Variables
Number of patients
%
PDPN positive area (%)
11.83 (0.45–36.29)
Age (years)
70.0 (46–83)
Sex
 Male
57
60
 Female
38
40
Histologic grade
 G1
28
29.5
 G2
55
57.9
 G3
12
12.6
Tumor size (mm)
27.9 (3–90)
Microscopic venous invasion
 Yes
83
87.4
 No
12
12.6
Microscopic lymphatic vessel invasion
 Yes
70
73.7
 No
25
26.3
UICC T category
 0
1
1.1
 1
6
6.3
 2
3
3.2
 3
84
88.4
 4
1
1.1
UICC N category
 0
39
41.1
 1
56
58.9
UICC stage
 0
1
1.1
 IA
5
5.3
 IB
3
3.2
 IIA
29
30.5
 IIB
56
58.9
 III
1
1.1
UICC Union for International Cancer Control, PDPN podoplanin

Immunohistochemistry for D2-40 and α-SMA

Formalin-fixed, paraffin-embedded tissue specimens were cut into 4-µm sections. Each section was mounted on a silane-coated glass slide, deparaffinized, and treated in antigen retrieval solution for 15 min at 120 °C using Dako REAL Target Retrieval Solution (Dako, Carpentaria, CA, USA). Endogenous peroxidase was quenched by incubation at room temperature in 0.3% H2O2, followed by rinsing with phosphate-buffered saline. Endogenous biotin was quenched using the Dako Biotin Blocking System (Dako). Sections were blocked using 5% normal blocking serum for 20 min. Mouse monoclonal to D2-40 antibodies (1:40; Abcam, Cambridge, UK) were applied overnight at 4 °C to stain PDPN. Rabbit polyclonal to α-smooth muscle actin antibodies (α-SMA, 1:200; Abcam) were applied for 2 h at room temperature. Following incubation, immunoperoxidase staining was completed using a Vectastain ABC elite kit (Vector Laboratories, Burlingame, CA, USA) and 3,3′-diaminobenzidine-tetrachloride as a chromogen. The D2-40-positive area was calculated from three different (100×) fields and is expressed as a percentage of the total area of the field using PhotoShop and Image J software. To calculate the cutoff value of the PDPN-positive area, a histogram was created (Fig. 1).

Statistical analysis

Data are expressed as mean ± standard error of the mean (SEM). Comparisons between two groups were assessed using the unpaired t test. Associations between different categorical variables were assessed using the χ 2 test. Survival rates were calculated using the Kaplan–Meier method, and significant differences in survival were determined by the log-rank test. The Cox proportional hazards model served for uni- and multivariable survival analysis. p < 0.05 was considered significant.

Results

Analysis of PDPN expression in pancreatic cancer by immunohistochemistry for D2-40

We performed immunohistochemical staining for D2-40 and α-SMA to evaluate PDPN expression in pancreatic cancer (Fig. 2). The areas that expressed PDPN in the pancreatic cancer also expressed α-SMA, a marker of stromal fibroblasts [23]. The PDPN-positive area in the pancreatic cancers ranged from 0.45 to 36.29% (median 11.83). A histogram was produced to establish the cutoff value of the PDPN-positive area, which was 11.83%, being the median value of the PDPN-positive areas. Patients with high expression of PDPN accounted for 52.6% of the patients (n = 50). There was no significant difference in clinicopathologic factors, except for the PDPN-positive area, between the group with high PDPN expression (high PDPN group) and the group with low PDPB expression (low PDPN group; Table 2).
Table 2
Relationships between podoplanin expression and clinicopathologic factors
Variables
High PDPN expression
Low PDPN expression
p value
n = 50 (52.6%)
n = 45 (47.4%)
Number of patients
%
Number of patients
%
PDPN positive area (%)
20.21
 
6.74
 
<0.0001
Age (years)
69.2
 
69
 
0.926
Sex
    
0.675
 Male
29
58
28
62.2
 
 Female
21
42
17
37.8
 
Histologic grade
    
0.241
 G1
12
24
16
35.6
 
 G2
33
66
22
48.9
 
 G3
5
10
7
15.6
 
Tumor size (mm)
28.3
 
27.5
 
0.791
Microscopic venous invasion
    
0.152
 Yes
46
92
37
82.2
 
 No
4
8
8
17.8
 
Microscopic lymphatic vessel invasion
    
0.141
 Yes
40
80
30
66.7
 
 No
10
20
15
33.3
 
UICC T category
    
0.569
 0
0
0
1
2.2
 
 1
4
8
2
4.4
 
 2
1
2
2
4.4
 
 3
44
88
40
88.9
 
 4
1
2
0
0
 
UICC N category
    
0.826
 0
20
40
19
42.2
 
 1
30
60
26
57.8
 
UICC stage
    
0.521
 0
0
0
1
2.2
 
 IA
4
8
1
2.2
 
 IB
1
2
2
4.4
 
 IIA
14
28
15
33.3
 
 IIB
30
60
26
57.8
 
 III
1
2
0
0
 
Recurrence
    
0.409
 Yes
33
66
26
57.8
 
 No
17
34
19
42.2
 
Statistical significances were calculated using the student’s t test or the χ 2 test
PDPN podoplanin

Correlation between PDPN expression in pancreatic cancer and prognosis

The high PDPN group had significantly poorer disease-free survival (DFS) and disease-specific survival (DSS) rates than the low PDPN group (Fig. 3). The median survival times for the high and low PDPN groups were 659 and 1212 days, respectively. We then analyzed survival according to the presence of lymph node metastasis. In the patients without lymph node metastasis, there was no significant difference in DFS or DSS according to PDPN expression, but in those with lymph node metastases, the high PDPN group had significantly poorer DFS and DSS rates than the low PDPN group (Fig. 4). There was no significant difference in the PDPN-positive area between patients with and those without lymph node metastasis (Fig. 5).
Focusing on the tumor size, in patients with tumors ≤20 mm, there was no significant difference in DFS or DSS according to PDPN expression, but in those with tumors >20 mm, the high PDPN group had significantly poorer DFS and DSS rates than the low PDPN group (Fig. 6). There was no correlation between PDPN expression and tumor size (Fig. 7).

Prognostic factors

We adopted factors found to be significant by univariate analysis, based on the multivariate Cox proportional hazards analysis. A high expression of PDPN was an independent risk factor for DSS (relative risk (RR) = 2.153, p = 0.022) and tumor size >20 mm was an independent risk factor for both DFS (RR = 2.514, p = 0.013) and DSS (RR = 2.535, p = 0.032; Table 3).
Table 3
Univariate and multivariate survival analysis (Cox regression model) of clinicopathologic factors and podoplanin expression
Factors
DFS
DSS
Univariate
Multivariate
Univariate
Multivariate
p value
Hazard ratio
95% CI
p value
p value
Hazard ratio
95% CI
p value
PDPN positive area: <11.83 vs. ≥11.83%
0.047
  
NS
0.006
2.153
1.116–4.153
0.022
Age: <65 vs. ≥65 years
0.547
  
NA
0.842
  
NA
Sex: female vs. male
0.663
  
NA
0.528
  
NA
Histologic grade: G1 vs. G2 and G3
0.189
  
NA
0.003
2.592
1.159–5.795
0.020
Tumor size: ≤20 vs. >20 mm
<0.001
2.514
1.211–5.220
0.013
0.002
2.535
1.081–5.943
0.032
Microscopic venous invasion: no vs. yes
0.366
  
NA
0.980
  
NA
Microscopic lymphatic vessel invasion: no vs. yes
0.126
  
NA
0.651
  
NA
UICC T category: T0, T1, and T2 vs. T3 and T4
0.014
  
NS
0.047
  
NS
UICC N category: N0 vs. N1
<0.001
  
NS
0.021
  
NS
UICC stage: 0-IIA vs. IIB/III
<0.001
2.223
1.203–4.105
0.011
0.011
  
NS
DFS disease-free survival, DSS disease-specific survival, CI confidence interval, NA not adopted, NS not significant, UICC Union for International Cancer Control, PDPN podoplanin

Discussion

The expression of stromal fibroblasts markers, including α-SMA, vimentin, desmin, fibroblast specific protein-1, and fibroblast-activation protein, is reported to vary depending on the local microenvironment of tumors [23]. Apte et al. reported that α-SMA-positive activated PSC also expressed glial fibrillary acidic protein or/and desmin in pancreatic cancer [24]. Recent studies investigating the roles of PSCs in pancreatic cancer have identified a mechanism of interaction among proliferation, invasion, and metastasis [7, 8, 2531]. In the present study, PDPN-expressing stromal cells in pancreatic cancer also expressed α-SMA, suggesting that they were most likely derived from PSCs (Fig. 2).
PDPN expression in stromal fibroblasts in pancreatic cancer was reported to be associated with lymphatic invasion, vascular invasion, the tumor size, histological grade, UICC classification T stage, and a shorter survival period [32]. Those results indicate that PDPN expression is associated with the progression of carcinoma in local recurrence, hematogenous metastasis, and lymphogenous metastasis [32, 33]. The present study found no significant difference in clinicopathologic factors between the high and low PDPN groups (Table 2). There were also no significant differences between PDPN expression and the incidence of lymph node metastases or the size of tumors (Figs. 5, 7). PDPN could be related to tumor growth, leading to poor prognosis, and its expression might be determined by the genotype of each tumor. Importantly, DFS and DSS were significantly poorer in the high PDPN group (Fig. 3). Thus, as PDPN expression is involved in the prognosis of pancreatic cancer patients, it may be a useful marker to identify patients with a poor prognosis after surgery.
Based on previous reports, lymphatic invasion, vascular invasion, tumor size, pathological grade, and UICC classification T stage are all useful for pathologic staging; however, no effective molecular targeting therapy after surgery has been established. In this regard, PDPN may be a useful and effective molecular target for therapy. Indeed, Kato et al. developed a cancer-specific monoclonal antibody against human PDPN, which reacted with PDPN-expressing cancer cells, but not with normal cells [34]. Although this antibody is promising for molecular targeting therapy against PDPN-expressing cancers, PDPN is expressed only on the stromal fibroblasts surrounding tumors in pancreatic cancer. Conversely, Suzuki-Inoue et al. reported that PDPN expressed in cancer cells promotes platelet aggregation and it may also be involved in migration, invasion, metastasis, and the malignant progression of cancer cells [35]. Thus, PDPN expressed in stromal fibroblasts may be involved in cancer progression, leading to a poor prognosis via mechanisms of multiple growth factors derived from activated platelets. Further investigations are needed to realize useful targeting therapy against PDPN in pancreatic cancer.

Conclusions

Podoplanin expression in stromal fibroblasts is associated with the poor prognosis of patients with large tumors or lymph node metastases of pancreatic cancer. Our findings suggest that patients with high expression of PDPN should be followed-up more closely after surgery. PDPN may become an important target of therapy for pancreatic cancer.

Compliance with ethical standards

Conflict of interest

Dr. Hirayama and his co-authors have no conflicts of interest.
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.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Yamada D, Eguchi H, Asaoka T, Tomohara H, Noda T, Wada H, et al. The basal nutritional state of PDAC patients is the dominant factor for completing adjuvant chemotherapy. Surg Today. 2017. doi:10.1007/s00595-017-1522-x. Yamada D, Eguchi H, Asaoka T, Tomohara H, Noda T, Wada H, et al. The basal nutritional state of PDAC patients is the dominant factor for completing adjuvant chemotherapy. Surg Today. 2017. doi:10.​1007/​s00595-017-1522-x.
2.
Zurück zum Zitat Fukuda Y, Yamada D, Eguchi H, Iwagami Y, Noda T, Asaoka T, et al. A novel preoperative predictor of pancreatic fistula using computed tomography after distal pancreatectomy with staple closure. Surg Today. 2017. doi:10.1007/s00595-017-1495-9.PubMed Fukuda Y, Yamada D, Eguchi H, Iwagami Y, Noda T, Asaoka T, et al. A novel preoperative predictor of pancreatic fistula using computed tomography after distal pancreatectomy with staple closure. Surg Today. 2017. doi:10.​1007/​s00595-017-1495-9.PubMed
3.
Zurück zum Zitat Tomihara H, Eguchi H, Yamada D, Gotoh K, Kawamoto K, Wada H, et al. Preoperative chemoradiotherapy does not compromise the feasibility of adjuvant chemotherapy for patients with pancreatic ductal adenocarcinoma. Surg Today. 2017;47:218–26.CrossRefPubMed Tomihara H, Eguchi H, Yamada D, Gotoh K, Kawamoto K, Wada H, et al. Preoperative chemoradiotherapy does not compromise the feasibility of adjuvant chemotherapy for patients with pancreatic ductal adenocarcinoma. Surg Today. 2017;47:218–26.CrossRefPubMed
4.
Zurück zum Zitat Ministry of Health, Labour and Welfare. Vital statistics Japan. Center for Cancer Control and Information Services, National Cancer Center, Japan. 2015. Ministry of Health, Labour and Welfare. Vital statistics Japan. Center for Cancer Control and Information Services, National Cancer Center, Japan. 2015.
5.
Zurück zum Zitat Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan GW, Korsten MA, et al. Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture. Gut. 1998;43:128–33.CrossRefPubMedPubMedCentral Apte MV, Haber PS, Applegate TL, Norton ID, McCaughan GW, Korsten MA, et al. Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture. Gut. 1998;43:128–33.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, et al. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115:421–32.CrossRefPubMed Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, et al. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115:421–32.CrossRefPubMed
7.
Zurück zum Zitat Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Investig. 2007;117:50–9.CrossRefPubMedPubMedCentral Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Investig. 2007;117:50–9.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer Res. 2008;68:7707–10.CrossRefPubMed Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer Res. 2008;68:7707–10.CrossRefPubMed
9.
Zurück zum Zitat Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 1999;154:385–94.CrossRefPubMedPubMedCentral Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 1999;154:385–94.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Matsui K, Breitender-Geleff S, Soleiman A, Kowalski H, Kerjaschki D. Podoplanin, a novel 43-kDa membrane protein, controls the shape of podocytes. Nephrol Dial Transplant. 1999;14(Suppl 1):9–11.CrossRefPubMed Matsui K, Breitender-Geleff S, Soleiman A, Kowalski H, Kerjaschki D. Podoplanin, a novel 43-kDa membrane protein, controls the shape of podocytes. Nephrol Dial Transplant. 1999;14(Suppl 1):9–11.CrossRefPubMed
11.
Zurück zum Zitat Vanderbilt JN, Dobbs LG. Characterization of the gene and promoter for RTI40, a differentiation marker of type I alveolar epithelial cells. Am J Respir Cell Mol Biol. 1998;19(4):662–71.CrossRefPubMed Vanderbilt JN, Dobbs LG. Characterization of the gene and promoter for RTI40, a differentiation marker of type I alveolar epithelial cells. Am J Respir Cell Mol Biol. 1998;19(4):662–71.CrossRefPubMed
12.
Zurück zum Zitat Wetterwald A, Hofstetter W, Cecchini MG, Lanske B, Wagner C, Fleish H, et al. Characterization and cloning of the E11 antigen, a marker expressed by rat osteoblasts and osteocytes. Bone. 1996;18:125–32.CrossRefPubMed Wetterwald A, Hofstetter W, Cecchini MG, Lanske B, Wagner C, Fleish H, et al. Characterization and cloning of the E11 antigen, a marker expressed by rat osteoblasts and osteocytes. Bone. 1996;18:125–32.CrossRefPubMed
13.
Zurück zum Zitat Schacht V, Dadras SS, Johnson LA, Jackson DG, Hong YK, Detmar M. Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol. 2005;166:913–21.CrossRefPubMedPubMedCentral Schacht V, Dadras SS, Johnson LA, Jackson DG, Hong YK, Detmar M. Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol. 2005;166:913–21.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Mishima K, Kato Y, Kaneko MK, Nishikawa R, Hirose T, Matsutani M. Increased expression of podoplanin in malignant astrocytic tumors as a novel molecular marker of malignant progression. Acta Neuropathol. 2006;111(5):483–8.CrossRefPubMed Mishima K, Kato Y, Kaneko MK, Nishikawa R, Hirose T, Matsutani M. Increased expression of podoplanin in malignant astrocytic tumors as a novel molecular marker of malignant progression. Acta Neuropathol. 2006;111(5):483–8.CrossRefPubMed
15.
Zurück zum Zitat Martín-Villar E, Scholl FG, Gamallo C, Yurrita MM, Muñoz-Guerra M, Cruces J, et al. Characterization of human PA2.26 antigen (T1α-2, podoplanin), a small membrane mucin induced in oral squamous cell carcinoma. Int J Cancer. 2005;113(6):899–910.CrossRefPubMed Martín-Villar E, Scholl FG, Gamallo C, Yurrita MM, Muñoz-Guerra M, Cruces J, et al. Characterization of human PA2.26 antigen (T1α-2, podoplanin), a small membrane mucin induced in oral squamous cell carcinoma. Int J Cancer. 2005;113(6):899–910.CrossRefPubMed
16.
Zurück zum Zitat Kato Y, Sasagawa I, Kaneko M, Osawa M, Fujita N, Tsuruo T. Aggrus: a diagnostic marker that distinguishes seminoma from embryonal carcinoma in testicular germ cell tumors. Oncogene. 2004;23(52):8552–6.CrossRefPubMed Kato Y, Sasagawa I, Kaneko M, Osawa M, Fujita N, Tsuruo T. Aggrus: a diagnostic marker that distinguishes seminoma from embryonal carcinoma in testicular germ cell tumors. Oncogene. 2004;23(52):8552–6.CrossRefPubMed
17.
18.
Zurück zum Zitat Kawase A, Ishii G, Nagai K, Ito T, Nagano T, Murata Y, et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. 2008;123:1053–9.CrossRefPubMed Kawase A, Ishii G, Nagai K, Ito T, Nagano T, Murata Y, et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. 2008;123:1053–9.CrossRefPubMed
19.
Zurück zum Zitat Schoppmann SF, Berghoff A, Dinhof C, Jakesz R, Gnant M, Dubsky P, et al. Podoplanin-expressing cancer-associated fibroblasts are associated with poor prognosis in invasive breast cancer. Breast Cancer Res Treat. 2012;134:237–44.CrossRefPubMed Schoppmann SF, Berghoff A, Dinhof C, Jakesz R, Gnant M, Dubsky P, et al. Podoplanin-expressing cancer-associated fibroblasts are associated with poor prognosis in invasive breast cancer. Breast Cancer Res Treat. 2012;134:237–44.CrossRefPubMed
20.
Zurück zum Zitat Tong L, Yuan S, Feng F, Zhang H. Role of podoplanin expression in esophageal squamous cell carcinoma: a retrospective study. Dis Esophagus. 2012;25:72–80.CrossRefPubMed Tong L, Yuan S, Feng F, Zhang H. Role of podoplanin expression in esophageal squamous cell carcinoma: a retrospective study. Dis Esophagus. 2012;25:72–80.CrossRefPubMed
21.
Zurück zum Zitat Hruban RH, Klöppel G, Boffetta P, Maitra A, Hiraoka N, Offerhaus GJA, et al. Ductal adenocarcinoma of the pancreas. In: WHO classification of tumors of the digestive system. World Health Organization; 2010. p. 281–91. Hruban RH, Klöppel G, Boffetta P, Maitra A, Hiraoka N, Offerhaus GJA, et al. Ductal adenocarcinoma of the pancreas. In: WHO classification of tumors of the digestive system. World Health Organization; 2010. p. 281–91.
22.
Zurück zum Zitat Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, editors. Exocrine and endocrine pancreas, AJCC cancer staging manual. 7th ed. New York: Springer; 2010. p. 241–8. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, editors. Exocrine and endocrine pancreas, AJCC cancer staging manual. 7th ed. New York: Springer; 2010. p. 241–8.
23.
Zurück zum Zitat Kallui R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401.CrossRef Kallui R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401.CrossRef
24.
Zurück zum Zitat Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004;29:179–87.CrossRefPubMed Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004;29:179–87.CrossRefPubMed
25.
Zurück zum Zitat Algul H, Treiber M, Lesina M, Schmid RM. Mechanisms of disease: chronic inflammation and cancer in pancreas—a potential role for pancreatic stellate cells? Nat Clin Pract Gastroenterol Hepatol. 2007;4:452–62.CrossRef Algul H, Treiber M, Lesina M, Schmid RM. Mechanisms of disease: chronic inflammation and cancer in pancreas—a potential role for pancreatic stellate cells? Nat Clin Pract Gastroenterol Hepatol. 2007;4:452–62.CrossRef
26.
Zurück zum Zitat Aoki H, Ohnishi H, Hama K, Ishijima T, Satoh Y, Hanatsuka K, et al. Autocrine loop between TGF-beta1 and IL-1 beta through Smad3- and ERK-dependent pathways in rat pancreatic stellate cells. Am J Physiol Cell Physiol. 2006;290:C1100–8.CrossRefPubMed Aoki H, Ohnishi H, Hama K, Ishijima T, Satoh Y, Hanatsuka K, et al. Autocrine loop between TGF-beta1 and IL-1 beta through Smad3- and ERK-dependent pathways in rat pancreatic stellate cells. Am J Physiol Cell Physiol. 2006;290:C1100–8.CrossRefPubMed
27.
Zurück zum Zitat Bachem MG, Zhou S, Buck K, Schneiderhan W, Siech M. Pancreatic stellate cells—role in pancreas cancer. Langenbecks Arch Surg. 2008;393:891–900.CrossRefPubMed Bachem MG, Zhou S, Buck K, Schneiderhan W, Siech M. Pancreatic stellate cells—role in pancreas cancer. Langenbecks Arch Surg. 2008;393:891–900.CrossRefPubMed
28.
Zurück zum Zitat Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68:918–26.CrossRefPubMedPubMedCentral Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68:918–26.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation is dependent on extracellular matrix proteins. Pancreas. 2004;28:38–44.CrossRefPubMed Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation is dependent on extracellular matrix proteins. Pancreas. 2004;28:38–44.CrossRefPubMed
30.
Zurück zum Zitat Sato N, Maehara N, Goggins M. Gene expression profiling of tumor-stromal interaction between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 2004;64:6950–6.CrossRefPubMed Sato N, Maehara N, Goggins M. Gene expression profiling of tumor-stromal interaction between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 2004;64:6950–6.CrossRefPubMed
31.
Zurück zum Zitat Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68:2085–93.CrossRefPubMed Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68:2085–93.CrossRefPubMed
32.
Zurück zum Zitat Shindo K, Aishima S, Ohuchida K, Fujiwara K, Fujino M, Mizuuchi Y, et al. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Mol Cancer. 2013;12:168–83.CrossRefPubMedPubMedCentral Shindo K, Aishima S, Ohuchida K, Fujiwara K, Fujino M, Mizuuchi Y, et al. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Mol Cancer. 2013;12:168–83.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Suzuki-Inoue K, Kato Y, Inoue O, Kaneko MK, Mishima K, Yatomi Y, et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem. 2007;282:25993–6001.CrossRefPubMed Suzuki-Inoue K, Kato Y, Inoue O, Kaneko MK, Mishima K, Yatomi Y, et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem. 2007;282:25993–6001.CrossRefPubMed
Metadaten
Titel
Expression of podoplanin in stromal fibroblasts plays a pivotal role in the prognosis of patients with pancreatic cancer
verfasst von
Kazuyoshi Hirayama
Hiroshi Kono
Yuuki Nakata
Yoshihiro Akazawa
Hiroyuki Wakana
Hisataka Fukushima
Hideki Fujii
Publikationsdatum
12.07.2017
Verlag
Springer Japan
Erschienen in
Surgery Today / Ausgabe 1/2018
Print ISSN: 0941-1291
Elektronische ISSN: 1436-2813
DOI
https://doi.org/10.1007/s00595-017-1559-x

Weitere Artikel der Ausgabe 1/2018

Surgery Today 1/2018 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

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