Open Access

Claudin‑9 is a novel prognostic biomarker for endometrial cancer

  • Authors:
    • Yuta Endo
    • Kotaro Sugimoto
    • Makoto Kobayashi
    • Yasuyuki Kobayashi
    • Manabu Kojima
    • Shigenori Furukawa
    • Shu Soeda
    • Takafumi Watanabe
    • Atsuko Y. Higashi
    • Tomohito Higashi
    • Yuko Hashimoto
    • Keiya Fujimori
    • Hideki Chiba
  • View Affiliations

  • Published online on: September 21, 2022     https://doi.org/10.3892/ijo.2022.5425
  • Article Number: 135
  • Copyright: © Endo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The tight‑junction protein claudin‑9 (CLDN9) is barely distributed in normal adult tissues but is ectopically expressed in various cancer types. Although multiple databases indicated upregulation of CLDN9 in endometrial cancers at the mRNA level, its protein expression and biological roles remain obscure. In the present study, the prognostic significance of CLDN9 expression in endometrial cancer was evaluated by immunohistochemical staining and semi‑quantification using formalin‑fixed paraffin‑embedded specimens obtained from 248 endometrial carcinoma cases. A total of 43 cases (17.3%) had high CLDN9 expression, whereas 205 cases (82.7%) exhibited low CLDN9 expression. The 5‑year disease‑specific survival rates in the high and low CLDN9 expression groups were 62.8 and 87.8% (P<0.001), respectively. In addition, multivariate analysis revealed that high CLDN9 expression was an independent prognostic factor (hazard ratio, 4.99; 95% CI, 1.96‑12.70; P<0.001). Furthermore, CLDN9 expression was significantly correlated with the expression of CLDN6 (P<0.001), which is the closest CLDN member to CLDN9 and a poor prognostic factor for endometrial carcinoma. The 5‑year disease‑specific survival rate of cases with CLDN6‑high/CLDN9‑high, CLDN6‑high/CLDN9‑low and CLDN6‑low/CLDN9‑high status was 30.0, 37.5 and 72.7%, respectively, whereas that of CLDN6‑low/CLDN9‑low was 89.8% (P=0.004). In conclusion, aberrant CLDN9 expression is a predictor of poor prognosis for endometrial cancer and may be utilized in combination with CLDN6 to achieve higher sensitivity.

Introduction

Endometrial cancer is the most common gynecological malignancy in developed countries and is increasing in both incidence and associated mortality (1-3). Most patients with endometrial cancer are diagnosed in the early stages and exhibit a favorable 5-year relative survival rate (95%), if the appropriate surgical procedure is provided (4). On the other hand, patients with regional spread and distant metastasis beyond the uterus have a poor 5-year relative survival rate (69 and 17%, respectively) (4). Although radiation therapy, hormonal therapy and chemotherapy with cytotoxic agents have been used for advanced and recurrent endometrial cancer, the effectiveness of these therapies is limited. It is reported that the efficacy of the first-line chemotherapy is 30-57% and that the median progression-free survival is <1 year (5,6). Of note, the effectiveness of second-line chemotherapy is more limited (6). Therefore, novel biomarkers are required to select patients with endometrial cancer with poor prognosis at the time of biopsy or initial surgery.

Claudins (CLDNs) are tetraspan proteins with a short cytoplasmic N-terminus, two extracellular loops and a C-terminal cytoplasmic domain (7). CLDNs form tight junctions and are composed of >20 subtypes in humans. They function as a physical barrier or gate of small molecules (8-11) and as signaling platforms to coordinate diverse cellular behaviors (10-12). CLDNs are expressed in distinct patterns in different tissues and cells. Furthermore, CLDNs are useful cancer biomarkers, as they are frequently upregulated and are associated with malignant traits of cancers, such as invasion, migration, metastasis and chemoresistance (13,14).

Recent studies by our group demonstrated that aberrant CLDN6 expression is a biomarker for poor prognosis in endometrial cancer, and that abnormal CLDN6 signaling enhances malignant behaviors by AKT-dependent phosphorylation of estrogen receptor-α (ERα) through the Src-family kinases (SFK)/PI3K/AKT signaling pathway (15-17). Among CLDN subtypes, CLDN9 is the closest member to CLDN6 (18) and their genes are located adjacent to each other on the human genome. While several experimental studies determined that CLDN9 overexpression promotes cancer malignancy (19,20), endogenous expression of CLDN9 in cancer tissues has not been indicated at the protein level and its clinicopathological significance remains obscure due to the unavailability of selective antibodies.

In the present study, it was demonstrated that high CLDN9 expression predicts poor prognosis in patients with endometrial cancer using a newly established specific monoclonal antibody (mAb) (21). In addition, it was indicated that the combination of CLDN9 and CLDN6 is beneficial for predicting poor outcome in endometrial cancer.

Materials and methods

Cell culture, expression vectors, and transfection

293T and Ishikawa cells were gifted by Professor Suzuki, Fukushima Medical University (Fukushima, Japan) and Professor Yamada, Wakayama Medical University (Wakayama, Japan), respectively. They were grown in Dulbecco's Modified Eagle Medium (Sigma-Aldrich; Merck KGaA) with 10% fetal bovine serum (Sigma-Aldrich; Merck KGaA) and 1% penicillin-streptomycin-amphotericin B suspension (cat. no. 161-23181; Fujifilm).

The protein-coding regions of the human CLDN1, CLDN4, CLDN5, CLDN6 and CLDN9 genes were amplified from complementary or genomic DNA extracted from 293T cells with the PrimeSTAR GXL (cat. no. R050A; Clontech) PCR kit following the manufacturer's protocol. They were cloned into the NotI/BamHI site of the CSII-EF-MCS-IRES2-Venus plasmid (cat. no. RDB04384; RIKEN BioResource Center). For transient expression of the target genes (CLDN1, CLDN4, CLDN5, CLDN6 and CLDN9), 5×106 293T cells were transfected with 10 µg of the indicated vectors using 30 µg of Polyethyleneimine Max (PEI Max; Cosmo Bio Co., Ltd.) eight hours after passage. The pTagRFP-laminB1 vector (cat. no. FP370; Evrogen) was co-transfected with the CLDN9-expression vector to visualize the nuclei in successfully transfected cells. The transfection efficiency was evaluated by Venus expression with a fluorescence microscope (IX71; Olympus Corporation).

Antibodies

Rat monoclonal antibodies (mAbs) against the cytoplasmic tail of human CLDN6 and CLDN9 were generated using an iliac lymph node method, as previously described (16,21). Clone #15 for CLDN6 and clone 1D1 for CLDN9 were used in the present study. A mouse anti-p53 mAb (cat. no. OP43; clone Ab-6; Calbiochem; Merck KGaA) was used for evaluation of p53 expression.

Immunoblotting

Total cell lysates were prepared using CelLytic™ MT Cell Lysis Reagent (cat. no. C3228; Sigma-Aldrich; Merck KGaA). The protein concentration of the total cell lysates was measured by Pierce™ BCA Protein Assay Kit (cat. no. 23225; Thermo Fisher Scientific, Inc.) and 0.5 µg was loaded per lane for one-dimensional SDS-PAGE (12.5%). Subsequently, the protein bands were electrophoretically transferred onto an Immobilon membrane (MilliporeSigma). The membrane was blocked with PBS containing 4% skimmed milk (Morinaga) for 30 min and treated with the supernatant of the rat anti-CLDN9 hybridomaprima for 60 min at room temperature. After washing with PBS three times, the membrane was incubated with 1:2,000-fold diluted HRP-conjugated anti-rat IgG (cat. no. NA935V; GE Healthcare). The signals were detected by chemiluminescence (cat. no. WSE-7110EzWestLumi One; ATTO).

Immunofluorescence

Ishikawa cells (5.0×105) were grown on coverslips coated with Cellmatrix Type I-A (Nitta Gelatin). After 48 h, the samples were fixed in ice-cold ethanol for 10 min. After washing with PBS, they were preincubated in PBS containing 5% skimmed milk (Morinaga) at room temperature. They were subsequently incubated overnight at 4°C with the supernatant of anti-CLDN9 hybridoma, followed by washing with PBS three times and incubation with the secondary antibody, 1:400 diluted Alexa Fluor 488 AffiniPure Donkey Anti-Rat IgG antibody (cat. no. 712-545-150; Jackson ImmunoResearch) for 60 min at room temperature. After washing with PBS, the specimens were mounted with Fluoro-Gel II with DAPI (cat. no. 17985-51; Electron Microscopy Sciences). The samples were observed and images were acquired with a fluorescent microscope (IX71; Olympus Corporation).

Tissue collection, immunostaining and histological analysis

Formalin-fixed paraffin-embedded (FFPE) tissue sections were obtained from 248 patients with endometrial cancer [age, mean ± standard deviation (SD) of 58.2±11.5 years; range, 30-83 years] who underwent hysterectomy and bilateral-salpingo-oophorectomy and/or retroperitoneal lymphadenectomy between January 2003 and March 2015 at Fukushima Medical University Hospital (FMUH; Fukushima, Japan). The subjects were limited to patients who were confirmed to have at least 5-year outcomes and those who had died due to endometrial cancer and metastasis. Detailed information, including postoperative pathology diagnosis reports, age, stage [International Federation of Gynecology and Obstetrics (FIGO) 2008] (22), histological type, histological grade, Bokhman subtype (23), lymphovascular space involvement (LVSI), lymph node metastasis, distant metastasis, recurrence status, disease-specific survival (DSS) and disease-free survival (DFS), was obtained. The staging of patients encountered between January 2003 and December 2007 was modified in accordance with the FIGO 2008 system. Distant metastasis was judged by diagnostic imaging. Normal adult tissues, including the pituitary gland, cerebrum, liver, lung, and kidney, were collected from autopsy specimens dissected at FMUH between January 2013 and December 2014. Three to four specimens among six cases (a 29-year-old male, 42-year-old female, 51-year-old female, 57-year-old male, 65-year-old male and a 71-year-old female) were examined and a representative image was presented for each organ.

For immunostaining, the FFPE tissue blocks were sliced into 5-µm-thick sections, deparaffinized with xylene and rehydrated using a graded series of ethanol. The sections were then immersed in 0.3% hydrogen peroxide in methanol for 20 min at room temperature to block endogenous peroxidase activity. Antigen retrieval was performed by incubating the sections in boiling 10 mM citric acid buffer (pH 5.0) using a microwave for 10 min. After blocking with 5% skimmed milk (Morinaga) at room temperature for 30 min, the sections were incubated overnight at 4°C with supernatants of the rat anti-CLDN6 or CLDN9 hybridoma. After washing with PBS, a secondary antibody reaction was performed by using the Histofine Simple Stain mouse MAX-PO kit (cat. no. 414311; Nichirei Biosciences, Inc.) for 3′,3′-diaminobenzidine as a chromogen according to the manufacturer's instructions. Immunostaining for CLDN6 was performed as previously described (16). p53 was stained following the manufacturer's protocol.

Immunostaining results were interpreted by two independent pathologists and one gynecologist using a semiquantitative scoring system. The immunostaining reactions were evaluated according to signal intensity (SI; 0, negative; 1, weak; 2, moderate; 3, strong). The receiver operating characteristic (ROC) curve was plotted and analyzed (Fig. S1) to determine the optimal cut-off values of the SI for CLDN9 expression. CLDN6 expression was assessed by the immunoreactive score (IRS) as described previously (16). p53 mutation was assessed following the most accepted criteria (24).

Statistical analysis

The χ2 test was used to evaluate the relationship between CLDN9 expression and clinicopathological parameters such as age, stage, histological type, histological grade, Bokhman subtype, LVSI, lymph node metastasis, distant metastasis, DSS and DFS. Survival analysis was performed using the Kaplan-Meier method and differences between groups were analyzed using the log-rank test. Cox regression according to the univariate and multivariate model was used to identify predictors of survival. In addition, the expression of CLDN9 and CLDN6 was compared and statistical analysis was performed in a similar way. Two-tailed P-values of <0.05 were considered to indicate a statistically significant result. When comparing the disease-specific and disease-free survival among four groups, P<0.125 was used as the threshold for a statistically significant result to counteract the multiple comparisons problem by applying Bonferroni correction. All statistical analyses were performed using SPSS version 24.0 software (IBM Corporation).

Results

Verification of anti-CLDN9 mAb

First, the reactivity and specificity of the anti-CLDN9 mAb were tested against the C-terminal cytoplasmic region of human CLDN9 (Fig. 1A), which was recently established by our group (21). To this end, 293T cells were transiently transfected with individual CLDN expression vectors, followed by western blot and immunohistochemistry using whole-cell extracts and cell blocks, respectively. As presented in Fig. 1B and C, the anti-CLDN9 mAb appeared to specifically recognize CLDN9 but not CLDN1, CLDN4, CLDN5 or CLDN6, which are the four closest members to CLDN9 among the CLDN family. In addition, immunofluorescence staining using the anti-CLDN9 mAb revealed positive signals along cell-cell borders in the endometrial cancer cell line Ishikawa overexpressing CLDN9 (Fig. 1D). Furthermore, CLDN9 expression was detected in normal human pituitary gland (Fig. 1E), but not in cerebral, liver, lung or kidney tissue (Fig. S2).

Differential expression of CLDN9 among endometrial cancer subjects

Next, the expression of CLDN9 was evaluated by immunohistochemistry in endometrial cancer tissues resected from 248 patients, whose demographic and clinicopathological characteristics are presented in Table I. CLDN9 was mainly distributed along the cell membranes of endometrial carcinoma cells, but it was diffusely localized in certain cases (Fig. 2). The expression of CLDN9 was semi-quantified by determining the SI, as the percentage of positive cells was not >10% in most cases (mean ± SD, 6.30±6.84%). The SI varied among the subjects, which was 3 in 18 subjects (7.3%), 2 in 25 (10.1%), 1 in 46 (18.5%) and 0 in 159 (64.1%) (Fig. S3). Based on the ROC analysis, the samples were divided into two groups: Low CLDN9 expression (SI <2) and high CLDN9 expression (SI ≥2; Fig. S1).

Table I

Clinicopathological characteristics of patients with endometrial cancer (n=248).

Table I

Clinicopathological characteristics of patients with endometrial cancer (n=248).

ParameterValue
Age, years58.1±11.5 (30-83)
Stage
 I182 (73.4)
 II6 (2.4)
 III40 (16.1)
 IV20 (8.1)
Histological type
 Endometrioid carcinoma226 (91.1)
  Grade 1145 (58.5)
  Grade 240 (16.1)
  Grade 341 (16.5)
 Serous carcinoma7 (2.8)
 Clear carcinoma6 (2.4)
 Mucinous carcinoma2 (0.8)
 Other7 (2.8)

[i] Values are expressed as n (%) or the mean ± standard deviation (range).

High CLDN9 expression is an independent poor prognostic marker for endometrial cancer

Kaplan-Meier plots revealed significantly shorter DSS and DFS in the high CLDN9 expression group than in the low expression group (Fig. 3A and B). The 5-year DSS rates in the low and high CLDN9 expression groups were 87.8 and 62.8%, and the DFS rates were 84.9 and 62.8%, respectively.

Among the clinicopathological factors, high CLDN9 expression was significantly associated with non-endometrioid histology (P=0.021) and lymph node metastasis (P=0.012; Table II). By contrast, younger age (P=0.370), stage III/IV (P=0.072), histological grade 3 (P=0.431), histological type II (which includes endometrioid carcinoma grade 3, serous carcinoma and clear cell carcinoma; P=0.101), lymphovascular space involvement (LVSI; P=0.070) and distant metastasis (P=0.414) were not related to high CLDN9 expression.

Table II

Relationship between CLDN9 expression and clinicopathological factors in patients with endometrial cancer (n=248).

Table II

Relationship between CLDN9 expression and clinicopathological factors in patients with endometrial cancer (n=248).

ParameterTotalCLDN9-high (n=43)CLDN9-low (n=205)P-value
Age, years0.370
 <5053 (21.4)7 (16.3)46 (22.4)
 ≥50195 (78.6)36 (83.7)159 (77.6)
Stage0.072
 I/II188 (75.8)28 (65.1)160 (78.0)
 III/IV60 (24.2)15 (34.9)45 (22.0)
Histological type0.021a
 Endometrioid226 (91.1)35 (81.4)191 (93.1)
 Serous7 (2.8)1 (2.3)6 (2.9)
 Clear6 (2.4)4 (9.3)2 (1.0)
 Mucinous2 (0.8)0 (0.0)2 (1.0)
 Others7 (2.8)3 (7.0)4 (2.0)
Histological grade0.431
 1/2185 (74.6)27 (62.8)158 (77.1)
 341 (16.5)8 (18.6)33 (16.1)
 Other22 (8.9)8 (18.6)14 (6.8)
Histological classification0.101
 Type I185 (74.6)27 (62.8)158 (77.1)
 Type II54 (21.8)13 (30.2)41 (20.0)
 Other9 (3.6)3 (7.0)6 (2.9)
LVSI0.070
  (−)178 (71.8)26 (60.5)152 (74.1)
  (+)70 (28.2)17 (39.5)53 (25.9)
Nodal stage0.012
 N0203 (81.9)30 (69.7)173 (84.4)
 N134 (13.7)11 (25.6)23 (11.2)
 Unknown11 (4.4)2 (4.7)9 (4.4)
Metastasis stage0.414
 M0229 (92.3)41 (95.3)188 (91.7)
 M119 (7.7)2 (4.6)17 (8.3)

a Endometrioid vs. non-endometrioid. Values are expressed as n (%). LVSI, lymphovascular space involvement; CLDN, claudin.

In the univariate analysis, stage III/IV [hazard ratio (HR)=15.69, 95% confidence interval (CI) 7.47-32.96, P<0.001], endometrioid type [HR=0.35 (95% CI, 0.16-0.76), P=0.008], histological grade 3 [HR=4.02 (95% CI, 2.01-8.02), P<0.001], histological type II [HR=3.85 (95% CI, 2.02-7.35), P<0.001], LVSI [HR=8.99 (95% CI, 4.50-17.97), P<0.001], lymph node metastasis [HR=12.70 (95% CI, 6.37-25.32), P<0.001], distant metastasis [HR=14.25 (95% CI, 7.45-27.26), P<0.001] and high CLDN9 expression [HR=3.64 (95% CI, 1.94-6.81), P<0.001] were significant prognostic factors for DSS of patients with endometrial cancer (Table III). In addition, p53 abnormality was not associated with CLDN9 in histological type II cases (Table SI).

Table III

Univariate analysis of disease-specific survival in patients with endometrial cancer.

Table III

Univariate analysis of disease-specific survival in patients with endometrial cancer.

VariableHR95% CIP-value
Age ≥50 years2.640.94-7.390.066
Stage III/IV15.697.47-32.96<0.001
Endometrioid type0.350.16-0.760.008
Histological grade 34.022.01-8.02<0.001
Type II3.852.02-7.35<0.001
LVSI (+)8.994.50-17.97<0.001
N112.706.37-25.32<0.001
M114.257.45-27.26<0.001
CLDN9-high3.641.94-6.81<0.001

[i] HR, hazard ratio; CI, confidence interval; LVSI, lymphovascular space involvement; N1, positive for lymph node metastasis; M1, positive for distant metastasis.

Subsequently, Cox multivariate analysis was performed to determine the independent predictors of survival. Among the variables analyzed, stage III/IV [HR=6.00 (95% CI, 1.94-18.56), P=0.002], LVSI [HR=3.34 (95% CI, 1.21-9.25), P=0.020], distant metastasis [HR=6.74 (95% CI, 2.32-19.57), P<0.001] and high CLDN9 expression [HR=4.99 (95% CI, 1.96-12.70), P<0.001] were independent prognostic factors for DSS of patients with endometrial cancer (Table IV). By contrast, older age, endometrioid type, histological grade 3, histological type II and lymph node metastasis were no independent prognostic variables for them.

Table IV

Multivariate analysis of disease-specific survival in patients with endometrial cancer.

Table IV

Multivariate analysis of disease-specific survival in patients with endometrial cancer.

VariableHR95% CIP-value
Stage III/IV6.001.94-18.560.002
LVSI (+)3.341.21-9.250.020
M16.742.32-19.57<0.001
CLDN9-high4.991.96-12.70<0.001

[i] HR, hazard ratio; CI, confidence interval; LVSI, lymphovascular space involvement; M1, positive for distant metastasis; CLDN, claudin.

CLDN9 expression correlates with CLDN6 expression in endometrial cancer

CLDN9 largely shares its alignment with CLDN6 (18) and its genetic locus is adjacent to CLDN6 (Fig. 4A and B). Furthermore, none of the two genes are primarily expressed in adult tissues, except for the inner ear, olfactory epithelium and anterior pituitary glands (21,25), suggesting that they are regulated by similar mechanisms. Thus, it was hypothesized that upregulation of CLDN6 and CLDN9 may be mutually correlated in endometrial cancer tissues. To compare the expression of CLDN9 and CLDN6, the 248 patients with endometrial cancer were classified into four groups according to the expression levels of CLDN9 and CLDN6 (Fig. 4C). In the high CLDN6 expression group (18 cases), 10 (55.6%) and 8 (44.4%) had high and low CLDN9 expression, respectively, whereas in the low CLDN6 expression group (n=230), 33 (14.3%) and 197 (85.7%) cases had high and low CLDN9 expression, respectively. It was determined that high expression of CLDN9 in endometrial cancer cases was significantly associated with high CLDN6 expression (P<0.001; Table V), although CLDN6 and CLDN9 were principally expressed in different tumor cells within endometrial cancer tissues (Fig. 4C, panels of CLDN6-high/CLDN9-high). Of note, unlike CLDN9, p53 abnormality was positively associated with CLDN6 in histological type II cases (Table SII).

Table V

Association between CLDN6 and CLDN9 expression in patients with endometrial cancer.

Table V

Association between CLDN6 and CLDN9 expression in patients with endometrial cancer.

CLDN6CLDN9
P-value
LowHigh
Low197 (79.4)33 (13.3)<0.001
High8 (3.2)10 (4.0)

[i] Values are expressed as n (%). CLDN, claudin.

Combination of CLDN6 and CLDN9 expression is advantageous in identifying patients with endometrial cancer with poor prognosis

Kaplan-Meier plots revealed that DSS and DFS in the high CLDN6 expression group were significantly lower than those in the low CLDN6 expression group regardless of the CLDN9 expression levels (Fig. 5). Furthermore, there were significant differences in both DSS and DFS between the subjects with CLDN6-low/CLDN9-low and those with CLDN6-low/CLDN9-high. Their 5-year DSS rate was 89.8% in the former and 72.7% in the latter group.

In the univariate analysis specifically for patients with endometrial cancer with low CLDN6 expression, stage III/IV [HR=18.42 (95% CI, 7.49-45.38), P<0.001], endometrioid type [HR=0.33 (95% CI, 0.13-0.86), P=0.023], histological grade 3 [HR=4.06 (95% CI, 1.80-9.14), P<0.001], histological type II [HR=3.82 (95% CI, 1.77-8.26), P<0.001], LVSI [HR=9.70 (95% CI, 4.29-21.93), P<0.001], lymph node metastasis [HR=16.47 (95% CI, 7.08-38.29), P<0.001], distant metastasis [HR=16.51 (95% CI, 7.71-35.37), P<0.001] and high CLDN9 expression [HR=2.98 (95% CI, 1.36-6.55), P=0.007] were significant prognostic variables for DSS. However, older age was not a prognostic factor for endometrial cancer with low CLDN6 expression (Table VI).

Table VI

Cox univariate analysis of disease-specific survival in patients with endometrial cancer with low CLDN6 expression.

Table VI

Cox univariate analysis of disease-specific survival in patients with endometrial cancer with low CLDN6 expression.

VariableHR95% CIP-value
Age ≥50 years1.900.66-5.450.201
Stage III/IV18.427.49-45.38<0.001
Endometrioid type0.330.13-0.860.023
Histological grade 34.061.80-9.14<0.001
Type II3.821.77-8.26<0.001
LVSI (+)9.704.29-21.93<0.001
N116.477.08-38.29<0.001
M116.517.71-35.37<0.001
CLDN9-high2.981.36-6.550.007

[i] HR, hazard ratio; CI, confidence interval; LVSI, lymphovascular space involvement; N1, positive for lymph node metastasis; M1, positive for distant metastasis; CLDN, claudin.

Discussion

The immunohistochemical analysis of the present study using the specific anti-CLDN9 mAb revealed that CLDN9 is differentially expressed in endometrial cancer tissues. For instance, the SI varied among the subjects with endometrial cancer and 43 in the 248 cases (17.3%) had high CLDN9 expression. In addition, there was a variation in the percentage of CLDN9-positive cells among the cases. Of note, similar heterogeneity was also observed in CLDN6 expression in endometrial cancer tissues (16).

Among the CLDN family, CLDN1 and CLDN2 are prone to be expressed in histological type I and II endometrial cancers, respectively, although the prognostic value remains to be determined (26,27). The present study demonstrated that high CLDN9 expression represents a poor prognostic marker for endometrial cancer. This conclusion was drawn from the following results: i) The DSS and DFS in the high CLDN9 expression group of endometrial cancer subjects were significantly lower compared with those in the low CLDN9 expression group; ii) high CLDN9 expression was significantly associated with adverse clinicopathological factors, including non-endometrioid type and lymph node metastasis; iii) univariate analysis indicated that high CLDN9 expression was a significant prognostic factor (HR=3.64); and iv) upon multivariate analysis, high CLDN9 expression appeared to be an independent prognostic factor for DSS of the endometrial cancer subjects (HR=4.99). Thus, not only aberrant CLDN6 expression (16) but also high CLDN9 expression predicts a poor outcome for endometrial cancer. The high CLDN9 expression significantly correlated with the high CLDN6 expression in endometrial cancer, further supporting this conclusion.

Recently, endometrial cancer was classified into four molecular groups based on genetic characteristics: Ultramutated, hypermutated, copy-number low and copy-number high (28,29). More recently, simplified classifications have also been proposed for broader clinical applications (30-32). However, these genetic classifications are not applicable for all cases mainly due to insufficient tumor cell content. In addition, protein expression does not necessarily correlate with gene mutations or mRNA expression (33-35). Therefore, a classification based on protein signals in FFPE specimens, which directly reflects molecular characteristics, is valuable. In the present study, it was demonstrated that the protein expression of CLDN9 and CLDN6 in FFPE specimens reflected the prognosis of patients with endometrial cancer. In more detail, the CLDN6-high group of patients with endometrial cancer exhibited markedly lower DSS and DFS irrespective of the expression levels of CLDN9. In addition, CLDN6-low/CLDN9-high cases of endometrial cancer had significantly decreased DSS and DFS compared with CLDN6-low/CLDN9-low cases. Furthermore, while the proportion of CLDN6-high cases was <10%, which was also in line with a previous study (16), nearly a quarter of cases had high expression of either CLDN6 or CLDN9. Thus, classification depending on the expression of CLDN6 and CLDN9 (CLDN6-high, CLDN6-low/CLDN9-high and CLDN6-low/CLDN9-low) would be beneficial in the aspects of easy use and broad clinical applications. In addition, abnormal expression of p53 was not associated with CLDN9-high, indicating that CLDN9-high predicts poor prognosis regardless of p53 mutation.

It remains elusive by which mechanisms high CLDN9 expression results in poor outcome for patients with endometrial cancer. However, it was previously reported that aberrant CLDN6 expression promotes endometrial cancer progression in vitro and in vivo via hijacking the CLDN6/SFK/PI3K/AKT/ERα pathway (12,17). For instance, abnormal CLDN6 signaling stimulates not only cell proliferation but also collective cell migration in the leading front of endometrial cancer cells. Since CLDN6 recruits and activates SFKs in second extracellular domain-dependent and Y196/200-dependent manners, both of which are highly conserved in CLDN9 (12), CLDN9 may similarly act as a cancer promoter. Alternatively, since CLDN2 activates Yes-associated protein (YAP) and stimulates self-renewal of human colorectal cancer stem-like cells (36), CLDN9 may also promote YAP activity to advance endometrial cancer progression.

It is premature to discuss how the expression of CLDN6 and CLDN9 is upregulated in endometrial cancer cells. However, a previous study by our group demonstrated that the CLDN6 signaling ligand independently activates the expression of endogenous CLDN6 gene in F9 embryonal carcinoma cells (15). Therefore, the positive loop of the CLDN6/SFK/AKT/RARγ cascade may contribute to inducing and maintaining CLDN6 expression in endometrial cancer cells. As the second extracellular domain and Y200 in CLDN6 are conserved in CLDN9 as described above, the expression of CLDN9 may be upregulated by a similar positive feedback mechanism.

As CLDNs are distributed on the cell surface, they are good druggable targets of antibody therapy. Indeed, antibody therapy against CLDN6, including CAR-T and antibody-drug conjugates, exhibits efficiency in solid tumors (37-39). Since CLDN9 has almost identical extracellular domains to those of CLDN6, it appears feasible to obtain a bispecific antibody targeting CLDN6 and CLDN9 together. Furthermore, CLDN9 and CLDN6 are rarely expressed in normal adult tissues, excluding the cochlea (25), olfactory epithelia and anterior pituitary glands (21), implying that the antibody therapy may be less toxic to non-tumor tissues. Therefore, taken together with the present findings that CLDN6 and CLDN9 are poor prognostic factors, antibody therapy targeting CLDN9 and CLDN6 would be promising for endometrial cancer treatment.

In conclusion, the present study demonstrated that upregulation of CLDN9 expression, determined using a selective mAb against CLDN9, predicted poor prognosis for patients with endometrial cancer. It is required to unveil the molecular mechanisms of CLDN9 signals in endometrial cancer to develop a new personalized medicine targeting CLDN9.

Supplementary Data

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

YE, KS, MakK, YK, ManK, AYH and TH performed experiments. YE, KS and HC drafted the manuscript. KS and HC conceived the study and supervised all experiments. KS, YE, YH and HC reviewed the pathological diagnosis and/or analyzed immunohistochemistry slides. SF, SS, TW and KF collected specimens and assembled the database. YH, KF and HC helped with the writing of the manuscript. YE, KS and HC confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent for publication

The study was approved by the Ethics Committee of Fukushima Medical University (Fukushima, Japan; approval code, 2019-311; approval date, Mar 18, 2020). The research was conducted in accordance with the 1964 Helsinki Declaration or comparable standards. This study was carried out by an opt-out method. Since it was conducted as a retrospective study using cases with a follow-up period of more than five years, the patients had already died or stopped visiting the hospital. The experimental protocol has been disclosed on the website and the patients or their representatives were able to decline to participate in the survey as they preferred.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors thank Ms. Seiko Watanabe and Ms. Keiko Watari, Fukushima Medical University for their technical assistance and the Scientific English Editing Section of Fukushima Medical University (Fukushima, Japan) for their help with the manuscript.

Funding

This work was supported by JSPS KAKENHI (grant nos. 19K16615, 20K18224 and 20K16176) and by the Takeda Science Foundation.

Abbreviations:

CI

confidence interval

CLDN

claudin

DFS

disease-free survival

DSS

disease-specific survival

ERα

estrogen receptor α

FFPE

formalin-fixed paraffin-embedded

FIGO

International Federation of Gynecology and Obstetrics

HR

hazard ratio

IRS

immunoreactive score

LVSI

lymphovascular space involvement

mAb

monoclonal antibody

RARγ

retinoic acid receptor γ

SI

signal intensity

SFK

Src-family kinase

SD

standard deviation

YAP

Yes-associated protein

References

1 

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, Forman D and Bray F: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer. 49:1374–1403. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Henley SJ, Ward EM, Scott S, Ma J, Anderson RN, Firth AU, Thomas CC, Islami F, Weir HK, Lewis DR, et al: Annual report to the nation on the status of cancer, part I: National cancer statistics. Cancer. 126:2225–2249. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Lu KH and Broaddus RR: Endometrial cancer. N Engl J Med. 383:2053–2064. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Aoki Y, Kanao H, Wang X, Yunokawa M, Omatsu K, Fusegi A and Takeshima N: Adjuvant treatment of endometrial cancer today. Jpn J Clin Oncol. 50:753–765. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Bestvina CM and Fleming GF: Chemotherapy for endometrial cancer in adjuvant and advanced disease settings. Oncologist. 21:1250–1259. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Furuse M, Fujita K, Hiiragi T, Fujimoto K and Tsukita S: Claudin-1 and -2: Novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol. 141:1539–1550. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Furuse M and Tsukita S: Claudins in occluding junctions of humans and flies. Trends Cell Biol. 16:181–188. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Van Itallie CM and Anderson JM: Claudins and epithelial paracellular transport. Annu Rev Physiol. 68:403–429. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Chiba H, Osanai M, Murata M, Kojima T and Sawada N: Transmembrane proteins of tight junctions. Biochim Biophys Acta. 1778:588–600. 2008. View Article : Google Scholar

11 

Zihni C, Mills C, Matter K and Balda MS: Tight junctions: From simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol. 17:564–580. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Sugimoto K and Chiba H: The claudin-transcription factor signaling pathway. Tissue Barriers. 9:19081092021. View Article : Google Scholar : PubMed/NCBI

13 

Li J: Context-dependent roles of claudins in tumorigenesis. Front Oncol. 11:6767812021. View Article : Google Scholar : PubMed/NCBI

14 

Tabariès S and Siegel PM: The role of claudins in cancer metastasis. Oncogene. 36:1176–1190. 2017. View Article : Google Scholar

15 

Sugimoto K, Ichikawa-Tomikawa N, Kashiwagi K, Endo C, Tanaka S, Sawada N, Watabe T, Higashi T and Chiba H: Cell adhesion signals regulate the nuclear receptor activity. Proc Natl Acad Sci USA. 116:24600–24609. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Kojima M, Sugimoto K, Tanaka M, Endo Y, Kato H, Honda T, Furukawa S, Nishiyama H, Watanabe T, Soeda S, et al: Prognostic significance of aberrant claudin-6 expression in endometrial cancer. Cancers (Basel). 12:27482020. View Article : Google Scholar :

17 

Kojima M, Sugimoto K, Kobayashi M, Ichikawa-Tomikawa N, Kashiwagi K, Watanabe T, Soeda S, Fujimori K and Chiba H: Aberrant claudin-6-adhesion signaling promotes endometrial cancer progression via estrogen receptor α. Mol Cancer Res. 19:1208–1220. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Lal-Nag M and Morin PJ: The claudins. Genome Biol. 10:2352009. View Article : Google Scholar : PubMed/NCBI

19 

Sharma RK, Chheda ZS, Das Purkayastha BP, Gomez-Gutierrez JG, Jala VR and Haribabu B: A spontaneous metastasis model reveals the significance of claudin-9 overexpression in lung cancer metastasis. Clin Exp Metastasis. 33:263–275. 2016. View Article : Google Scholar

20 

Liu H, Wang M, Liang N and Guan L: Claudin-9 enhances the metastatic potential of hepatocytes via Tyk2/Stat3 signaling. Turk J Gastroenterol. 30:722–731. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Higashi AY, Higashi T, Furuse K, Ozeki K, Furuse M and Chiba H: Claudin-9 constitutes tight junctions of folliculostellate cells in the anterior pituitary gland. Sci Rep. 11:216422021. View Article : Google Scholar

22 

Mutch DG and Prat J: 2014 FIGO staging for ovarian, fallopian tube and peritoneal cancer. Gynecol Oncol. 133:401–404. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Conlon N, Da Cruz Paula A, Ashley CW, Segura S, De Brot L, da Silva EM, Soslow RA, Weigelt B and DeLair DF: Endometrial carcinomas with a 'serous' component in young women are enriched for DNA mismatch repair deficiency, lynch syndrome, and POLE exonuclease domain mutations. Am J Surg Pathol. 44:641–648. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Köbel M, Ronnett BM, Singh N, Soslow RA, Gilks CB and McCluggage WG: Interpretation of P53 immunohistochemistry in endometrial carcinomas: Toward increased reproducibility. Int J Gynecol Pathol. 38(Suppl 1): S123–S131. 2019. View Article : Google Scholar

25 

Nakano Y, Kim SH, Kim HM, Sanneman JD, Zhang Y, Smith RJH, Marcus DC, Wangemann P, Nessler RA and Bánfi B: A claudin-9-based ion permeability barrier is essential for hearing. PLoS Genet. 5:e10006102009. View Article : Google Scholar : PubMed/NCBI

26 

Sobel G, Németh J, Kiss A, Lotz G, Szabó I, Udvarhelyi N, Schaff Z and Páska C: Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol. 103:591–598. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Szabó I, Kiss A, Schaff Z and Sobel G: Claudins as diagnostic and prognostic markers in gynecological cancer. Histol Histopathol. 24:1607–1615. 2009.PubMed/NCBI

28 

Urick ME and Bell DW: Clinical actionability of molecular targets in endometrial cancer. Nat Rev Cancer. 19:510–521. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Cancer Genome Atlas Research Network; Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, et al: Integrated genomic characterization of endometrial carcinoma. Nature. 497:67–73. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Stelloo E, Nout RA, Osse EM, Jürgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC, van der Steen-Banasik EM, Nijman HW, Putter H, Bosse T, et al: Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts. Clin Cancer Res. 22:4215–4224. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Kommoss S, McConechy MK, Kommoss F, Leung S, Bunz A, Magrill J, Britton H, Kommoss F, Grevenkamp F, Karnezis A, et al: Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Ann Oncol. 29:1180–1188. 2018. View Article : Google Scholar : PubMed/NCBI

32 

León-Castillo A, de Boer SM, Powell ME, Mileshkin LR, Mackay HJ, Leary A, Nijman HW, Singh N, Pollock PM, Bessette P, et al: Molecular classification of the PORTEC-3 trial for high-risk endometrial cancer: Impact on prognosis and benefit from adjuvant therapy. J Clin Oncol. 38:3388–3397. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Vogel C and Marcotte EM: Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 13:227–232. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Edfors F, Danielsson F, Hallström BM, Käll L, Lundberg E, Pontén F, Forsström B and Uhlén M: Gene-specific correlation of RNA and protein levels in human cells and tissues. Mol Syst Biol. 12:8832016. View Article : Google Scholar : PubMed/NCBI

35 

Fortelny N, Overall CM, Pavlidis P and Freue GVC: Can we predict protein from mRNA levels? Nature. 547:E19–E20. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Paquet-Fifield S, Koh SL, Cheng L, Beyit LM, Shembrey C, Mølck C, Behrenbruch C, Papin M, Gironella M, Guelfi S, et al: Tight junction protein claudin-2 promotes self-renewal of human colorectal cancer stem-like cells. Cancer Res. 78:2925–2938. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Reinhard K, Rengstl B, Oehm P, Michel K, Billmeier A, Hayduk N, Klein O, Kuna K, Ouchan Y, Wöll S, et al: An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 367:446–453. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Kong FE, Li GM, Tang YQ, Xi SY, Loong JHC, Li MM, Li HL, Cheng W, Zhu WJ, Mo JQ, et al: Targeting tumor lineage plasticity in hepatocellular carcinoma using an anti-CLDN6 antibody-drug conjugate. Sci Transl Med. 13:eabb62822021. View Article : Google Scholar : PubMed/NCBI

39 

Matsuzaki J, Lele S, Odunsi K and Tsuji T: Identification of Claudin 6-specific HLA class I- and HLA class II-restricted T cell receptors for cellular immunotherapy in ovarian cancer. Oncoimmunology. 11:20209832022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 61 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Endo Y, Sugimoto K, Kobayashi M, Kobayashi Y, Kojima M, Furukawa S, Soeda S, Watanabe T, Higashi AY, Higashi T, Higashi T, et al: Claudin‑9 is a novel prognostic biomarker for endometrial cancer. Int J Oncol 61: 135, 2022
APA
Endo, Y., Sugimoto, K., Kobayashi, M., Kobayashi, Y., Kojima, M., Furukawa, S. ... Chiba, H. (2022). Claudin‑9 is a novel prognostic biomarker for endometrial cancer. International Journal of Oncology, 61, 135. https://doi.org/10.3892/ijo.2022.5425
MLA
Endo, Y., Sugimoto, K., Kobayashi, M., Kobayashi, Y., Kojima, M., Furukawa, S., Soeda, S., Watanabe, T., Higashi, A. Y., Higashi, T., Hashimoto, Y., Fujimori, K., Chiba, H."Claudin‑9 is a novel prognostic biomarker for endometrial cancer". International Journal of Oncology 61.5 (2022): 135.
Chicago
Endo, Y., Sugimoto, K., Kobayashi, M., Kobayashi, Y., Kojima, M., Furukawa, S., Soeda, S., Watanabe, T., Higashi, A. Y., Higashi, T., Hashimoto, Y., Fujimori, K., Chiba, H."Claudin‑9 is a novel prognostic biomarker for endometrial cancer". International Journal of Oncology 61, no. 5 (2022): 135. https://doi.org/10.3892/ijo.2022.5425