Gene expression profile analysis of lung cancers revealed the transactivation of an immunoglobulin-like molecule Nectin-4 in the majority of non–small cell lung cancers (NSCLC). Immunohistochemical staining of 422 NSCLCs showed that a high level of Nectin-4 expression was associated with poor prognosis for NSCLC patients (P < 0.0001), and multivariate analysis confirmed its independent prognostic value (P < 0.0001). We established an ELISA to measure serum Nectin-4 and found that serum Nectin-4 levels were significantly higher in NSCLC patients than in healthy volunteers. The proportion of the serum Nectin-4–positive cases was 88 of 164 (53.7%) NSCLCs, whereas only 3 of 131 (2.3%) healthy volunteers were falsely diagnosed as positive, which was superior to carcinoembryonic antigen (CEA) and cytokeratin 19-fragment (CYFRA21-1) in sensitivity and specificity. A combined ELISA for both Nectin-4 and CEA increased sensitivity and classified 65.0% of lung adenocarcinomas as positive with false-positive rate of 4.6%. The use of both Nectin-4 and CYFRA21-1 classified 68.3% of lung squamous cell carcinomas as positive with false-positive rate of 6.1%. Treatment of lung cancer cells with small interfering RNAs against Nectin-4 suppressed its expression and cell growth. In addition, exogenous expression of Nectin-4 increased the lamellipodia formation and the invasive ability of mammalian cells through activation of small GTPase Rac1. Nectin-4 might play a significant role in lung carcinogenesis, and it should be a new candidate serum and tissue biomarker, as well as a therapeutic target. [Cancer Res 2009;69(16):6694–703]

Lung cancer is the leading cause of cancer death worldwide (1). About 30% of patients who are diagnosed to have non–small cell lung cancer (NSCLC) are able to undergo curative resection, whereas the remaining patients with an advanced disease are mainly treated with chemotherapy alone or in combination with a local treatment modality (2). In fact, the overall 5-year survival rate of NSCLC patients still remains at only 15% (3, 4). A number of targeted therapies were shown to be effective for a subset of advanced NSCLC patients (59). However, issues of toxicity limit these treatment regimens to selected patients.

To identify new molecules involved in pathways of carcinogenesis, we had performed genome-wide expression profile analysis of 101 lung cancers with the cDNA microarray containing 27,648 genes and expressed sequence tags (ESTs) (1015). To verify the biological and clinicopathologic significance of the respective gene products, we have been performing tumor-tissue microarray analysis of clinical lung cancer materials as well as RNA interference assays (1637). This systematic approach revealed that Nectin-4 (alias poliovirus receptor–related 4, PVRL4) was transactivated in the majority of NSCLCs.

The Nectin family is the Ca2+-independent immunoglobulin-like molecules consisting of four members (Nectin-1, Nectin-2, Nectin-3, and Nectin-4), which are supposed to trans-interact homophilically and heterophilically, and play a role in cell-cell adhesion (38). Nectins 1, 2, and 3 are widely expressed in adult tissues, but Nectin-4 was expressed specifically in the embryo and placenta (39, 40). Recently, Nectin-4 was indicated to be overexpressed in breast carcinoma and have a soluble form that is released into the blood (41, 42). In spite of the recent evidence of Nectin-4 overexpression in cancer, the biological significance of Nectin-4 activation in human cancer progression and its clinical potential as a therapeutic target were not fully described.

Here, we report that Nectin-4 plays a significant role in cancer cell growth and invasion, and could be a potential target for the development of therapeutic agents as well as a novel serum and tissue biomarker for lung cancer.

Clinical tissue samples. Primary lung cancer samples as well as their corresponding normal tissues adjacent to resection margins from patients, who had no anticancer treatment before tumor resection, had been obtained earlier with informed consent (11). The histologic classification of the tumor specimens was based on WHO criteria (43). All tumors were staged on the basis of the pTNM pathologic classification of the International Union Against Cancer (Table 1A; ref. 44). A total of 422 formalin-fixed samples of primary NSCLCs including 265 adenocarcinomas (ADC), 116 squamous cell carcinomas (SCC), 28 large cell carcinomas, 13 adenosquamous cell carcinomas and adjacent normal lung tissues, had been obtained earlier along with clinicopathologic data from patients who had undergone surgery at Saitama Cancer Center (Saitama, Japan). This study and the use of all clinical materials were approved by the individual Institutional Research Ethics Committees.

Table 1.
A. Association between Nectin-4 positivity in NSCLC tissues and patients' characteristics (n = 422)
Total
Nectin-4 strong positive
Nectin-4 weak positive
Nectin-4 absent
P: strong vs weak/absent
n = 422n = 245n = 119n = 58
Gender      
    Male 290 172 76 42 0.4577 
    Female 132 73 43 16  
Age (y)      
    <65 218 126 63 29 0.9217 
    ≥65 204 119 56 29  
Histologic type      
    ADC 265 167 68 30 0.0059*, 
    SCC 116 54 41 21  
    Others 41 24 10  
pT factor      
    T1 143 69 51 23 0.0048 
    T2–T4 279 176 68 35  
pN factor      
    N0 259 142 81 36 0.105 
    N1+N2 163 103 38 22  
Smoking      
    Nonsmoker 127 70 40 17 0.4523 
    Smoker 295 175 79 41  
     
B. Cox's proportional hazards model analysis of prognostic factors in patients with NSCLC
 
    
Variables
 
Hazards ratio
 
95% CI
 
Unfavorable/favorable
 
P
 
Univariate analysis     
    Nectin-4 2.116 1.551–2.887 Strong (2+)/Weak (1+) or (−) <0.0001 
    Age (y) 1.473 1.107–1.962 ≥65/< 65 0.0079 
    Gender 1.738 1.247–2.422 Male/Female 0.0011 
    Histologic type 1.402 1.053–1.867 Non-ADC/ADC 0.0208 
    pT factor 2.660 1.857–3.810 T2–T4/T1 <0.0001 
    pN factor 2.393 1.799–3.181 N1+N2/N0 <0.0001 
    Smoking 1.248 0.907–1.716 Smoker/nonsmoker 0.1734 
Multivariate analysis     
    Nectin-4 2.145 1.558–2.954 Strong (2+)/Weak (1+) or (−) <0.0001 
    Age (y) 1.679 1.250–2.254 ≥65/< 65 0.0006 
    Gender 1.437 0.996–2.072 Male/Female 0.0522 
    Histologic type 1.144 0.828–1.580 Non-ADC/ADC 0.4158 
    pT factor 1.916 1.319–2.785 T2–T4/T1 0.0006 
    pN factor 2.324 1.728–3.124 N1+N2/N0 <0.0001 
A. Association between Nectin-4 positivity in NSCLC tissues and patients' characteristics (n = 422)
Total
Nectin-4 strong positive
Nectin-4 weak positive
Nectin-4 absent
P: strong vs weak/absent
n = 422n = 245n = 119n = 58
Gender      
    Male 290 172 76 42 0.4577 
    Female 132 73 43 16  
Age (y)      
    <65 218 126 63 29 0.9217 
    ≥65 204 119 56 29  
Histologic type      
    ADC 265 167 68 30 0.0059*, 
    SCC 116 54 41 21  
    Others 41 24 10  
pT factor      
    T1 143 69 51 23 0.0048 
    T2–T4 279 176 68 35  
pN factor      
    N0 259 142 81 36 0.105 
    N1+N2 163 103 38 22  
Smoking      
    Nonsmoker 127 70 40 17 0.4523 
    Smoker 295 175 79 41  
     
B. Cox's proportional hazards model analysis of prognostic factors in patients with NSCLC
 
    
Variables
 
Hazards ratio
 
95% CI
 
Unfavorable/favorable
 
P
 
Univariate analysis     
    Nectin-4 2.116 1.551–2.887 Strong (2+)/Weak (1+) or (−) <0.0001 
    Age (y) 1.473 1.107–1.962 ≥65/< 65 0.0079 
    Gender 1.738 1.247–2.422 Male/Female 0.0011 
    Histologic type 1.402 1.053–1.867 Non-ADC/ADC 0.0208 
    pT factor 2.660 1.857–3.810 T2–T4/T1 <0.0001 
    pN factor 2.393 1.799–3.181 N1+N2/N0 <0.0001 
    Smoking 1.248 0.907–1.716 Smoker/nonsmoker 0.1734 
Multivariate analysis     
    Nectin-4 2.145 1.558–2.954 Strong (2+)/Weak (1+) or (−) <0.0001 
    Age (y) 1.679 1.250–2.254 ≥65/< 65 0.0006 
    Gender 1.437 0.996–2.072 Male/Female 0.0522 
    Histologic type 1.144 0.828–1.580 Non-ADC/ADC 0.4158 
    pT factor 1.916 1.319–2.785 T2–T4/T1 0.0006 
    pN factor 2.324 1.728–3.124 N1+N2/N0 <0.0001 

NOTE: Others, large-cell carcinoma (LCC) plus adenosquamous-cell carcinoma (ASC).

*

ADC vs non-ADC.

P < 0.05 (Fisher's exact test).

P < 0.05.

Serum samples. Serum samples were obtained with informed consent from 131 healthy volunteers as controls [88 males and 43 females; median age (±1 SD), 55.5 ± 9.6 y; range, 31–83; Supplementary Table S1B] and from 86 nonneoplastic lung disease patients with chronic obstructive pulmonary disease (COPD) enrolled as a part of the Japanese Project for Personalized Medicine (BioBank Japan) or admitted to Hiroshima University and its affiliated Hospitals (76 males and 10 females; median age (±1 SD), 66.4 ± 5.6 y; range, 54–73; Supplementary Table S1C). All of these COPD patients were current and/or former smokers [the mean (±1 SD) of pack-year index was 66.7 ± 44.4; pack-year index was defined as the number of cigarette packs (20 cigarette per pack) consumed a day multiplied by years]. Serum samples were obtained with informed consent from 164 NSCLC patients (123 ADCs and 41 SCCs) admitted to Hiroshima University Hospital, as well as Kanagawa Cancer Center Hospital [122 males and 42 females; median age (±1 SD), 64.5 ± 10.4 y; range, 30–85; Table 2]. To investigate the prognostic value of serum Nectin-4, two additional sets of serum samples with precise follow-up record after treatments were also obtained. Group 1: serum samples obtained with informed consent before curative surgical resection at Kanagawa Cancer Center Hospital from 95 early stage NSCLC patients [stage I, 48 males and 47 females; median age (±1 SD), 66.8 ± 9.6 y; range, 38–84]. Group 2: serum samples obtained at diagnosis with informed consent from 62 advanced stage NSCLC patients (stage IIIB–IV) treated later with an identical protocol of the first-line chemotherapy using both carboplatin and paclitaxel admitted to Hiroshima University Hospital [46 males and 16 females; median age (±1 SD), 61.7 ± 10.2 y; range, 35–79]. Patient samples were selected for the study on the basis of the following criteria: (a) patients were newly diagnosed and previously untreated and (b) their tumors were pathologically diagnosed as NSCLCs (stages I–IV). Serum was obtained at the time of diagnosis and stored at −150°C.

Table 2.

Association between serum Nectin4 positivity and NSCLC patients' characteristics (n = 164)

Total
Serum Nectin-4 positive
Serum Nectin-4 absent
P: positive vs absent
n = 164n = 88n = 76
Gender     
    Male 122 69 53 0.215 
    Female 42 19 23  
Age (y)     
    <65 74 34 40 0.0845 
    ≥65 90 54 36  
Histologic type     
    ADC 123 66 57 >0.9999 
    SCC 41 22 19  
pT factor     
    T1 35 11 24 0.0039* 
    T2–T4 129 77 52  
pN factor     
    N0 50 21 29 0.0612 
    N1–N3 114 67 47  
Smoking     
    Nonsmoker 50 24 26 0.3959 
    Smoker 114 64 50  
Total
Serum Nectin-4 positive
Serum Nectin-4 absent
P: positive vs absent
n = 164n = 88n = 76
Gender     
    Male 122 69 53 0.215 
    Female 42 19 23  
Age (y)     
    <65 74 34 40 0.0845 
    ≥65 90 54 36  
Histologic type     
    ADC 123 66 57 >0.9999 
    SCC 41 22 19  
pT factor     
    T1 35 11 24 0.0039* 
    T2–T4 129 77 52  
pN factor     
    N0 50 21 29 0.0612 
    N1–N3 114 67 47  
Smoking     
    Nonsmoker 50 24 26 0.3959 
    Smoker 114 64 50  
*

P < 0.05 (Fisher's exact test).

Generation of murine anti–Nectin-4 monoclonal antibody. The anti–Nectin-4 monoclonal antibodies (clones 19–33 and 66–97) were produced using standard method after successive i.p. injections of 20 μg soluble recombinant protein (Nectin-4 ectodomain) to mice.

Flow cytometry and immunocytochemical analyses. Flow cytometric and immunocytochemical analyses were performed using anti–Nectin-4 antibody (clone 19–33), as previously described (17).

Immunohistochemistry and tissue microarray. Tumor-tissue microarrays were constructed using 422 formalin-fixed primary NSCLCs as described elsewhere (4547). To investigate the status of the Nectin-4 protein in clinical lung cancer samples that had been embedded in paraffin blocks, we stained the sections using 16.25 μg/mL of mouse anti–Nectin-4 antibody (clone 19–33) as previously described (34). Because the intensity of staining within each tumor tissue core was mostly homogenous, the intensity of Nectin-4 staining was semiquantitatively evaluated by three independent investigators without prior knowledge of clinicopathologic data using following criteria: strong positive (scored as 2+), dark brown staining in >50% of tumor cells completely obscuring plasma membrane and cytoplasm; weak positive (1+), any lesser degree of brown staining appreciable in tumor cell plasma membrane and cytoplasm; absent (scored as 0), no appreciable staining in tumor cells. Cases were defined as strongly positive if all of the three reviewers independently classified them as such.

ELISA. Serum levels of Nectin-4 were measured by sandwich-type ELISA that was originally developed using mouse anti–human Nectin-4 antibodies (please see above). In brief, for detection of Nectin-4 in serum, 96-well flexible microtiter plates (439454; NALGE NUNC International) were coated with 5 μg/mL of capturing monoclonal antibody to Nectin-4 (clone 19–33) overnight. Wells were blocked with 300 μL PBS (pH 7.4) containing 5% bovine serum albumin (BSA) for 2 h and then incubated for 2 h with 6-fold diluted serum samples in PBS (pH 7.4) containing 1% BSA. After washing with PBS (pH 7.4) containing 0.05% Tween 20, the wells were incubated for 2 h with 10 ng/mL of biotin-conjugated monoclonal anti–Nectin-4 antibody (clone 66–97), followed by reaction with avidin-conjugated peroxidase (P347; Dako Cytomation) using a Substrate Reagent (R&D Systems). The color reaction was terminated by addition of 50 μL 2N sulfuric acid. Color intensity was determined by a photometer at a wavelength of 450 nm, with a reference wave length of 570 nm. Standard curve was drawn for each plate using recombinant soluble Nectin-4 proteins (Nectin-4 ectodomain). Serum levels of Nectin-4 were calculated using dilutions of the recombinant Nectin-4 protein (raging from 0.1–300 ng/mL) as a reference. Levels of carcinoembryonic antigen (CEA) in serum were measured by ELISA with a commercially available enzyme test kit (Hope Laboratories) according to the supplier's recommendations. Levels of cytokeratin 19-fragment (CYFRA21-1) in serum were measured by ELISA with a commercially available kit (DRG).

Statistical analysis. We used contingency tables to analyze the relationship of Nectin-4 levels in tissues or serum and clinicopathologic variables of NSCLC patients. Survival curves were calculated from the date of surgery or diagnosis to the time of death, or to the last follow-up observation. Kaplan-Meier curves were calculated for each relevant variable, and for Nectin-4 expression in lung tumors or for serum Nectin-4 levels; differences in survival times among patient subgroups were analyzed using the log-rank test. Univariate and multivariate analyses were performed with the Cox proportional hazard regression model to determine associations between clinicopathologic variables and cancer-related mortality.

In ELISA, differences in the serum levels of Nectin-4, CEA, and CYFRA21-1 between tumor groups and a healthy control were analyzed by Mann-Whitney U tests.

RNA interference assay. To evaluate the biological functions of Nectin-4 in lung cancer cells, we used small interfering RNA (siRNA) duplexes against the target genes (Dharmacon) as described (37). The target sequences of the synthetic oligonucleotides for RNA interference were as follows: control 1 (a nonspecific control oligo); control 2 (Luciferase: Photinus pyralis luciferase gene), 5′-CGUACGCGGAAUACUUCGA-3′; siRNA-Nectin-4-#1, 5′-ACAGUUACCACGUCUGAGGUU-3′, siRNA-Nectin-4-#2, 5′-AAUGGUUCAUGGCCUGUUUUU-3′.

Rac1 activation assay. Activation of Rac1 by Nectin-4 overexpression was detected according to the supplier's recommendations using Rac1 activation assay kit (Cell Biolabs), and COS-7 and NIH-3T3 cells transfected with Nectin-4–expressing pcDNA3.1-myc/His plasmid or mock plasmid and further cultured for 48 h.

Nectin-4 expression in lung tumors and normal tissues. To search for novel target molecules for the development of therapeutic agents and/or diagnostic biomarkers for NSCLC, we first screened genes that showed more than a 5-fold higher level of expression in cancer cells than in normal cells, in the majority of NSCLCs analyzed by cDNA microarray. Among 27,648 genes and ESTs screened, we identified Nectin-4 to be a good candidate that showed more than a 5-fold higher level of expression in 87.5% of NSCLCs. We confirmed its transactivation by semiquantitative reverse transcription-PCR (RT-PCR) experiments in 11 of 14 additional NSCLC cases (6 of 7 ADCs; 5 of 7 SCCs), and in 7 of 20 lung cancer cell lines (Supplementary Fig. S1). We subsequently attempted to generate mouse monoclonal antibodies specific to human Nectin-4 and obtained two independent clones, 19-33 and 66-97. As Nectin-4 was indicated to be a type I membrane protein, we examined Nectin-4 expression on the surface of lung cancer cells using flow cytometry with anti–Nectin-4 monoclonal antibody (clone 19–33). This analysis indicated that Nectin-4 protein was highly expressed and localized at the plasma membranes of NCI-H2170 and NCI-H358 cells, in which Nectin-4 protein had been detected at a high level by semiquantitative RT-PCR, but not in those of A549 and SBC-5 cells that did not express Nectin-4 (Fig. 1A). Immunocytochemical staining of the same set of lung cancer cells with anti–Nectin-4 monoclonal antibody (clone 19–33) indicated that Nectin-4 was detected at plasma membrane and cytoplasm in NCI-H2170 and NCI-H358, but not in A549 and SBC-5 cells (Fig. 1B). Since the ectodomain of Nectin-4 (43.5 kDa) was suggested to be secreted by cleavage of its extracellular portion (41), we applied ELISA to examine its presence in the culture media of the lung cancer cell lines. The amounts of detectable Nectin-4 in the culture media was concordant to the expression levels of Nectin-4 detected with RT-PCR, flow cytometry, and immunofluorescence analyses (Fig. 1C), further supporting the specific binding affinity of our monoclonal antibodies to Nectin-4. Attempts to measure endogenous Nectin-4 protein levels in the NCI-H2170 and NCI-H358 cell lines by Western blotting using the two monoclonal antibodies (clones 19–33 and 66–97) failed to detect any bands on Western blots, indicating that these antibodies can be used for recognition of a native form of Nectin-4. We further confirmed that the two monoclonal antibodies (19–33 and 66–97) could specifically immunoprecipitate Nectin-4 protein in myc-tagged Nectin-4–overexpressing COS-7 cells (Supplementary Fig. S2, left), but not in mock-transformant COS-7 cells (Supplementary Fig. S2, right), suggesting that these antibodies have an ability to specifically recognize the native Nectin-4.

Figure 1.

Expression and cellular localization of Nectin-4 in lung cancers. A, flow cytometric analysis of the levels of endogenous Nectin-4 protein on the cell surface using nti-Nectin-4 monoclonal antibody and four lung cancer cell lines. Green peaks, the fluorescent intensities (Y-axis) detected by anti–Nectin-4 monoclonal antibody; black peaks, those of cells incubated with nonimmunized mouse IgG (control). B, subcellular localization of endogenous Nectin-4 protein, determined using anti–Nectin-4 monoclonal antibody. C, levels of secreted endogenous Nectin-4 protein detected with ELISA in culture medium from the four lung cancer cell lines.

Figure 1.

Expression and cellular localization of Nectin-4 in lung cancers. A, flow cytometric analysis of the levels of endogenous Nectin-4 protein on the cell surface using nti-Nectin-4 monoclonal antibody and four lung cancer cell lines. Green peaks, the fluorescent intensities (Y-axis) detected by anti–Nectin-4 monoclonal antibody; black peaks, those of cells incubated with nonimmunized mouse IgG (control). B, subcellular localization of endogenous Nectin-4 protein, determined using anti–Nectin-4 monoclonal antibody. C, levels of secreted endogenous Nectin-4 protein detected with ELISA in culture medium from the four lung cancer cell lines.

Close modal

Northern blotting using Nectin-4 cDNA as a probe identified the 3.7-kb transcript as a very faint signal in placenta and to a lesser degree in trachea among the 23 normal human tissues examined (data not shown), which was concordant with previous report (39). We subsequently examined expression of Nectin-4 protein in six normal tissues (heart, lung, liver, kidney, trachea, and placenta), as well as lung cancers using anti–Nectin-4 monoclonal antibody, and found that it was hardly detectable in the former five tissues, whereas positive Nectin-4 staining appeared in placenta and lung tumor tissues. The expression levels of Nectin-4 protein in lung cancer were significantly higher than those in placenta (Fig. 2A).

Figure 2.

Expression of Nectin-4 in normal tissues and lung tumors, and its association with poor prognosis for NSCLCs. A, immunohistochemical evaluation of Nectin-4 protein using anti–Nectin-4 antibody (clone 19–33) in lung adenocarcimona and six normal tissues (×200). B, immunohistochemical staining pattern of Nectin-4 protein in representative lung adenocarcimonas using anti–Nectin-4 antibody on tissue microarrays (×100). C, immunohistochemical staining of Nectin-4 protein using anti–Nectin-4 antibody in eight representative paired lung tumors and adjacent normal lung tissues (×200). D, Kaplan-Meier analysis of survival in patients with NSCLC according to Nectin-4 expression.

Figure 2.

Expression of Nectin-4 in normal tissues and lung tumors, and its association with poor prognosis for NSCLCs. A, immunohistochemical evaluation of Nectin-4 protein using anti–Nectin-4 antibody (clone 19–33) in lung adenocarcimona and six normal tissues (×200). B, immunohistochemical staining pattern of Nectin-4 protein in representative lung adenocarcimonas using anti–Nectin-4 antibody on tissue microarrays (×100). C, immunohistochemical staining of Nectin-4 protein using anti–Nectin-4 antibody in eight representative paired lung tumors and adjacent normal lung tissues (×200). D, Kaplan-Meier analysis of survival in patients with NSCLC according to Nectin-4 expression.

Close modal

Association of Nectin-4 expression with poor clinical outcomes for NSCLC patients. To further verify the biological and clinicopathologic significance of Nectin-4, we examined the expression of Nectin-4 protein by means of tissue microarrays consisting of NSCLC tissues from 422 patients who underwent surgical resection. We classified a pattern of Nectin-4 expression on the tissue array ranging from absent/weak (scored as 0–1+) to strong (2+; Fig. 2B). Of the 422 NSCLC cases examined, Nectin-4 was strongly stained in 245 cases (58.1%; score 2+), weakly stained in 119 cases (28.2%; score 1+), and not stained in 58 cases (13.7%; score 0), whereas their adjacent normal lung tissues were not stained (Fig. 2C). We then evaluated the association between Nectin-4 status and clinicopathologic parameters. As shown in Table 1A, histological type (higher in ADC; P = 0.0059 by Fisher's exact test) and pT factor (higher in T2–T4; P = 0.0048 by Fisher's exact test) were significantly associated with the strong Nectin-4 positivity (score 2+). NSCLC patients whose tumors showed strong Nectin-4 expression revealed shorter survival periods compared with those with absent/weak Nectin-4 expression (P < 0.0001 by log-rank test; Fig. 2D). We also applied univariate analysis to evaluate associations between patient prognosis and other factors including age (<65 versus ≥65), gender (female versus male), histologic type (ADC versus non-ADC), pT classification (T1 versus T2-4), pN classification (N0 versus N1+N2), smoking history (nonsmoker versus smoker), and Nectin-4 expression status (score 0+, 1+ versus 2+). Among those parameters, strong Nectin-4 positivity (P < 0.0001), elderly (P = 0.0079), male (P = 0.0011), non-ADC (P = 0.0208), advanced pT stage (P < 0.0001), and advanced pN stage (P < 0.0001) were significantly associated with poor prognosis. Multivariate analysis of these prognostic factors revealed that strong Nectin-4 expression, elderly, larger tumor size, and lymph node metastasis were independent prognostic factors for NSCLC patients (P < 0.0001, 0.0006, 0.0006, and <0.0001, respectively; Table 1B).

Serum levels of Nectin-4 in NSCLC patients. As the in vitro findings had suggested a possibility of application of Nectin-4 as a serum biomarker (Fig. 1C), we investigated whether the Nectin-4 is secreted into sera of patients with NSCLC. ELISA experiments detected Nectin-4 in serologic samples from the majority of the 164 patients with NSCLC (Fig. 3A); serum levels of Nectin-4 in NSCLC patients were 2.0 ± 2.5 ng/mL (mean ± 1 SD). In contrast, the mean (±1 SD) serum levels of Nectin-4 in 131 healthy volunteers were 0.6 ± 0.4 ng/mL, and those in 86 patients with COPD, who were current and/or former smokers, were 0.6 ± 0.9 ng/mL. The difference in the levels of serum Nectin-4 protein between NSCLC patients and healthy volunteers was significant with P value of <0.0001 (Mann-Whitney U test). The difference between healthy volunteers and COPD patients was not significant (P = 0.103 by Mann-Whitney U test). When classified according to histologic types, the serum levels of Nectin-4 were 2.0 ± 2.5 ng/mL in ADC patients and 2.0 ± 2.3 ng/mL in SCC patients (Fig. 3A,, left). Serum Nectin-4 was detected even in patients with operative stage tumors (stages I–IIIA; Fig. 3A,, right). Using receiver operating characteristic (ROC) curves drawn with the data of these 164 NSCLC patients and 131 healthy donors (Fig. 3B,, left), the cutoff level in this assay was set to provide optimal diagnostic accuracy and likelihood ratios (minimal false-negative and false-positive results) for Nectin-4, i.e., 1.0 ng/mL with a sensitivity of 53.7% [88 of 164 NSCLC; 6 of 24 (25%) stage I, 10 of 20 (50%); stage II–IIIA; and 72 of 120 (60%) stage IIIB–IV tumors] and a specificity of 97.7% (128 of 131; Supplementary Table S1A). To evaluate the potential of serum Nectin-4 level as a cancer-specific biomarker, we next examined the relationship between serum Nectin-4 positivity and patients' or healthy individuals' characteristics (Supplementary Table S1B and C; Table 2). Serum Nectin-4 positivity was significantly associated with pT factor (higher in T2–T4; P = 0.0039 by Fisher's exact test) in NSCLC patients (Table 2). Importantly, serum Nectin-4 positivity was not associated with smoking history, gender, and age in individual groups of lung cancer, COPD, and healthy volunteer (Supplementary Table S1B and C; Table 2). In addition, there was no significant association between its positivity and respiratory function in COPD patients (Supplementary Table S1C). To evaluate the feasibility of using serum Nectin-4 level as a tumor detection biomarker, we also measured by ELISA serum levels of two conventional tumor markers (CEA and CYFRA21-1) for NSCLC patients, in the same set of serum samples from cancer patients and control individuals. ROC analyses (Fig. 3B , left) determined the cutoff value of CEA for NSCLC detection to be 2.5 ng/mL with a sensitivity of 42.7% [70 of 164 NSCLC; 4 of 24 (16.7%) for stage I, 3 of 20 (15%) for stage II–IIIA, and 63 of 120 for (52.5%) stage IIIB–IV tumors] and a specificity of 97.7%, and also determined the cutoff value of CYFRA21-1 to be 2.0 pg/mL with a sensitivity of 39.0% [64 of 164 NSCLC; 1 of 24 (4.2%) for stage I, 3 of 20 (15%) for stage II–IIIA, and 60 of 120 (50%) for stage IIIB–IV tumors] and a specificity of 96.2% (please see also Supplementary Table S1A). The sum of the area under the ROC curve for serum Nectin-4 value was larger than that for serum CEA or CYFRA21-1, suggesting that Nectin-4 is likely to be a better diagnostic biomarker for NSCLC in the aspect of specificity and likelihood.

Figure 3.

Serologic concentration of Nectin-4 in NSCLC patients and its association with poor prognosis for NSCLCs. A, left, Serum Nectin-4 levels from patients with lung ADC or SCC determined by ELISA. Right, distribution of Nectin-4 in sera from patients at various clinical stages of lung cancer. B, left, ROC curve analysis of Nectin-4 (red), CEA (yellow), and CYFRA21-1 (green) as serum tumor biomarkers for NSCLC. Right, Serum Nectin-4 levels before and after surgical treatment in patients with NSCLC. C, serum Nectin-4 levels and the expression levels of Nectin-4 in primary tumor tissues in the same NSCLC patients. D, Kaplan-Meier analysis of survival after surgery in 95 patients with stage I NSCLC (top) or after the first-line chemotherapy with a combination of two drugs (carboplatin and paclitaxel) in 62 patients with stage IIIB to IV NSCLC (bottom), according to serum Nectin-4 positivity.

Figure 3.

Serologic concentration of Nectin-4 in NSCLC patients and its association with poor prognosis for NSCLCs. A, left, Serum Nectin-4 levels from patients with lung ADC or SCC determined by ELISA. Right, distribution of Nectin-4 in sera from patients at various clinical stages of lung cancer. B, left, ROC curve analysis of Nectin-4 (red), CEA (yellow), and CYFRA21-1 (green) as serum tumor biomarkers for NSCLC. Right, Serum Nectin-4 levels before and after surgical treatment in patients with NSCLC. C, serum Nectin-4 levels and the expression levels of Nectin-4 in primary tumor tissues in the same NSCLC patients. D, Kaplan-Meier analysis of survival after surgery in 95 patients with stage I NSCLC (top) or after the first-line chemotherapy with a combination of two drugs (carboplatin and paclitaxel) in 62 patients with stage IIIB to IV NSCLC (bottom), according to serum Nectin-4 positivity.

Close modal

We then evaluated the feasibility of using serum Nectin-4 level as a tumor detection biomarker in combination with CEA or CYFRA21-1. Measuring both Nectin-4 and CEA in serum can improve overall sensitivity for detection of lung ADC to 65.0% (for diagnosing ADC, the sensitivity of CEA alone is 42.3% and that of Nectin-4 is 54.5%). False-positive results for either of the two tumor markers among 131 healthy volunteers (control group) amounted to 4.6% (6 of 131), whereas the false-positive rates for CEA and Nectin-4 in the same control group were 2.3% (3 of 131) each. On the other hand, Nectin-4 and CYFRA21-1 in serum can improve overall sensitivity for detection of lung SCC to 68.3% (for diagnosing SCC, the sensitivity of CYFRA21-1 alone is 53.7% and that of Nectin-4 is 51.2%). False-positive results for either of the two tumor markers among 131 healthy volunteers (control group) amounted to 6.1% (8 of 131), whereas the false-positive rates for CYFRA21-1 and Nectin-4 in the same control group were 3.8% (5 of 131) and 2.3% (3 of 131), respectively.

We then performed ELISA experiments using paired preoperative and postoperative (2 months after the surgery) serum samples from NSCLC patients to monitor the levels of serum Nectin-4 in the same patients. The concentration of serum Nectin-4 was decreased after surgical resection of primary tumors (Fig. 3B,, right). We further compared the serum levels of Nectin-4 with the expression levels of Nectin-4 in primary tumors in the same set of 12 NSCLC cases whose serum had been collected before surgery (six patients with Nectin-4–positive tumors and six with Nectin-4–negative tumors). The serum levels of Nectin-4 showed good correlation with the expression levels of Nectin-4 in primary tumors (Fig. 3C), further suggesting that serum Nectin-4 was secreted from lung tumor.

Association of serum Nectin-4 positivity with poor clinical outcomes for NSCLC patients. We next examined by ELISA the serum Nectin-4 levels before curative surgery in additional 95 patients with stage I NSCLC and those at diagnosis in 62 patients with advanced NSCLC (stage IIIB–IV) who were newly diagnosed and previously untreated, and whose clinicopathologic background affecting prognosis was mostly identical, and found the correlation of serum Nectin-4 positivity with clinical outcomes. The median survival time of serum Nectin-4–positive patients with stage I NSCLC after curative surgery or advanced NSCLC treated with an identical protocol of chemotherapy was shorter than those of patients with serum Nectin-4 negative (P = 0.0219 and 0.0269, respectively, by log-rank test; Fig. 3D , top and bottom). We also applied univariate analysis to evaluate associations between patient prognosis and other factors including age, gender, histologic type, disease stage, smoking history, and serum Nectin-4 positivity. Univariate analysis indicated that only serum Nectin-4 positive was significantly associated with poor prognosis for early stage NSCLC patients after surgery (P = 0.0301; Supplementary Table S2A). In advanced NSCLCs, univariate analysis indicated that serum Nectin-4 positive (P = 0.036) and poorer Performance Status (P = 0.0004) were significantly associated with poor prognosis for newly diagnosed advanced NSCLC patients (Supplementary Table S2B). Multivariate analysis revealed that both serum Nectin-4 positivity and poorer Performance Status were independent prognostic factors for the NSCLC patients (P = 0.0394 and 0.0004, respectively; Supplementary Table S2B). These results independently support the high specificity and the great potentiality of serum Nectin-4 as a biomarker for detection of cancer at an early stage and for predicting the early progression of the disease.

Effect of Nectin-4 on the growth of NSCLC cells. To assess whether up-regulation of Nectin-4 plays a significant role in the growth and/or survival of lung cancer cells, we transfected siRNAs for Nectin-4 to lung cancer cell lines NCI-H2170 and NCI-H358 that overexpressed endogenous Nectin-4. The levels of Nectin-4 expression in the cells transfected with siRNA for Nectin-4 (si-Nectin-4-#1 and -#2) were significantly reduced compared with those transfected with any of the two control siRNAs (Fig. 4A). In accordance with the suppressive effect of the si-Nectin-4-#1, and -#2 on Nectin-4 expression, colony numbers and cell viability measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were significantly reduced (Supplementary Fig. S4A and B), but no such effect was observed by two control siRNAs.

Figure 4.

Inhibition of growth of NSCLC cells by siRNA against Nectin-4, and effect of Nectin-4 overexpression on activation of Rac1. A, expression of Nectin-4 in NCI-H2170 and NCI-H358 cells treated with si-Nectin-4 or control siRNAs analyzed by semiquantitative RT-PCR. B, MTT assay of the cells transfected with Nectin-4–specific siRNAs. All assays were performed thrice, and in triplicate wells. C, subcellular localization of exogenous Nectin-4 and F-actin in COS-7 and NIH-3T3 cells, detected with Alexa488-conjugated secondary antibodies to detect myc/His-tagged Nectin-4 and Alexa594-conjugated phalloidin. D, the elevated levels of GTP-Rac1 in COS-7 and NIH-3T3 cells transfected with Nectin-4–expressing plasmid.

Figure 4.

Inhibition of growth of NSCLC cells by siRNA against Nectin-4, and effect of Nectin-4 overexpression on activation of Rac1. A, expression of Nectin-4 in NCI-H2170 and NCI-H358 cells treated with si-Nectin-4 or control siRNAs analyzed by semiquantitative RT-PCR. B, MTT assay of the cells transfected with Nectin-4–specific siRNAs. All assays were performed thrice, and in triplicate wells. C, subcellular localization of exogenous Nectin-4 and F-actin in COS-7 and NIH-3T3 cells, detected with Alexa488-conjugated secondary antibodies to detect myc/His-tagged Nectin-4 and Alexa594-conjugated phalloidin. D, the elevated levels of GTP-Rac1 in COS-7 and NIH-3T3 cells transfected with Nectin-4–expressing plasmid.

Close modal

To further examine the effect of Nectin-4 on the growth of lung cancer cells, we transfected plasmids designed to express Nectin-4 or mock plasmids into PC-14 cells that did not express endogenous Nectin-4, and established two independent PC-14 cell lines overexpressing exogenous Nectin-4 (PC-14-Nectin-4-#A and PC-14-Nectin-4-#B) and two control cells (PC-14-Mock-#A and PC-14-Mock-#B). MTT assay revealed that growth of the two PC-14-Nectin-4 cells was promoted at a significant degree compared with control PC-14-Mock cells (Supplementary Fig. S3A). To investigate a potential role of Nectin-4 in vivo tumor growth, we s.c. transplanted either PC-14-Nectin-4-#B cells or PC-14-Mock-#B cells into BALB/cAJcl-nu/nu mice. During the 20-day observation, all three mice that were individually transplanted with PC-14-Nectin-4-#B cells showed significantly more rapid tumor growth compared with three independent mice transplanted with PC-14-Mock-#B cells (Supplementary Fig. S3B). These findings imply an in vivo and in vitro growth promoting effect of Nectin-4.

Enhancement of cellular invasion by overexpression of Nectin-4. As the immunohistochemical analysis on tissue microarray had indicated that NSCLC patients with Nectin-4–strong positive tumors showed shorter survival period than those with Nectin-4–negative or weak positive tumors, we examined a possible role of Nectin-4 in cellular migration and invasion by Matrigel assays using mammalian COS-7 and NIH-3T3 cells. As shown in Supplementary Fig. S4B, cells transfected with myc/His-tagged Nectin-4–expressing plasmids showed significant invasive activity through Matrigel compared with those transfected with mock vector.

Next, we examined the effect of Nectin-4 on the cell morphology by transfecting myc/His-tagged Nectin-4–expressing plasmid into COS-7 and NIH-3T3 cells. Immunocytochemical analysis using anti-myc antibody for exogenous Nectin-4 and phalloidin for filamentous actin (F-actin) clearly detected that the membrane protrusions, which were strongly stained with phalloidin, was significantly increased after transfection of Nectin-4 (Fig. 4C , top and bottom). No such effect was observed in the cells transfected with mock vector. Interestingly, exogenous Nectin-4 was strongly stained and partly colocalized with F-actin at these protrusions. These results suggest that overexpression of Nectin-4 induced lamellipodia formation in these cells.

Activation of Rac1 by overexpression of Nectin-4. The extension of protrusions, such as lamellipodia, is essential for cell motility, and the formation of lamellipodia requires activation of small GTPases Rac1 (48). We therefore examined whether Rac1 activity could be involved in Nectin-4–dependent enhancement of the cell motility. Like other small GTPases, Rac1 regulates molecular events by cycling between an inactive GDP-bound form and an active GTP-bound form. In its active (GTP-bound) state, Rac1 binds specifically to the p21-binding domain of p21-activated protein kinase to control downstream signaling cascades. Based on this mechanism, we attempted to investigate a possible activation of Rac1 by Nectin-4 overexpression, using lysate from COS-7 and NIH-3T3 cells transfected with Nectin-4–expressing plasmid or mock plasmid, which were mixed with p21-activated protein kinase-p21-binding domain agarose beads to affinity-precipitate the protein complex containing GTP-Rac1 (active form). The levels of GTP-Rac1 were elevated in COS-7 and NIH-3T3 cells transfected with Nectin-4–expressing plasmid, compared with those transfected with mock vector (Fig. 4D). These results indicate that Nectin-4 could enhance the lamellipodia formation and cellular invasion possibly through activation of Rac1.

Nectin family members are both homophilic and heterophilic cell adhesion molecules that bind afadin, an actin filament (F-actin)–binding protein through their cytoplasmic tails and associate with the actin cytoskeleton, and could regulate many other cellular activities such as movement, differentiation, polarization, and the entry of viruses, in cooperation with other cell adhesion molecules and cell surface membrane receptors (38, 39). In our study, we have shown that activation of Nectin-4 is likely to be an essential contributor to the cell growth and highly malignant phenotype of tumors through the Rac1-signaling pathway. Although detailed functional association between Nectin-4 transactivation and lung carcinogenesis remains to be clarified, targeting the Nectin-4 pathway provides a large possibility for developing new types of therapeutic drugs such as nucleic acid drugs, monoclonal antibodies, and cancer vaccines that are expected to have a powerful biological activity against cancer with a minimal risk of adverse events in patients.

Possible mechanisms of antibody-based cancer therapy can be classified into two categories (49). One is the direct action, which can be subcategorized into three modes, (a) blocking function, (b) stimulating function, and (c) targeting function. The other mechanism of antibody therapy is the indirect action by the immune system, including complement-dependent cytotoxicity and antibody-dependent cytotoxicity (49). We confirmed that two mouse monoclonal antibodies raised against the ectodomain of Nectin-4 (clones 19–33 and 66–97) for ELISA had no antitumor activity in vitro when treated Nectin-4–overexressing lung cancer cells only with the antibodies (data not shown). Because Nectin-4 was highly expressed on the surface of lung cancer cells and the ectodomain of Nectin-4 was secreted (Fig. 1A), we need to clarify in the future study the mechanism of carcinogenesis involving the cell surface and/or extracellular Nectin-4. Based on the data, the production of monoclonal antibodies targeting cell surface Nectin-4 that could induce antibody-dependent cytotoxicity/complement-dependent cytotoxicity activity and/or block oncogenic receptor signals, or those neutralizing the secreted Nectin-4 would be warranted.

We established our original ELISA system by the two mouse monoclonal antibodies to measure serum levels of Nectin-4 and showed that serum Nectin-4 levels were significantly higher in lung cancer patients than in healthy volunteers. This is a great advantage to develop practical and standardized diagnostic kits in the clinic. Extracellular part of Nectin-4 is reported to be processed by TACE/ADAM-17 and a soluble Nectin-4 is shed from the cell surface and released into the extracellular space (41). Metalloprotease-mediated cleavage of some single-pass type I transmembrane proteins, such as L1CAM released by ADAM10-mediated membrane-proximal cleavage, has been shown to stimulate cellular migration and neurite outgrowth (50). The ectodomain shedding of Nectin-4 might exert some signals important for cancer progression. Importantly, serum Nectin-4 levels measured by our ELISA system showed higher sensitivity and specificity (53.7% and 97.7%, respectively) than conventional serum tumor markers for NSCLC (CEA or CYFRA21-1) that are being used in clinical practice (Supplementary Table S1A). Furthermore, serum Nectin-4 in patients with operable stages of NSCLC also showed higher sensitivity (25% in stage I; 50% in stage II–IIIA) than serum CEA (16.7% in stage I; 15% in stage II–IIIA) or CYFRA21-1 (4.2% in stage I; 15% in stage II–IIIA). Although further validation using a larger set of serum samples covering various clinical stages will be required, our data presented here also indicate a potential clinical usefulness of Nectin-4 as a serologic biomarker for NSCLC that could be widely used in clinical practice, such as detection of cancer, prediction of the malignant potential of tumor, and monitoring the disease control condition after any anticancer treatment. Finally, it should be noted that we observed activation of Nectin-4 in more than half of a series of other types of cancers such as bladder and cervical carcinomas we examined (data not shown), suggesting its diagnostic and therapeutic application to a wide-range of tumors.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Ahmedin J, Rebecca S, Elizabeth W, et al. Cancer Statistics, 2007.
CA Cancer J Clin
2007
;
57
:
43
–66.
2
Parkin DM. Global cancer statistics in the year 2000.
Lancet Oncol
2001
;
2
:
533
–43.
3
Naruke T, Tsuchiya R, Kondo H, Asamura H. Prognosis and survival after resection for bronchogenic carcinoma based on the 1997 TNM-staging classification: the Japanese experience.
Ann Thorac Surg
2001
;
71
:
1759
–64.
4
Schiller JH, Harrington D, Belani CP, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer.
N Engl J Med
2002
;
346
:
92
–8.
5
Thatcher N. The place of targeted therapy in the patient management of non-small cell lung cancer.
Lung Cancer
2007
;
57
Suppl 2:
S18
–23.
6
Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer.
N Engl J Med
2006
;
355
:
2542
–50.
7
Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer.
N Engl J Med
2005
;
353
:
123
–32.
8
Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer).
Lancet
2005
;
366
:
1527
–37.
9
Cesare G, Paolo M, Filomena G, et al. Sorafenib and Sunitinib in the Treatment of advanced non-small cell lung cancer.
The Oncologist
2007
;
12
:
191
–200.
10
Daigo Y, Nakamura Y. From cancer genomics to thoracic oncology: discovery of new biomarkers and therapeutic targets for lung and esophageal carcinoma.
Gen Thorac Cardiovasc Surg
2008
;
56
:
43
–53.
11
Kikuchi T, Daigo Y, Katagiri T, et al. Expression profiles of non-small cell lung cancers on cDNA microarrays: identification of genes for prediction of lymph-node metastasis and sensitivity to anti-cancer drugs.
Oncogene
2003
;
22
:
2192
–205.
12
Kakiuchi S, Daigo Y, Tsunoda T, Yano S, Sone S, Nakamura Y. Genome-wide analysis of organ-preferential metastasis of human small cell lung cancer in mice.
Mol Cancer Res
2003
;
1
:
485
–99.
13
Kakiuchi S, Daigo Y, Ishikawa N, et al. Prediction of sensitivity of advanced non-small cell lung cancers to gefitinib (Iressa, ZD1839).
Hum Mol Genet
2004
;
13
:
3029
–43.
14
Kikuchi T, Daigo Y, Ishikawa N, et al. Expression profiles of metastatic brain tumor from lung adenocarcinomas on cDNA microarray.
Int J Oncol
2006
;
28
:
799
–805.
15
Taniwaki M, Daigo Y, Ishikawa N, et al. Gene expression profiles of small-cell lung cancers: molecular signatures of lung cancer.
Int J Oncol
2006
;
29
:
567
–75.
16
Suzuki C, Daigo Y, Kikuchi T, Katagiri T, Nakamura Y. Identification of COX17 as a therapeutic target for non-small cell lung cancer.
Cancer Res
2003
;
63
:
7038
–41.
17
Ishikawa N, Daigo Y, Yasui W, et al. ADAM8 as a novel serological and histochemical marker for lung cancer.
Clin Cancer Res
2004
;
10
:
8363
–70.
18
Kato T, Daigo Y, Hayama S, et al. A novel human tRNA-dihydrouridine synthase involved in pulmonary carcinogenesis.
Cancer Res
2005
;
65
:
5638
–46.
19
Furukawa C, Daigo Y, Ishikawa N, et al. Plakophilin 3 oncogene as prognostic marker and therapeutic target for lung cancer.
Cancer Res
2005
;
65
:
7102
–10.
20
Ishikawa N, Daigo Y, Takano A, et al. Increases of amphiregulin and transforming growth factor-α in serum as predictors of poor response to gefitinib among patients with advanced non-small cell lung cancers.
Cancer Res
2005
;
65
:
9176
–84.
21
Suzuki C, Daigo Y, Ishikawa N, et al. ANLN plays a critical role in human lung carcinogenesis through the activation of RHOA and by involvement in the phosphoinositide 3-kinase/AKT pathway.
Cancer Res
2005
;
65
:
11314
–25.
22
Ishikawa N, Daigo Y, Takano A, et al. Characterization of SEZ6L2 cell-surface protein as a novel prognostic marker for lung cancer.
Cancer Sci
2006
;
97
:
737
–45.
23
Takahashi K, Furukawa C, Takano A, et al. The neuromedin u-growth hormone secretagogue receptor 1 b/neurotensin receptor 1 oncogenic signaling pathway as a therapeutic target for lung cancer.
Cancer Res
2006
;
66
:
9408
–19.
24
Hayama S, Daigo Y, Kato T, et al. Activation of CDCA1-2, members of centromere protein complex, involved in pulmonary carcinogenesis.
Cancer Res
2006
;
66
:
10339
–48.
25
Kato T, Hayama S, Yamabuki Y, et al. Increased expression of insulin-like growth factor-II messenger RNA-binding protein 1 Is associated with tumor progression in patients with lung cancer.
Clin Cancer Res
2007
;
13
:
434
–42.
26
Suzuki C, Takahashi K, Hayama S, et al. Identification of Myc-associated protein with JmjC domain as a novel therapeutic target oncogene for lung cancer.
Mol Cancer Ther
2007
;
6
:
542
–51.
27
Yamabuki T, Takano A, Hayama S, et al. Dickkopf-1 as a novel serologic and prognostic biomarker for lung and esophageal carcinomas.
Cancer Res
2007
;
67
:
2517
–25.
28
Hayama S, Daigo Y, Yamabuki T, et al. Phosphorylation and activation of cell division cycle associated 8 by aurora kinase B plays a significant role in human lung carcinogenesis.
Cancer Res
2007
;
67
:
4113
–22.
29
Kato T, Sato N, Hayama S, et al. Activation of HJURP (Holliday Junction-Recognizing Protein) involved in the chromosomal stability and immortality of cancer cells.
Cancer Res
2007
;
67
:
8544
–53.
30
Taniwaki M, Takano A, Ishikawa N, et al. Activation of KIF4A as a prognostic biomarker and therapeutic target for lung cancer.
Clin Cancer Res
2007
;
13
:
6624
–31.
31
Ishikawa N, Takano A, Yasui W, et al. Cancer-testis antigen lymphocyte antigen 6 complex locus K is a serologic biomarker and a therapeutic target for lung and esophageal carcinomas.
Cancer Res
2007
;
67
:
11601
–11.
32
Mano Y, Takahashi, K, Ishikawa N, et al. Fibroblast growth factor receptor 1 oncogene partner as a novel prognostic biomarker and therapeutic target for lung cancer.
Cancer Sci
2007
;
98
:
1902
–13.
33
Suda T, Tsunoda T, Daigo Y, Nakamura Y, Tahara H. Identification of human leukocyte antigen-A24-restricted epitope peptides derived from gene products upregulated in lung and esophageal cancers as novel targets for immunotherapy.
Cancer Sci
2007
;
98
:
1803
–8.
34
Kato T, Sato N, Takano A, et al. Activation of placenta specific transcription factor distal-less homeobox 5 predicts clinical outcome in primary Lung cancer patients.
Clin Cancer Res
2008
;
14
:
2363
–70.
35
Mizukami Y, Kono K, Daigo Y, et al. Detection of novel cancer-testis antigen-specific T-cell responses in TIL, regional lymph nodes, and PBL in patients with esophageal squamous cell carcinoma.
Cancer Sci
2008
;
99
:
1448
–54.
36
Harao M, Hirata S, Irie A, et al. HLA-A2-restricted CTL epitopes of a novel lung cancer-associated cancer testis antigen, cell division cycle associated 1, can induce tumor-reactive CTL.
Int J Cancer
2008
;
123
:
2616
–25.
37
Hirata D, Yamabuki T, Ito T, et al. Involvement of epithelial cell transforming sequence 2 (ECT2) oncoantigen in lung and esophageal cancer progression.
Clin Cancer Res
2009
;
15
:
256
–66.
38
Takai Y, Miyoshi J, Ikeda W, Ogita H. Nectins and nectin-like molecules: roles in contact inhibition of cell movement and proliferation.
Nat Rev Mol Cell Biol
2008
;
9
:
603
–15.
39
Reymond N, Fabre S, Lecocq E, Adelaide J, Dubreuil P, Lopez M. Nectin4/PRR4, a new afadin-associated member of the nectin family that trans-interacts with nectin1/PRR1 through V domain interaction.
J Biol Chem
2001
;
276
:
43205
–15.
40
Fabre S, Reymond N, Cocchi F, et al. Prominent role of the Ig-like V domain in trans-interactions of nectins. Nectin3 and nectin 4 bind to the predicted C-C′-C″-D β-strands of the nectin1 V domain.
J Biol Chem
2002
;
277
:
27006
–13.
41
Fabre-Lafay S, Garrido-Urbani S, Reymond N, Goncalves A, Dubreuil P, Lopez M. Nectin-4, a new serological breast cancer marker, is a substrate for tumor necrosis factor-α-converting enzyme (TACE)/ADAM-17.
J Biol Chem
2005
;
280
:
19543
–50.
42
Fabre-Lafay S, Monville F, Garrido-Urbani S, et al. Nectin-4 is a new histological and serological tumor associated marker for breast cancer.
BMC Cancer
2007
;
7
:
73
.
43
Travis WD, Colby TV, Corrin B, Shimosato Y, Brambilla E. Histological Typing of Lung and Pleural Tumors: World Health Organization International Histological Classification of Tumors, 3rd edn. Berlin: Springer; 1999.
44
Sobin L, Wittekind CH. TNM Classification of Malignant Tumours, 6th ed. New York: Wiley-Liss; 2002.
45
Chin SF, Daigo Y, Huang HE, et al. A simple and reliable pretreatment protocol facilitates fluorescent in situ hybridisation on tissue microarrays of paraffin wax embedded tumour samples.
Mol Pathol
2003
;
56
:
275
–9.
46
Callagy G, Cattaneo E, Daigo Y, et al. Molecular classification of breast carcinomas using tissue microarrays.
Diagn Mol Pathol
2003
;
12
:
27
–34.
47
Callagy G, Pharoah P, Chin SF, et al. Identification and validation of prognostic markers in breast cancer with the complementary use of array-CGH and tissue microarrays.
J Pathol
2005
;
205
:
388
–96.
48
Takai Y, Sasaki T, Matozaki T. Small GTP-Binding Proteins.
Physiol Rev
2001
;
81
:
153
–208.
49
Imai K, Takaoka A. Comparing antibody and small-molecule therapies for cancer.
Nat Rev Cancer
2006
;
6
:
714
–27.
50
Fogel M, Gutwein P, Mechtersheimer S, et al. L1 expression as a predictor of progression and survival in patients with uterine and ovarian carcinomas.
Lancet
2003
;
362
:
869
–75.