Main

Gastric cancer is the second most common cause of cancer-related death worldwide (Health and Welfare Statistics Association: Tokyo, 2006; Katanoda and Yako-Suketomo, 2009). Although complete removal of the tumour by surgical resection is an ideal treatment option for patients with gastric cancer, many patients with advanced-stage gastric cancer need to be treated with intensive chemotherapy. Gastric cancer patients exhibit high relapse rates even after curative surgery and unresponsiveness to chemotherapy, resulting in dismal survival rates (Sasako et al, 2011). Several methods for the prediction and early detection of subclinical ‘minimal residual cancer’ after surgery (Austrup et al, 2000; Klein et al, 2002) or relapse have been developed, for example, peritoneal lavage, positron emission tomography, gene profiling, and so on. (Motoori et al, 2006; Makino et al, 2010; Graziosi et al, 2011), reliable markers that can specifically reflect gastric cancer disease status have not been determined.

Analysing serum level of tumour markers is employed for cancer detection, monitoring patients’ disease status, and prognosis prediction. Several organ-specific tumour markers are used in the clinic, for example, prostate-specific antigen and prostatic acid phosphatase for prostate cancer (Seamonds et al, 1986; Ferro et al, 1987) and protein induced by vitamin K absence-II for liver cancer (Fujiyama et al, 1986). As no gastric cancer-specific markers have been determined, a combination of several nonspecific tumour markers, for example, carcinoembryonic antigen (CEA), CA19-9,and so on, is merely applicable for monitoring treatment efficacy, but not the diagnosis of gastric cancer (Takahashi et al, 1995, 2003). Carcinoembryonic antigen and CA19-9 are found in the sera of 20–60% of gastric cancer patients, and their expression levels in gastric cancer are related to clinical events, such as relapse (Kodera et al, 1996). Carcinoembryonic antigen value, in particular, is indicative of the formation of a large tumour, liver or peritoneal metastasis, and/or a high risk of relapse and poor prognosis (Ikeda et al, 1993; Yamamoto et al, 2004). However, as CEA, a cell surface-anchored glycoprotein, is expressed in normal cell membranes, 5% of CEA-positive cases are pseudopositives, that is, caused by heavy smoking, endometriosis, and ageing, and so on. (Alexander et al, 1976), suggesting the importance of novel markers for gastric cancer.

NY-ESO-1 antigen, a cancer/testis (CT) antigen, was originally identified in oesophageal cancer by serological expression cloning using autologous patient serum and has been shown to be strongly immunogenic. Spontaneous NY-ESO-1 antibody production is often observed in patients with NY-ESO-1-expressing tumours, for example, 9.4% of melanoma patients, 12.5% of ovarian cancer patients, 7.7–26.5% of breast cancer patients, 4.2–20.0% of lung cancer patients, and 52% of prostate cancer patients, but has not been detected in non-cancerous donors (Stockert et al, 1998; Nakada et al, 2003; Türeci et al, 2006; Chapman et al, 2007; Isobe et al, 2009; Gati et al, 2011). Thus, it is possible that the NY-ESO-1 humoral immune response could be used as a serological marker for detecting these cancers and to facilitate the clinical management of some patients with particular types of cancer (Gnjatic et al, 2006). Jäger et al (1999) found that the change in the NY-ESO-1 humoral immune response reflected the overall tumour load in 10 out of 12 patients with various cancers. However, there is ongoing controversy regarding the association between the NY-ESO-1 immune response and prognostic criteria (Yuan et al, 2011). To address these issues in gastric cancer, we investigated the clinical usefulness of the NY-ESO-1 humoral immune response for diagnosis, monitoring, and relapse prediction in gastric cancer patients.

Materials and methods

Serum sample and tissue specimen collection from gastric cancer patients

In all, 363 patients with histologically confirmed gastric cancer, who underwent surgical resection or chemotherapy at one of four institutions between 2004 and 2011, were included in this study after providing written informed consent. Serum samples were obtained from the 363 patients during their admission to hospital for surgical treatment and/or chemotherapy, and afterwards, serial serum samples were obtained at each follow-up visit from 25 patients who displayed NY-ESO-1 humoral immune responses. All serum samples were collected as surplus samples after routine blood tests and stored. Fixed and frozen gastric cancer tissue samples were obtained from 60 out of 363 patients during surgery and stored. The samples were subsequently subjected to expression analysis. Information regarding blood test results, tumour stage, histological type, depth of invasion, lymph node metastasis, and distant metastasis, which were obtained from pathological examinations and CT scans, were collected from the relevant patient databases. Serum samples obtained from 50 healthy donors were used as controls. This study was approved by the institutional review boards of Osaka University Hospital, Toyonaka Municipal Hospital, Ikeda City Hospital, and Minoh City Hospital.

Reverse transcription–polymerase chain reaction

Total cellular RNA was extracted from the frozen tissue using TRIZOL reagent (Invitrogen, Carlsbad, CA, USA). The total RNA (1 μg) was subjected to the reverse transcription (RT) in 20 μl buffer with oligo-(dT)15 primer using a RT system (Promega, Madison, WI, USA). Conventional polymerase chain reaction (PCR) was performed in a 25-μl reaction mixture containing 1 μl of cDNA template, 500 nM of each primer, and 1 U of Taq DNA polymerase (AmpliTaq Gold, Roche Molecular Systems, Pleasanton, CA, USA) in the following conditions: one cycle of 95 °C for 12 min; followed by 35 cycles of 94 °C for 1 min, 60 °C for 1 min, and 72 °C for 1.5 min; and then a final step of 72 °C for 10 min. The sequences of the primers for NY-ESO-1 were as follows: ESO1-1, 5′-AGTTCTACCTCGCCATGCCT-3′; and ESO1-2, 5′-TCCTCCTCCAGCGACAAACAA-3′. The integrity of each RNA sample was verified by performing RT–PCR for porphobilinogen deaminase (PBGD). The PCR products were subjected to electrophoresis on a 2% agarose gel and visualised with ethidium bromide.

Immunohistochemistry

Formalin-fixed, paraffin-embedded tissues were used for the immunohistochemistry (IHC) analyses. Slides were incubated with the primary antibody overnight at 4 °C. The monoclonal antibody E978, which was previously generated by our group, was used to detect NY-ESO-1. The slides were then subjected to a heat-based antigen retrieval technique by immersing them in a preheated buffer solution (hipH solution; Dako, Carpinteria, CA, USA). A polymer-based antibody detection system (PowerVision; Leica Microsystems, Buffalo Grove, IL, USA) was used as the secondary reagent, and 3,3-diaminobenzidine tetrahydrochloride (Liquid DAB; Biogenex, San Ramon, CA, USA) was used as the chromogen. Normal adult testis tissue as a positive control and appropriate negative controls were included for each case.

Enzyme-linked immunosorbent assay

A measure of 100 μl of 1 μg ml−1 recombinant protein in coating buffer (pH 9.6) were added to each well of 96-well PolySorp immunoplates (Nunc, Roskilde, Denmark) and incubated overnight at 4 °C. The plates were then washed with PBS and blocked with 200 μl per well of 5% FCS/PBS for 1 h at room temperature. After being washed again, 100 μl of serially diluted serum were added to each well and incubated for 2 h at room temperature. Then, after extensive washing, goat anti-human IgG (Medical & Biological Laboratories, Nagoya, Japan) was added to the wells as a secondary antibody, and the plates were incubated for 1 h at room temperature. The plates were washed again, and the signals were developed with 100 μl per well of 0.03% o-phenylene diamine dihydrochloride, 0.02% hydrogen peroxide, and 0.15 M citrate buffer, and absorbance was read at 490 nm using an enzyme-linked immunosorbent assay (ELISA) reader (Benchmark Microplate Reader; Bio-Rad, Hercules, CA, USA). Ovalbumin (OVA; Sigma, St Louis, MO, USA) was used as the control protein. Levels of NY-ESO-1 humoral response were assessed using optical density (OD) values.

CEA and CA19-9

Serum CEA and CA19-9 levels were measured at each hospital’s clinical laboratory department. Carcinoembryonic antigen and CA19-9 positivity were defined as serum levels of CEA and CA19-9 of >5.0 ng ml−1 and >37 U ml−1, respectively.

Statistical analysis

Fisher’s exact test was used to assess the associations between NY-ESO-1 antibody expression and clinicopathological parameters. Kaplan–Meier curves were plotted to assess the effect of the NY-ESO-1 antibody on overall survival. Survival curves were compared using the log-rank test.

Results

Determination of NY-ESO-1 humoral immune response positivity

We first determined the OD cutoff value for NY-ESO-1 humoral immune response positivity. When the serum samples from the 50 healthy donors were examined for reactivity to the NY-ESO-1 recombinant protein by ELISA, their OD values ranged from 0.08 to 0.20, and their mean and standard deviation values were 0.15 and 0.05, respectively, at a dilution of 1 : 200. Thus, NY-ESO-1 humoral immune response positivity was defined as an OD value of >0.25 at a dilution of 1 : 200 (95% accuracy level) and >3 times of the OD value against control protein (OVA).

NY-ESO-1 humoral immune responses of gastric cancer patients

Serum samples were obtained from 363 gastric cancer patients, including 176 stage I, 45 stage II, 67 stage III, and 75 stage IV patients at admission (Table 1). The NY-ESO-1 antibody was detected in 3.4% (6 of 176) of stage I, 4.4% (2 of 45) of stage II, 25.3% (17 of 67) of stage III, and 20.0% (16 of 75) of stage IV gastric cancer patients, resulting in an overall detection rate of 11.1% (41 of 363). An analysis of the gastric cancer patients’ characteristics found that NY-ESO-1 antibody positivity was significantly correlated with gender (male>female) and tumour progression (Table 2). In particular, the patients with progressive gastric cancer involving deeper tumour invasion, positive lymph node metastasis, positive distant metastasis, or a higher clinical stage tended to produce the NY-ESO-1 antibody.

Table 1 Frequencies of NY-ESO-1 antibody, CEA, and CA19-9 in gastric cancer patients
Table 2 Relationship between NY-ESO-1 antibody positivity and clinicopathological features in gastric cancer patients

Analysis of NY-ESO-1 antigen expression

NY-ESO-1 mRNA and NY-ESO-1 protein expression were analysed by RT–PCR and IHC, respectively, in gastric cancer tissues obtained from 60 patients for whom both frozen and formalin-fixed specimens were available, including 12 stage I, 12 stage II, 20 stage III, and 16 stage IV patients (Table 3). NY-ESO-1 mRNA was detected in six specimens. NY-ESO-1 was immunohistochemically detected in 19 specimens, including 6 and 13 that were positive and negative for NY-ESO-1 mRNA, respectively. Most of the specimens displayed a heterogeneous staining pattern (data not shown).

Table 3 Frequency of NY-ESO-1 antibody positives in gastric cancer patients in whom the NY-ESO-1 antigen was or was not detected by IHC or RT–PCR

NY-ESO-1 antibody and antigen expression

We analysed the frequency of NY-ESO-1 antibody positivity in gastric cancer patients in whom NY-ESO-1 antigen expression was or was not detected by RT–PCR or IHC. As shown in Table 3, 9 out of the 60 gastric cancer patients whose specimens were available for expression analysis possessed the NY-ESO-1 antibody in their sera. The NY-ESO-1 antibody was detected in 8 of 19 (42.1%) patients with IHC-positive gastric cancer and 5 of 6 (83.3%) patients with RT–PCR (and IHC)-positive gastric cancer, whereas only 1 of 41 patients in whom both RT–PCR and IHC analysis produced negative results displayed an NY-ESO-1 humoral immune responses.

Frequencies of NY-ESO-1 humoral immune responses and conventional tumour markers in gastric cancer patients

The frequency of the NY-ESO-1 humoral immune response was compared with those of conventional tumour markers in gastric cancer patients. The serum CEA and CA19-9 levels of 363 gastric cancer patients were measured at admission (Table 1). Carcinoembryonic antigen and CA19-9 positivity were observed in 21.2% (77 of 363) and 14.9% (54 of 363) of the gastric cancer patients, respectively, and, except for CA19-9 in the stage III patients, they displayed higher frequencies than the NY-ESO-1 humoral immune response in all stages of the disease. We then analysed whether the addition of the NY-ESO-1 humoral immune response to CEA and CA19-9 increased the diagnostic frequency of gastric cancer. The combined use of CEA and CA19-9 tests produced positivity rates of 15.3% (27 of 176) in stage I, 24.4% (11 of 45) in stage II, 37.3% (25 of 67) in stage III, and 53.3% (40 of 75) in stage IV gastric cancer patients, resulting in an overall positivity rate of 28.4% (103 of 363). When the NY-ESO-1 humoral immune response was added to these two conventional tumour markers, the positivity rates of all stages increased, resulting in information gains of 14.9% (from 25 to 35 patients; 10 of 67) in stage III and 11.2% (from 65 to 81 patients; 16 of 142) in stage III and IV gastric cancer patients.

Changes in the NY-ESO-1 humoral immune responses of the patients during their clinical courses

Serial serum samples were obtained from 25 gastric cancer patients who displayed positive NY-ESO-1 antibody at admission, and the changes in their NY-ESO-1 humoral immune responses were examined throughout their clinical courses. In all, 6 stage I, 2 stage II, and 8 stage III patients received curative surgical treatment, and 14 did not suffer recurrence. The NY-ESO-1 immune response levels of the patients who did not suffer recurrence decreased after treatment and had fallen below the cutoff level by 9 months after surgery in most cases and did not subsequently increase (Figure 1). The half-lives of their NY-ESO-1 humoral immune response levels were 1.5, 1.6, 2.1, 3.2, and 6.6 months in the stage I patients; 3.0 and 4.0 months in the stage II patients; and 1.6, 1.9, 2.3, 3.0, 3.2, 4.1, and 6.7 months in the stage III patients (mean: 3.0 months). On the other hand, the two patients who underwent curative surgery but subsequently suffered recurrence, M-2 (stage I) and M-11 (stage III), displayed not only incomplete decreases in their NY-ESO-1 humoral immune response levels but also their subsequent restoration to pretreatment levels (Figure 1 and Figure 2A and B). In a comparison between the patients’ conventional tumour marker levels and their NY-ESO-1humoral immune response levels, we found that the changes in their CEA and CA19-9 levels were consistent with their NY-ESO-1 immune response levels in patient M-2, whereas patient M-11 was negative for both CEA and CA19-9 throughout their clinical course. Nine stage IV patients who received chemotherapy alone maintained high NY-ESO-1 humoral immune response levels throughout their clinical courses, including some patients who achieved partial tumour responses after chemotherapy (Figure 1).

Figure 1
figure 1

Change in the NY-ESO-1 humoral immune responses of gastric cancer patients after treatment. The serum NY-ESO-1 humoral immune responses of patients with stage I, II, III, or IV gastric cancer in whom NY-ESO-1 antibody production was detected before surgical treatment or chemotherapy were serially analysed. In all, 6 stage I, 2 stage II, and 8 stage III patients received curative surgery, and only 2 patients (, ■) suffered recurrence. Other 14 patients did not suffer recurrence. Nine patients with stage IV gastric cancer received chemotherapy alone after the initial detection of NY-ESO-1 antibody. Each mark represents a patient. Optical density (OD) values were measured at a serum dilution of 1 : 200.

Figure 2
figure 2

NY-ESO-1 humoral immune response, CEA, and carbohydrate antigen (CA)19-9 levels of patients who relapsed after curative surgery. The NY-ESO-1 humoral immune response (, ■ Figure 1), CEA (), and CA19-9 () levels of two patients, M-2 (stage I) (A) and M-11 (stage III) (B), who underwent curative surgery but subsequently suffered recurrence, were serially analysed. OD values were measured at a serum dilution of 1 : 200. The closed marks indicate CEA or CA19-9 positivity.

Prognostic value of the NY-ESO-1 humoral immune response in gastric cancer

The prognostic value of the NY-ESO-1 immune response was evaluated in gastric cancer patients. An analysis of the cumulative overall survival of the gastric cancer patients indicated that there was no difference in the survival rates of the patients who did and did not display positive NY-ESO-1 humoral immune responses (Figure 3A). However, among the patients with higher stage gastric cancer, overall survival was better in the patients in whom NY-ESO-1humoral immune responses were detected, although the difference was not significant (Figure 3B). NY-ESO-1 protein expression, as detected by IHC, did not affect the overall survival rate (data not shown).

Figure 3
figure 3

Prognostic role of NY-ESO-1 antibody in gastric cancer patients. The cumulative overall survival rate was analysed in all patients (n=310; A) and stage III and IV (n=126; B) gastric cancer patients in whom NY-ESO-1 antibodies were (continuous line) and were not detected (dotted line). The detection of NY-ESO-1 protein by IHC analysis did not affect the overall survival rate (data not shown). Survival curves were plotted using the Kaplan–Meier method. The log-rank test was used for comparisons between groups. P-values <0.05 were considered significant.

Discussion

NY-ESO-1 antibody was detected in 23.2% of stage III and IV gastric cancer patients, and the combinatorial use of the NY-ESO-1 antibody with CEA and CA19-9 as tumour markers increase the percentage of tumour detection from 45.8 to 57.0%. As the frequency of NY-ESO-1 humoral immune response was relatively low in the patients with early-stage gastric cancer, analysing serum NY-ESO-1 antibody levels alone might not be useful for screening for early-stage gastric cancer. Nevertheless, the expression of NY-ESO-1, a CT antigen, is restricted to tumour tissues and NY-ESO-1 antibody is only detectable in patients with NY-ESO-1-expressing tumours (Stockert et al, 1998), indicating the highly specific nature of NY-ESO-1 humoral immune responses in cancer patients. Given that NY-ESO-1 expression by malignant cells is required for antibody induction (Stockert et al, 1998), the detection of NY-ESO-1 antibody would be helpful for diagnosing malignancy, although extensive analysis of serum samples from patients with non-cancerous disease, for example, liver or renal disorders, autoimmune diseases, and so on, would be necessary to confirm. In our expression analysis, more NY-ESO-1-positive cases were detected by IHC (19 of 60) than by RT–PCR (6 of 60). This was probably due to the heterogeneous expression of NY-ESO-1 in gastric cancer and the fact that a limited number of biopsy samples were used for the RT–PCR, whereas multiple slices from whole tumour specimens were used for the IHC. Extensive IHC analysis should be used for NY-ESO-1 expression studies of gastric cancer.

We detected a correlation between the NY-ESO-1 humoral immune response levels and the clinical outcome after therapy in gastric cancer patients. The patients who underwent surgery and did not suffer a subsequent relapse displayed consistent decreases in their NY-ESO-1 humoral immune response levels or even the complete disappearance of the NY-ESO-1 antibody from their sera. It is generally accepted that constant immunological stimulation is necessary to maintain a strong humoral immune response (Jager et al, 1999). Thus, reduction of antigen doses by the removal of NY-ESO-1-expressing tumour is one possible reason for the observed decreases in these patients’ NY-ESO-1 humoral immune response levels after surgery. Patients M-2 and M-11, in whom NY-ESO-1 humoral immune responses remained high for 1 year after surgery and increased thereafter, may have a subclinical residual disease of the so-called ‘minimal residual cancer’ (Austrup et al, 2000; Klein et al, 2002) after curative surgery. Local recurrent tumours of 23 and 25 mm in diameter subsequently developed in M-2 and M-11, respectively, suggesting that even a small tumour burden is sufficient to stimulate antibody production. Patient M-2 showed a partial decrease in their NY-ESO-1 humoral immune response levels after the resection of the relapsed tumour, and we are carefully observing the progression of this tumour.

Nine patients with stage IV gastric cancer received chemotherapy alone. Among them, six patients displayed stable disease, two patients displayed progressive disease, and one patient (M-19) achieved a partial response. Serial analysis of the NY-ESO-1 humoral immune responses of these nine patients including M-19 showed that they barely changed throughout their clinical courses, suggesting that even small tumours are enough to provoke strong NY-ESO-1 humoral immune responses. In this regard, the NY-ESO-1 humoral immune response might not be suitable as a clinical marker for palliative therapy.

We have performed serial cancer vaccine clinical trials with NY-ESO-1 because of its strong immunogenicity and high specificity (Uenaka et al, 2007; Wada et al, 2008; Kakimi et al, 2011). The NY-ESO-1humoral immune response could be a reliable marker of the induction of immune response, as well as for predicting clinical responses in these trials. Furthermore, antibody-based examinations detected both intra- and intermolecular antigen spreading in the sera of patients who had been vaccinated with NY-ESO-1 protein (Kawada et al, 2012), suggesting the possible correlation of NY-ESO-1 humoral immne responses and clinical status. In addition, we have started a phase I study of vaccination with NY-ESO-1 protein mixed with Hiltonol (Poly ICLC), Picibanil (OK-432), and Montanide (ISA-51) in patients with NY-ESO-1-expressing cancers (UMIN000007954). Furthermore, NY-ESO-1 vaccine involving modulators of immune checkpoints, for example, anti-CTLA4 antibody and anti-PD-1 antibody, and reagents that are antagonistic to regulatory T cells, for example, anti-CCR4 antibody (Pardoll, 2012) should be considered.

Recently, the antibody against p53, another tumour antigen, has been recognised as a useful tumour marker (Lubin et al, 1995). Shimada et al (2000)) reported that p53 antibody was detected in 35% of serum samples from patients with in situ oesophageal cancer and that it disappeared after endoscopic mucosal resection, proposing that p53 antibody is useful for the early detection and subsequent monitoring of oesophageal cancer. In addition, Müller et al (2006) reported that p53 antibody was found in 23.4% of serum samples from cancer patients with 100% accuracy and was correlated with poor prognosis in hepatocellular carcinoma and breast cancer.

Here, we have demonstrated that the NY-ESO-1 humoral immune response could also be valuable as a marker for detecting advanced gastric cancer and inferring whether residual tumour cells remain after treatment, although its frequency in gastric cancer is not very high. We have started a prospective multi-institutional clinical study of NY-ESO-1 humoral immune responses in higher stage gastric cancer patients. In this new study, the NY-ESO-1 humoral immune responses of approximately 100 patients who relapsed after curative surgery will be serially analysed and then followed up. This trial has been registered as UMIN000007925 in Japan.