Purpose: The development of local and distant recurrences is a major problem in the treatment of rectal cancer patients. In this study, we investigated whether epithelial human leukocyte antigen-DR (HLA-DR) expression allowed discrimination between high and low tumor recurrence rates, and analyzed the mechanism behind its expression.

Experimental Design: The role of IFNγ in HLA-DR expression was studied in rectal cancer cell lines and tumors by promoter-specific analyses of class II transactivator (CIITA). The predictive value of epithelial HLA-DR expression was investigated by immunohistochemical evaluation of 1,016 rectal tumors, obtained from a large prospective trial. Associations with recurrences and survival were determined by univariate and multivariate log-rank testing.

Results: HLA-DR was induced by IFNγ in rectal cancer cell lines. Activity of the IFNγ-inducible pIV-CIITA promoter correlated with epithelial HLA-DR expression in rectal tumors. Patients with HLA-DR–positive tumors developed less frequent local and distant recurrences [1.6% versus 9.1% (P = 0.0015) and 15.3% versus 29.9% (P < 0.0001), respectively, after 5 years of follow-up] and had better survival (78.6% versus 61.3%; P < 0.0001) than patients with HLA-DR–negative tumors. Epithelial HLA-DR was more often found in lower tumor-node-metastasis (TNM) stages. Next to TNM and circumferential resection margin, HLA-DR expression was independently associated with lower distant recurrence rates and prolonged survival.

Conclusions: Epithelial HLA-DR expression can be used as a marker to discriminate patients with high or low risk of developing recurrences. The possible involvement of IFNγ, the relationship with lower TNM stages, and the independent effect on recurrence development together suggest that the host immune response plays an important role in controlling tumor cells.

In the treatment of rectal cancer patients, local and distant recurrences are a major problem because these are associated with both high mortality and morbidity. The introduction of total mesorectal excision (TME) surgery in combination with preoperative radiotherapy has been shown to be useful in reducing local recurrences. However, more than 25% of the patients develop metastases within 5 years after surgical treatment with curative intent (1, 2).

We and others have shown that (colo)rectal cancer patients develop less frequent recurrences when high numbers of infiltrating immune cells are present in the tumor (36). An optimal antitumor immune response usually requires activation of CD4+ and CD8+ T lymphocytes with a tumor-associated antigen. CD8+ T cells are activated by antigen presented by human leukocyte antigen (HLA) class I molecules whereas CD4+ T cells are activated by antigen presented by HLA class II molecules, normally expressed on professional antigen-presenting cells such as dendritic cells and macrophages. We have recently shown that a subgroup of tumors expressed high levels of HLA classes I and II and several other immune-related genes. Importantly, most of these tumors expressed the HLA class II protein HLA-DR on epithelial cells (7). These findings suggest an important role for the host immune response in targeting tumor cells, implying a better prognosis for patients with tumors expressing epithelial HLA-DR. Indeed, most studies on colorectal cancer patients have shown that epithelial expression of HLA-DR relates to a better prognosis (811). Because of the limited amount of data for rectal cancer patients, it is unclear whether the prognostic value of HLA-DR observed in colorectal cancer patients (with 33-44% rectal cancer patients) can also be applied specifically to rectal cancer patients. We therefore analyzed the expression of epithelial HLA-DR in a large number of rectal tumors, obtained from a prospective trial randomizing surgery alone or short-term preoperative radiotherapy followed by surgery (1). The standardized high-quality surgery technique done in this trial decreased the number of recurrences due to inadequate surgery (12), and therefore allowed a reliable investigation of the influence of intrinsic tumor characteristics, such as HLA-DR expression, on recurrences and survival. In addition, we analyzed the involvement of IFNγ in epithelial HLA-DR expression in vivo.

Cell lines. Colon cancer cell line HT29, rectal cancer cell lines SW837 and SW1463, and Ramos B-cell line were all cultured in Iscove's modified Dulbecco's medium (Cambrex) supplemented with glutamine, penicillin/streptomycin, and 8% heat-inactivated FCS, at 37°C. Where indicated, exponentially growing cells were treated with 100 units/mL human recombinant IFNγ for the indicated time periods.

Visualization of HLA-DR on surface of cell lines. For these experiments, IFNγ-treated or untreated growing cells were harvested, washed, and resuspended in PBS supplemented with 0.5% bovine serum albumin and 0.05% sodium azide (PBA). Cells were incubated with a monoclonal FITC-labeled anti-HLA-DR antibody at 1:50 (BD Biosciences) for 30 min at 4°C. PBA alone was used as negative control. Antibody binding was detected with the use of a FACSCalibur (BD Biosciences). Propidium iodide was added to allow exclusion of dead cells.

Promoter-specific reverse transcription-PCR for class II transactivator. Isolation of RNA was done as previously described (13). Complementary DNA was synthesized from 1.0-μg aliquots of total RNA using random primers (Ambion) and SuperScript III (Invitrogen) according to the manufacturer's instructions. Class II transactivator (CIITA) products were amplified as previously described with the following primers: pIV-CIITA, sense 5′-AGCTGGCGGGAGGGAGAGGCCACC-3′ and antisense 5′-CATACTGGTCCAGTTCCGCGATATTGG-3′; pIII-CIITA, sense 5′-GATTCCTACACAATGCGTTGCCTGG-3′ and antisense as for pIV-CIITA (14). As a control, glyceraldehyde-3-phosphate dehydrogenase was amplified with the following primers: sense 5′-AAGGTGAAGGTCGGAGTCAAC-3′ and antisense 5′-TGGAAGATGGTGATGGGATT-3′.

Western blotting. Adherent cells were washed and scraped in 20 mmol/L Tris-HCl (pH 7.6), 137 mmol/L NaCl (TBS). Cells were lysed in cold cell lysis buffer (Cell Signaling Technology) supplemented with complete protease inhibitor cocktail (Roche Applied Science). After clearing the lysates by high-speed centrifugation, protein concentrations were determined with bicinchoninic acid protein assay (Pierce Biotechnology). Equal amounts of total protein were resolved on SDS-PAGE gels and blotted on Hybond-P (Amersham). Ponceau S stainings were done to verify protein quantities transferred onto the membranes. Membranes were blocked with 5% nonfat dry milk in TBS supplemented with 0.2% Tween 20 (TBS-T) for 2 h. Primary antibody incubations were carried out in 1% milk/TBS-T overnight at 4°C and followed by three washes with TBS-T. Membranes were incubated with antirabbit IgG-horseradish peroxidase conjugates (Amersham) for 45 min, washed thrice, and developed with ECL Plus reagents (Amersham). The primary antibodies used were rabbit monoclonal antibodies recognizing human phospho-Tyr701 signal tranducer and activator of transcription 1 (STAT1; Cell Signaling Technology) at 1:3,000; STAT1 at 1:1,000 (Cell Signaling Technology); IFN regulatory factor 1 (IRF1) at 1:1,000 (C20; Santa Cruz Biotechnology); and extracellular signal–regulated kinase 2 (kindly provided by B. Burgering, University Medical Center Utrecht, Utrecht, the Netherlands).

Patients and tumor samples. Tumor samples were obtained at the time of surgery from rectal cancer patients participating in the Dutch TME trial, a large multicenter trial in which patients were randomized for short-term radiotherapy (5 × 5 Gy) followed by TME surgery, or TME surgery alone (1). Radiotherapeutic, surgical, and pathologic procedures were standardized and quality controlled. Tumor staging was determined with the tumor-node-metastasis (TNM) classification (15). Paraffin-embedded tumors (1,047) were available in tissue microarray format, as previously described (16). Patients who were ineligible (n = 25) or TNM stage 0 (n = 6) were excluded from the analyses, leaving 1,016 patients for evaluation of the prognostic value of HLA-DR expression. The median follow-up time of these patients was 68 months.

Immunohistochemistry. Stainings were done as previously described (7). Briefly, 4-μm sections were deparaffinized and endogenous peroxidise activity was blocked with 0.3% hydrogen peroxide in methanol for 30 min. Antigen retrieval was done by boiling for 10 min in 0.01 mol/L citrate buffer (pH 6.0). Preincubation with 20% normal goat serum in PBS/1% bovine serum albumin for 30 min was followed by overnight incubation with the prediluted monoclonal HLA-DR antibody (LN3; Signet Laboratories). Sections were incubated for 1 h with biotinylated goat anti-mouse (1:200; DakoCytomation) followed by incubation with streptavidin complexed with biotinylated peroxidase (DakoCytomation) and developed with 3,3-diaminobenzidine (DakoCytomation). Finally, sections were counterstained with Mayer's hematoxylin. As a negative control, PBS alone was used in place of the primary antibody.

Scoring. For each tumor, one 2-mm-diameter punch was evaluated. Epithelial HLA-DR stainings were scored by determining the percentage of positively stained tumor epithelium. Most tumors (n = 674; 66%) were completely negative for epithelial HLA-DR, 126 (12%) tumors showed <20% positive epithelium, 52 (5%) tumors showed 20% to 50%, and 164 (16%) tumors showed ≥50% positive epithelium. To analyze the correlation between different variables, tumors were regarded positive if at least 20% of the epithelial cells stained positive for HLA-DR. This retrospectively selected cutoff value was chosen to ensure an adequate sample size in the HLA-DR–positive group. Analyses were repeated with ≥5% and ≥50% as cutoff values to show that results were independent of cutoff value.

Statistical analysis. All analyses were done with SPSS statistical software (version 12.0 for Windows, SPSS, Inc.). Data were analyzed with Mann-Whitney or Kruskal-Wallis tests to compare quantitative and ordered variables and with Student's t tests to compare normally distributed data between two groups. χ2 tests were used to compare proportions. A two-sided P value of ≤0.05 was considered statistically significant.

Recurrence and overall survival analyses were done using the Kaplan-Meier method with time of surgery as entry date. Log-rank testing was used for comparison of groups. To show survival curves for the HLA-DR–positive and HLA-DR–negative patients that account for variables associated with HLA-DR expression, we carried out a stratified Cox regression analysis with TNM, circumferential resection margin (CRM), and treatment as covariates in the model and HLA-DR status as stratum variable. In this way, the curves were adjusted for differences between the HLA-DR–positive and HLA-DR–negative patients with respect to the other variables, but the curves were not forced to have the same shape, as is the case in the ordinary Cox regression model.

For multivariate analyses, variables associated with HLA-DR expression or with significant effect on either recurrence or survival in univariate analyses (P < 0.10) were further examined using the Cox proportional hazards regression model with backward stepwise elimination. Variables were eliminated if P > 0.10.

Role of IFNγ in epithelial HLA-DR expression in rectal cancer cell lines. We have recently shown, following gene expression analysis on 47 nonirradiated rectal tumors, that a subgroup of tumors expressed HLA-DR, a member of the HLA class II proteins, in epithelial cells (7). To obtain insight into the mechanism of epithelial HLA-DR expression, we first tested whether IFNγ is able to induce HLA-DR expression in vitro in rectal cancer cells, as has been described for colon cancer cells (17). Indeed, fluorescence-activated cell sorting analysis showed up-regulated surface expression of HLA-DR following treatment with IFNγ. Specifically, IFNγ induced HLA-DR expression in SW1463 cells, which do not normally express HLA-DR, and markedly enhanced the HLA-DR expression in SW837 cells above the endogenous expression (Fig. 1A).

Fig. 1.

Effect of IFNγ on HLA-DR expression in tumor cells in vitro. A, quantitative comparison of surface HLA-DR expression of rectal cancer cell lines SW1463 and SW837; HT29 (colon) and Ramos (B cells) were used as controls. Cells were cultured for 48 h in the absence (gray line) or presence (black line) of IFNγ 100 units/mL. Gray fills indicate negative PBA control. Results are representative of four independent experiments. B, Western blot of whole-cell lysates of SW1463 and SW837 cells, harvested at the indicated time points after addition of IFNγ (100 units/mL). Blots were analyzed for STAT1 phosphorylation and reprobed to determine for STAT1 expression (top), or analyzed for IRF1 expression and reprobed for extracellular signal–regulated kinase 2 (Erk2) expression as loading control. Results are representative of three independent experiments.

Fig. 1.

Effect of IFNγ on HLA-DR expression in tumor cells in vitro. A, quantitative comparison of surface HLA-DR expression of rectal cancer cell lines SW1463 and SW837; HT29 (colon) and Ramos (B cells) were used as controls. Cells were cultured for 48 h in the absence (gray line) or presence (black line) of IFNγ 100 units/mL. Gray fills indicate negative PBA control. Results are representative of four independent experiments. B, Western blot of whole-cell lysates of SW1463 and SW837 cells, harvested at the indicated time points after addition of IFNγ (100 units/mL). Blots were analyzed for STAT1 phosphorylation and reprobed to determine for STAT1 expression (top), or analyzed for IRF1 expression and reprobed for extracellular signal–regulated kinase 2 (Erk2) expression as loading control. Results are representative of three independent experiments.

Close modal

IFNγ induces HLA-DR through the STAT1 signaling pathway, which promotes transcription of IRF1, leading to transcription of CIITA (18). Western blot analyses of rectal cancer cells treated with IFNγ indeed showed rapid phosphorylation of STAT1 and up-regulation of IRF1 after 6 h on treatment (Fig. 1B). Together, the data show that IFNγ is able to induce epithelial HLA-DR expression in these rectal cancer cells, and therefore suggest that epithelial HLA-DR expression in tumors might be driven by IFNγ.

Role of IFNγ in epithelial HLA-DR expression in rectal tumors. To investigate the role of IFNγ in epithelial HLA-DR expression in vivo, we examined the promoter activity of CIITA in rectal tumors. CIITA is controlled by four independent promoters, of which promoter pIV has been described to regulate IFNγ-induced CIITA expression, and pIII to regulate constitutive CIITA expression such as in B cells and some melanomas (1921). PCR analyses, with promoter-specific primers, enabled us to determine the promoter activity in rectal tumors and showed both active pIII and pIV in HLA-DR–positive tumors. Whereas pIII-CIITA was detected in some HLA-DR–negative tumors, activity of pIV-CIITA could not be found in HLA-DR–negative tumors (Fig. 2, left). In addition, analysis of IFNγ-treated cells showed that both promoters, pIII and pIV, were activated on IFNγ treatment. Although SW837 displayed pIV-derived CIITA in the absence of IFNγ, its activity was markedly enhanced on IFNγ treatment (Fig. 2, right). Together, these observations suggest that IFNγ is likely involved in epithelial HLA-DR expression in vivo.

Fig. 2.

CIITA promoter activity in tumors in vivo and on IFNγ treatment in vitro. A, complementary DNA of seven HLA-DR–positive and seven HLA-DR–negative tumor samples was analyzed for CIITA promoter activity by PCR using primers specific for pIII (top) or pIV (middle). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH; bottom) served as control for amounts of cDNA. Results are representative of three independent experiments. B, cell lines cultured in the absence (−) or presence (+) of IFNγ (100 units/mL) for 48 h were analyzed for CIITA promoter activity by PCR. mRNA expression of pIII-CIITA (top), pIV-CIITA (middle), and glyceraldehyde-3-phosphate dehydrogenase (bottom). Results are representative of three independent experiments.

Fig. 2.

CIITA promoter activity in tumors in vivo and on IFNγ treatment in vitro. A, complementary DNA of seven HLA-DR–positive and seven HLA-DR–negative tumor samples was analyzed for CIITA promoter activity by PCR using primers specific for pIII (top) or pIV (middle). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH; bottom) served as control for amounts of cDNA. Results are representative of three independent experiments. B, cell lines cultured in the absence (−) or presence (+) of IFNγ (100 units/mL) for 48 h were analyzed for CIITA promoter activity by PCR. mRNA expression of pIII-CIITA (top), pIV-CIITA (middle), and glyceraldehyde-3-phosphate dehydrogenase (bottom). Results are representative of three independent experiments.

Close modal

Correlation between epithelial HLA-DR expression and patients' characteristics. We previously analyzed 47 rectal cancer patients for epithelial HLA-DR expression and showed epithelial HLA-DR expression in 9 tumors (7). Interestingly, none of the patients with HLA-DR–positive tumors developed recurrences after 3 years of follow-up, whereas 19% of the patients with HLA-DR–negative tumors developed recurrences within this period of time. The number of patients is obviously too small to provide reliable data. To further investigate this, we evaluated HLA-DR expression in 1,016 paraffin-embedded rectal tumors obtained from a prospective trial randomizing for preoperative radiotherapy, followed by surgery. Most tumors (66%) did not show any HLA-DR expression in the epithelial compartment. These were grouped together with the 12% of tumors that showed <20% positivity and considered HLA-DR negative. The 216 (21%) tumors that expressed HLA-DR in at least 20% of the epithelial cells were considered HLA-DR positive.

HLA-DR expression in relation to clinicopathologic variables is shown in Table 1. Epithelial HLA-DR expression was associated with lower TNM stages, and HLA-DR–positive tumors had significantly more negative CRMs. HLA-DR–positive tumors were more often found in irradiated patients, but analysis with the percentage of HLA-DR–positive cells as a continuous variable revealed no significant difference between irradiated and nonirradiated patients (P = 0.137). The association with TNM stages and CRM remained statistically significant (P < 0.001 and P = 0.015, respectively).

Table 1.

Clinical and pathologic characteristics of patients with HLA-DR–positive and HLA-DR–negative tumors

HLA-DRn (%)HLA-DR+n (%)P
Age (median) 65 65 0.911 
Sex    
    Female 277 (35) 84 (39) 0.245 
    Male 523 (65) 132 (61)  
Treatment    
    TME 430 (54) 97 (45) 0.021 
    RT + TME 370 (46) 119 (55)  
TNM stage    
    I 209 (26) 96 (44) <0.001 
    II 226 (28) 67 (31)  
    III 307 (38) 49 (23)  
    IV 58 (7) 4 (2)  
Differentiation    
    Well 38 (5) 8 (4) 0.675 
    Moderate 528 (66) 143 (66)  
    Poor/undif 233 (29) 65 (30)  
Distance to anal verge, cm    
    ≥10 227 (28) 54 (25) 0.229 
    5-10 333 (42) 87 (41)  
    <5 238 (30) 72 (34)  
Operation type    
    LAR 520 (65) 137 (63) 0.516 
    APR 234 (29) 71 (33)  
    Hartmann 45 (6) 8 (4)  
CRM    
    Negative 647 (81) 191 (88) 0.033 
    Positive 153 (19) 25 (12)  
HLA-DRn (%)HLA-DR+n (%)P
Age (median) 65 65 0.911 
Sex    
    Female 277 (35) 84 (39) 0.245 
    Male 523 (65) 132 (61)  
Treatment    
    TME 430 (54) 97 (45) 0.021 
    RT + TME 370 (46) 119 (55)  
TNM stage    
    I 209 (26) 96 (44) <0.001 
    II 226 (28) 67 (31)  
    III 307 (38) 49 (23)  
    IV 58 (7) 4 (2)  
Differentiation    
    Well 38 (5) 8 (4) 0.675 
    Moderate 528 (66) 143 (66)  
    Poor/undif 233 (29) 65 (30)  
Distance to anal verge, cm    
    ≥10 227 (28) 54 (25) 0.229 
    5-10 333 (42) 87 (41)  
    <5 238 (30) 72 (34)  
Operation type    
    LAR 520 (65) 137 (63) 0.516 
    APR 234 (29) 71 (33)  
    Hartmann 45 (6) 8 (4)  
CRM    
    Negative 647 (81) 191 (88) 0.033 
    Positive 153 (19) 25 (12)  

Abbreviations: RT, radiotherapy; undif, undifferentiated; LAR, low anterior resection; APR, abdominoperineal resection.

Epithelial HLA-DR expression is related to a better prognosis for rectal cancer patients. The association between epithelial HLA-DR expression in tumors and the development of recurrences in the cancer patients is displayed in Fig. 3. Local recurrence rates were significantly lower for patients with HLA-DR–positive tumors compared with patients with HLA-DR–negative tumors: 1.6% versus 9.1% at 5 years of follow-up (P = 0.0015; Fig. 3A, left). A similar result was found for distant recurrence rates: 15.3% versus 29.9% (P < 0.0001; Fig. 3A, middle). The lower number of recurrences resulted in prolonged overall survival for patients with epithelial HLA-DR–positive tumors when compared with patients with HLA-DR–negative tumors: 78.6% versus 61.3% (P < 0.0001) of the patients were still alive at 5 years of surgery (Fig. 3A, right). In this study, tumors were classified as being HLA-DR positive if at least 20% of the epithelial tumor cells stained positive. To evaluate the consistency of the data, the analyses have been repeated with the cutoff values ≥5% and ≥50%. These analyses showed similar results (i.e. significantly lower recurrence rates and better survival for patients with HLA-DR–positive tumors).

Fig. 3.

Prognostic values of epithelial HLA-DR expression for tumor recurrences and survival. Local (left) and distant (middle) recurrence rates and overall survival (right) for patients with HLA-DR–positive tumors (black) and patients with HLA-DR–negative tumors (gray). A, Kaplan-Meier curves obtained with univariate log-rank analysis; B, curves obtained with Cox regression analysis that account for TNM, CRM, and treatment. The N below the plots indicates the absolute number of patients at risk.

Fig. 3.

Prognostic values of epithelial HLA-DR expression for tumor recurrences and survival. Local (left) and distant (middle) recurrence rates and overall survival (right) for patients with HLA-DR–positive tumors (black) and patients with HLA-DR–negative tumors (gray). A, Kaplan-Meier curves obtained with univariate log-rank analysis; B, curves obtained with Cox regression analysis that account for TNM, CRM, and treatment. The N below the plots indicates the absolute number of patients at risk.

Close modal

The curves in Fig. 3A showed that epithelial HLA-DR can discriminate patients with high or low recurrence risks. Nevertheless, several important variables favor the HLA-DR–positive group (Table 1). Curves that are corrected for the effects of TNM, CRM, and treatment are displayed in Fig. 3B. These curves also showed better prognosis for patients with HLA-DR–positive tumors.

Stratified analysis based on the treatment regimen revealed that both nonirradiated and irradiated patients had lower distant recurrence rates and better survival when the tumor stained positive for HLA-DR (Table 2). We also observed better local control for HLA-DR–positive tumors in irradiated patients (P = 0.011), and a similar trend in nonirradiated patients (P = 0.064). Interestingly, none of the irradiated patients with HLA-DR–positive tumors developed local recurrences within 5 years of surgery. Stratification based on TNM showed a comparable trend for TNM stage I/II and TNM stage III/IV tumors: lower recurrence rates and better survival for the HLA-DR–positive group. Although the predictive value of epithelial HLA-DR on recurrence rates was not statistically significant for the CRM-positive tumors (local recurrences, P = 0.11; distant recurrences, P = 0.10), it was associated with a better survival (P = 0.01) for this group of patients. For the CRM-negative tumors, positive HLA-DR was significantly associated with lower recurrence rates and prolonged survival.

Table 2.

Prognostic values of epithelial HLA-DR expression for local and distant recurrences and survival on stratification for treatment, TNM, or CRM

HLA-DRLocal recurrences
Distant recurrences
Overall survival
PosNegPHR (95% CI)PosNegPHR (95% CI)PosNegPHR (95% CI)
Overall 1.6 9.5 0.002 3.9 (1.6-9.7) 15.3 29.9 <0.0001 2.1 (1.5-3.1) 78.6 61.3 <0.0001 1.8 (1.4-2.4) 
TME 3.5 10.6 0.06 2.5 (0.9-7.1) 14.6 30.2 0.005 2.1 (1.2-3.8) 79.4 61.7 0.006 1.7 (1.2-2.5) 
RT + TME 0.0 7.2 0.01 8.7 (1.1-64.5) 16.0 29.5 0.002 2.1 (1.3-3.5) 77.9 60.9 0.0002 2.0 (1.4-2.8) 
TNM I/II 0.0 4.1 0.08 3.4 (0.8-14.6) 10.7 15.9 0.15 1.5 (0.9-2.5) 81.5 74.8 0.07 1.4 (1.0-1.9) 
TNM III/IV 4.8 16.1 0.07 2.8 (0.9-9.0) 30.4 49.4 0.01 1.9 (1.1-3.3) 69.4 45.4 0.004 1.9 (1.2-2.9) 
CRM 1.2 7.3 0.01 3.5 (1.2-9.7) 12.1 25.1 0.0005 2.2 (1.4-3.3) 80.0 66.7 0.0004 1.7 (1.3-2.2) 
CRM+ 4.3 18.6 0.11 4.5 (0.6-33.6) 38.9 53.2 0.10 1.8 (0.9-3.6) 67.9 38.8 0.01 2.1 (1.2-3.8) 
HLA-DRLocal recurrences
Distant recurrences
Overall survival
PosNegPHR (95% CI)PosNegPHR (95% CI)PosNegPHR (95% CI)
Overall 1.6 9.5 0.002 3.9 (1.6-9.7) 15.3 29.9 <0.0001 2.1 (1.5-3.1) 78.6 61.3 <0.0001 1.8 (1.4-2.4) 
TME 3.5 10.6 0.06 2.5 (0.9-7.1) 14.6 30.2 0.005 2.1 (1.2-3.8) 79.4 61.7 0.006 1.7 (1.2-2.5) 
RT + TME 0.0 7.2 0.01 8.7 (1.1-64.5) 16.0 29.5 0.002 2.1 (1.3-3.5) 77.9 60.9 0.0002 2.0 (1.4-2.8) 
TNM I/II 0.0 4.1 0.08 3.4 (0.8-14.6) 10.7 15.9 0.15 1.5 (0.9-2.5) 81.5 74.8 0.07 1.4 (1.0-1.9) 
TNM III/IV 4.8 16.1 0.07 2.8 (0.9-9.0) 30.4 49.4 0.01 1.9 (1.1-3.3) 69.4 45.4 0.004 1.9 (1.2-2.9) 
CRM 1.2 7.3 0.01 3.5 (1.2-9.7) 12.1 25.1 0.0005 2.2 (1.4-3.3) 80.0 66.7 0.0004 1.7 (1.3-2.2) 
CRM+ 4.3 18.6 0.11 4.5 (0.6-33.6) 38.9 53.2 0.10 1.8 (0.9-3.6) 67.9 38.8 0.01 2.1 (1.2-3.8) 

NOTE: Five-year recurrence rates and survival for patients with HLA-DR–positive tumors (pos) and HLA-DR–negative tumors (neg) and P values were determined by univariate log-rank analyses. Hazard ratios (HR) with 95% confidence intervals (95% CI) were determined by Cox regression analysis with HLA-DR as covariate.

The independent predictive value for HLA-DR was tested in multivariate Cox regression analysis with backward stepwise elimination in the context of the following variables: CRM, TNM stage, differentiation, distance to anal verge, and treatment. No significant interactions were observed between the variables. Although statistically not significant, epithelial HLA-DR expression tended to be predictive for local recurrences (hazard ratio, 2.4; P = 0.07) in addition to CRM, TNM, and treatment. Importantly, HLA-DR expression on epithelial tumor cells had independent prognostic value for distant recurrence rate (hazard ratio, 1.6; P = 0.01) and survival (hazard ratio, 1.4; P = 0.01) in additional to TNM and CRM (Table 3).

Table 3.

Prognostic values of epithelial HLA-DR expression for distant and local recurrences and survival in multivariate analysis

HR (95% CI)
Local recurrenceDistant recurrenceSurvival
HLA-DR    
    Negative 2.4 (0.9-6.0) 1.6 (1.1-2.4) 
    Positive 1.4 (1.1-1.9) 
CRM    
    Negative 1.7 (1.3-2.1) 
    Positive 1.9 (1.1-3.3) 1.8 (1.3-2.4) 
TNM stage    
    I 3.0 (2.2-4.0) 
    II 3.9 (1.3-11.7) 2.5 (1.6-4.1) 1.3 (1.1-1.7) 
    III 8.2 (2.9-23.4) 6.4 (4.1-10.0) 
Distance to anal verge, cm    
    ≥10 ns ns 
    5-10  1.4 (1.0-2.0)  
    <5  1.8 (1.3-2.5)  
Treatment    
    TME 1.8 (1.0-3.0) ns ns 
    RT + TME   
HR (95% CI)
Local recurrenceDistant recurrenceSurvival
HLA-DR    
    Negative 2.4 (0.9-6.0) 1.6 (1.1-2.4) 
    Positive 1.4 (1.1-1.9) 
CRM    
    Negative 1.7 (1.3-2.1) 
    Positive 1.9 (1.1-3.3) 1.8 (1.3-2.4) 
TNM stage    
    I 3.0 (2.2-4.0) 
    II 3.9 (1.3-11.7) 2.5 (1.6-4.1) 1.3 (1.1-1.7) 
    III 8.2 (2.9-23.4) 6.4 (4.1-10.0) 
Distance to anal verge, cm    
    ≥10 ns ns 
    5-10  1.4 (1.0-2.0)  
    <5  1.8 (1.3-2.5)  
Treatment    
    TME 1.8 (1.0-3.0) ns ns 
    RT + TME   

NOTE: P values and hazard ratios with 95% confidence intervals were determined by Cox regression analysis. “ns” indicates that this variable was not statistically significant in the backward stepwise logistic regression analysis and was therefore eliminated.

In this study, we found that HLA-DR was expressed on epithelial cells in ∼20% of rectal tumors, and showed that rectal cancer patients with HLA-DR–positive tumors had better survival than patients with HLA-DR–negative tumors. In addition, epithelial HLA-DR expression correlated with a reduction in both local and distant recurrence rates.

The expression of HLA-DR was inducible by IFNγ in rectal cancer cell lines, which is consistent with published data for colon cancer cell lines (17). The influence of IFNγ on HLA-DR expression in vivo was previously suggested following detection of IFNγ mRNA in HLA-DR–positive tumors (11). Nevertheless, the presence of IFNγ can also be a consequence of epithelial HLA-DR because HLA-DR–expressing tumor cells recruit and activate CD4+ T helper 1 and natural killer cells, resulting in local production of IFNγ (22, 23). To obtain more insight into HLA-DR expression in vivo, we analyzed CIITA promoter activity and showed that activity of pIV correlated with HLA-DR expression, in contrast to that of pIII, which is involved in constitutive expression (20, 21). This suggests that epithelial HLA-DR expression in vivo is regulated by CIITA via the IFNγ-inducible pIV promoter. It therefore follows that HLA-DR expression could result from the presence of IFNγ.

Epithelial HLA-DR expression as prognostic factor has previously been described for colorectal cancer patients (811). However, stratified analyses according to tumor location revealed better prognosis for HLA-DR–positive tumors in colon cancer patients but not in rectal cancer patients (9). The prognostic value of HLA-DR obtained in colorectal patients might be skewed by its association with microsatellite instability (9), which is a rare event in rectal tumors (24, 25). To our knowledge, this is the first comprehensive study investigating a correlation between epithelial HLA-DR expression and survival of rectal cancer patients. The high number of patients studied and the standardized high-quality TME surgery enabled us to show remarkably lower recurrence rates and better survival for rectal cancer patients with HLA-DR–positive tumors. Interestingly, the results did not depend on the cutoff value because similar results were obtained with the cutoff values ≥5%, ≥20%, and ≥50% HLA-DR–positive epithelium.

Several mechanisms have been proposed to explain the good prognosis for colorectal cancer patients with HLA-DR–positive tumors. A role for IFNγ has been described, as it affects multiple genes involved in apoptosis, cell growth, and genetic instability (10, 2628). Other studies focused on immunologic tumor control. Animal experiments showed that CIITA-transfected tumor cells were rejected via activation of the host immune response (22, 23). In vitro experiments with human tumor cells also showed enhanced immunogenictiy on (IFNγ-induced) expression of CIITA (29, 30). In agreement with this, we and others have shown that high amounts of tumor infiltrating lymphocytes correlate with a better prognosis in (colo)rectal cancer patients (3, 4, 6). In our study, HLA-DR–positive tumors were more often found in low TNM stages. This might be explained by reduced proliferation of HLA-DR–positive cells (10), as well as by immunologic control of the primary tumor. The less frequent distant recurrence development supports the hypothesis that HLA-DR–positive tumor cells exert an immunologic control. Whereas HLA-DR expression independently predicted lower distant recurrence rates and prolonged survival, its predictive value for local recurrences was not statistically significant in the context of TNM, CRM, and treatment. This loss of significance indicates different regulation of local versus distant recurrences. Whereas the immune response plays an important role in preventing HLA-DR–positive cells from metastasizing to lymph nodes or other organs, local control also depends on other mechanisms, such as the effect of radiotherapy.

TNM stage, CRM, and radiotherapy were independently associated with local control, whereas HLA-DR only showed a trend toward better local control. These data indicate that radiation and a negative CRM could be more important factors than epithelial HLA-DR expression in preventing local recurrences. The role of HLA-DR expression may, however, be underestimated due to the small number of events.

In conclusion, epithelial HLA-DR expression is associated with a reduced recurrence rates and better survival. Because this expression is an intrinsic tumor characteristic, evaluation of HLA-DR expression can be used to identify patients with high risk for disease recurrences who need adjuvant treatment (31). In addition, we showed that epithelial HLA-DR expression in rectal tumors is likely to result from the presence of IFNγ. It is also known that HLA-DR expression enhances the immune system and results in local IFNγ production. This implies a self-amplifying system between HLA-DR and IFNγ. If this is the case, then the induction of HLA-DR could be a promising therapeutic strategy. A recently published phase II trial in metastatic melanoma patients showed a long-lasting expression of HLA-DR on IFNγ treatment (32), indicating that up-regulation of HLA-DR can indeed be achieved by administration of IFNγ in vivo. In these melanoma patients, HLA-DR induction did not correlate with tumor regression. However, melanomas are not directly comparable to rectal tumors because they are known to be highly immunogenic and have several mechanisms to escape immune destruction (26, 3338). It remains to be elucidated whether treatment-induced HLA-DR expression will improve prognosis to comparable levels as endogenous epithelial HLA-DR expression for rectal cancer patients. In light of our study, future investigation into the therapeutic benefit of HLA-DR induction for the treatment of rectal cancer patients is warranted.

Grant support: Dutch Cancer Society grant RUL 2002-2733.

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.

Note: C.A.M. Marijnen and J.P. Medema share senior authorship.

We thank Peter van den Elsen (Leiden University Medical Center, Leiden, the Netherlands) for kindly providing the CIITA promoter-specific primers, and Fiona Kimberley (LEXOR, Academic Medical Center, the Netherlands) for critical reading of the manuscript.

1
Kapiteijn E, Marijnen CA, Nagtegaal ID, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer.
N Engl J Med
2001
;
345
:
638
–46.
2
Peeters KC, Marijnen CA, Nagtegaal ID, et al. The TME trial after a median follow-up of 5 years: increased local control but no survival benefit in irradiated patients with resectable rectal carcinoma.
Ann Surg
2007
;
246
:
693
–701.
3
Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome.
Science
2006
;
313
:
1960
–4.
4
Nagtegaal ID, Marijnen CA, Kranenbarg EK, et al. Local and distant recurrences in rectal cancer patients are predicted by the nonspecific immune response; specific immune response has only a systemic effect—a histopathological and immunohistochemical study.
BMC Cancer
2001
;
1
:
7
.
5
Naito Y, Saito K, Shiiba K, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer.
Cancer Res
1998
;
58
:
3491
–4.
6
Pages F, Berger A, Camus M, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer.
N Engl J Med
2005
;
353
:
2654
–66.
7
de Bruin EC, van de PS, van de Velde CJ, et al. Gene expression profiling reveals two separate mechanisms regulating apoptosis in rectal carcinomas in vivo.
Apoptosis
2007
;
12
:
1671
–80.
8
Andersen SN, Rognum TO, Lund E, Meling GI, Hauge S. Strong HLA-DR expression in large bowel carcinomas is associated with good prognosis.
Br J Cancer
1993
;
68
:
80
–5.
9
Lovig T, Andersen SN, Thorstensen L, et al. Strong HLA-DR expression in microsatellite stable carcinomas of the large bowel is associated with good prognosis.
Br J Cancer
2002
;
87
:
756
–62.
10
Matsushita K, Takenouchi T, Shimada H, et al. Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: Possible involvement of c-myc suppression by interferon-γ in situ.
Cancer Sci
2006
;
97
:
57
–63.
11
Matsushita K, Takenouchi T, Kobayashi S, et al. HLA-DR antigen expression in colorectal carcinomas: influence of expression by IFN-γ in situ and its association with tumour progression.
Br J Cancer
1996
;
73
:
644
–8.
12
Kapiteijn E, Putter H, van de Velde CJ. Impact of the introduction and training of total mesorectal excision on recurrence and survival in rectal cancer in the Netherlands.
Br J Surg
2002
;
89
:
1142
–9.
13
de Bruin EC, van de PS, Lips EH, et al. Macrodissection versus microdissection of rectal carcinoma: minor influence of stroma cells to tumor cell gene expression profiles.
BMC Genomics
2005
;
6
:
142
.
14
van der Stoep N, Biesta P, Quinten E, van den Elsen PJ. Lack of IFN-γ-mediated induction of the class II transactivator (CIITA) through promoter methylation is predominantly found in developmental tumor cell lines.
Int J Cancer
2002
;
97
:
501
–7.
15
Quirke P, Durdey P, Dixon MF, Williams NS. Local recurrence of rectal adenocarcinoma due to inadequate surgical resection. Histopathological study of lateral tumour spread and surgical excision.
Lancet
1986
;
2
:
996
–9.
16
de Bruin EC, van de Velde CJ, van de PS, et al. Prognostic value of apoptosis in rectal cancer patients of the dutch total mesorectal excision trial: radiotherapy is redundant in intrinsically high-apoptotic tumors.
Clin Cancer Res
2006
;
12
:
6432
–6.
17
Satoh A, Toyota M, Ikeda H, et al. Epigenetic inactivation of class II transactivator (CIITA) is associated with the absence of interferon-γ-induced HLA-DR expression in colorectal and gastric cancer cells.
Oncogene
2004
;
23
:
8876
–86.
18
Muhlethaler-Mottet A, Di Berardino W, Otten LA, Mach B. Activation of the MHC class II transactivator CIITA by interferon-γ requires cooperative interaction between Stat1 and USF-1.
Immunity
1998
;
8
:
157
–66.
19
Deffrennes V, Vedrenne J, Stolzenberg MC, et al. Constitutive expression of MHC class II genes in melanoma cell lines results from the transcription of class II transactivator abnormally initiated from its B cell-specific promoter.
J Immunol
2001
;
167
:
98
–106.
20
Muhlethaler-Mottet A, Otten LA, Steimle V, Mach B. Expression of MHC class II molecules in different cellular and functional compartments is controlled by differential usage of multiple promoters of the transactivator CIITA.
EMBO J
1997
;
16
:
2851
–60.
21
Reith W, LeibundGut-Landmann S, Waldburger JM. Regulation of MHC class II gene expression by the class II transactivator.
Nat Rev Immunol
2005
;
5
:
793
–806.
22
Meazza R, Comes A, Orengo AM, Ferrini S, Accolla RS. Tumor rejection by gene transfer of the MHC class II transactivator in murine mammary adenocarcinoma cells.
Eur J Immunol
2003
;
33
:
1183
–92.
23
Mortara L, Castellani P, Meazza R, et al. CIITA-induced MHC class II expression in mammary adenocarcinoma leads to a Th1 polarization of the tumor microenvironment, tumor rejection, and specific antitumor memory.
Clin Cancer Res
2006
;
12
:
3435
–43.
24
Ikeda Y, Oda S, Abe T, Ohno S, Maehara Y, Sugimachi K. Features of microsatellite instability in colorectal cancer: comparison between colon and rectum.
Oncology
2001
;
61
:
168
–74.
25
Fernebro E, Bendahl PO, Dictor M, Persson A, Ferno M, Nilbert M. Immunohistochemical patterns in rectal cancer: application of tissue microarray with prognostic correlations.
Int J Cancer
2004
;
111
:
921
–8.
26
Reu FJ, Bae SI, Cherkassky L, et al. Overcoming resistance to interferon-induced apoptosis of renal carcinoma and melanoma cells by DNA demethylation.
J Clin Oncol
2006
;
24
:
3771
–9.
27
Kroger A, Stirnweiss A, Pulverer JE, et al. Tumor suppression by IFN regulatory factor-1 is mediated by transcriptional down-regulation of cyclin D1.
Cancer Res
2007
;
67
:
2972
–81.
28
Xiao S, Li D, Zhu HQ, et al. RIG-G as a key mediator of the antiproliferative activity of interferon-related pathways through enhancing p21 and p27 proteins.
Proc Natl Acad Sci U S A
2006
;
103
:
16448
–53.
29
Thompson JA, Dissanayake SK, Ksander BR, Knutson KL, Disis ML, Ostrand-Rosenberg S. Tumor cells transduced with the MHC class II Transactivator and CD80 activate tumor-specific CD4+ T cells whether or not they are silenced for invariant chain.
Cancer Res
2006
;
66
:
1147
–54.
30
Schmitz-Winnenthal FH, Escobedo LV, Beckhove P, et al. Specific immune recognition of pancreatic carcinoma by patient-derived CD4 and CD8 T cells and its improvement by interferon-γ.
Int J Oncol
2006
;
28
:
1419
–28.
31
Wolpin BM, Meyerhardt JA, Mamon HJ, Mayer RJ. Adjuvant treatment of colorectal cancer.
CA Cancer J Clin
2007
;
57
:
168
–85.
32
Propper DJ, Chao D, Braybrooke JP, et al. Low-dose IFN-γ induces tumor MHC expression in metastatic malignant melanoma.
Clin Cancer Res
2003
;
9
:
84
–92.
33
van Houdt IS, Oudejans JJ, van den Eertwegh AJ, et al. Expression of the apoptosis inhibitor protease inhibitor 9 predicts clinical outcome in vaccinated patients with stage III and IV melanoma.
Clin Cancer Res
2005
;
11
:
6400
–7.
34
Medema JP, de Jong J, Peltenburg LT, et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors.
Proc Natl Acad Sci U S A
2001
;
98
:
11515
–20.
35
Irmler M, Thome M, Hahne M, et al. Inhibition of death receptor signals by cellular FLIP.
Nature
1997
;
388
:
190
–5.
36
Bullani RR, Wehrli P, Viard-Leveugle I, et al. Frequent down-regulation of Fas (CD95) expression and function in melanoma.
Melanoma Res
2002
;
12
:
263
–70.
37
Hahne M, Rimoldi D, Schroter M, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape.
Science
1996
;
274
:
1363
–6.
38
Bullani RR, Huard B, Viard-Leveugle I, et al. Selective expression of FLIP in malignant melanocytic skin lesions.
J Invest Dermatol
2001
;
117
:
360
–4.