Experimental evidence suggests that the let-7 family of noncoding RNAs suppresses adaptive immune responses, contributing to immune evasion by the tumor. We hypothesized that the amount of let-7a and let-7b expression in colorectal carcinoma might be associated with limited T-lymphocyte infiltrates in the tumor microenvironment and worse clinical outcome. Utilizing the molecular pathological epidemiology resources of 795 rectal and colon cancers in two U.S.-nationwide prospective cohort studies, we measured tumor-associated let-7a and let-7b expression levels by quantitative reverse-transcription PCR, and CD3+, CD8+, CD45RO (PTPRC)+, and FOXP3+ cell densities by tumor tissue microarray immunohistochemistry and computer-assisted image analysis. Logistic regression analysis and Cox proportional hazards regression were used to assess associations of let-7a (and let-7b) expression (quartile predictor variables) with T-cell densities (binary outcome variables) and mortality, respectively, controlling for tumor molecular features, including microsatellite instability, CpG island methylator phenotype, LINE-1 methylation, and KRAS, BRAF, and PIK3CA mutations. Compared with cases in the lowest quartile of let-7a expression, those in the highest quartile were associated with lower densities of CD3+ [multivariate odds ratio (OR), 0.40; 95% confidence interval (CI), 0.23–0.67; Ptrend = 0.003] and CD45RO+ cells (multivariate OR, 0.31; 95% CI, 0.17–0.58; Ptrend = 0.0004), and higher colorectal cancer-specific mortality (multivariate hazard ratio, 1.82; 95% CI, 1.42–3.13; Ptrend = 0.001). In contrast, let-7b expression was not significantly associated with T-cell density or colorectal cancer prognosis. Our data support the role of let-7a in suppressing antitumor immunity in colorectal cancer and suggest let-7a as a potential target of immunotherapy. Cancer Immunol Res; 4(11); 927–35. ©2016 AACR.

Accumulating evidence indicates that tumor molecular pathologic changes elicit immune reactions and that immune cells in the tumor microenvironment influence tumor evolution (1–3). Hence, tumor cells need to evade antitumor immune response. In colorectal cancer, the degree of infiltration of T lymphocytes, such as CD8+ cytotoxic T cells and CD45RO (PTPRC)+ effector memory T cells, has been associated with high-level microsatellite instability (MSI-high; refs. 4, 5) and favorable prognosis (5–7). Therapeutic antibodies against immune checkpoint molecules, including CTLA4, PDCD1 (programmed cell death 1; PD-1), and CD274 (PDCD1 ligand 1; PD-L1), can enhance antitumor T-cell activity and improve clinical outcome in various tumor types (8). Immune checkpoint blockade has been found to be effective in the MSI-high subtype of colorectal cancer (9). MSI-high colorectal cancers generate immunogenic neoantigens that can elicit lymphocytic immune response (10), resulting in upregulation of multiple immune checkpoint pathways (11). Beyond MSI status and neoantigen load, it is of interest to identify other factors that influence immune cell infiltration in colorectal cancer.

MicroRNAs are small noncoding RNAs that epigenetically regulate diverse biological and pathologic processes, including immune response and tumor progression (12). It has been shown that synthetic oligonucleotides can be delivered into cells or tissues to modulate the function of endogenous microRNAs, and small molecules may modify the expression or function of microRNAs (13). Through a novel delivery system that can target the tumor microenvironment, antisense oligomers have been shown to reduce tumor volumes of miR155-addicted lymphoma in mice, demonstrating the therapeutic potential of microRNA targeting (14). Our group has shown that tumor expression of microRNA MIR21 is inversely associated with densities of CD3+ and CD45RO+ T cells in colorectal cancer tissue (15). In addition, the let-7 microRNA family has been shown to play an important role in host immunity (16–18). Experimental evidence suggests that let-7 suppresses TLR4-mediated immune activation during infection, and that downregulation of let-7 expression restores immune response against pathogens (16, 17). In addition, decreased let-7 expression is associated with the self-renewal of memory T cells (18). Although tumor-associated let-7 expression has been associated with patient survival in lung (19), ovarian (20, 21), and other cancers, its prognostic association in colorectal cancer remains unclear. In human, let-7a and let-7b together account for the vast majority of let-7 RNA molecules present in colonic epithelia (22). Therefore, we hypothesized that let-7a or let-7b expression might be associated with a lower density of T cells in colorectal cancer tissue and worse clinical outcome.

To test our hypotheses, we examined let-7a and let-7b expression in colorectal cancer tissues from two U.S.-nationwide prospective cohort studies, the Nurses’ Health Study (NHS) and the Health Professionals Follow-up Study (HPFS), in relation to CD3+, CD8+, CD45RO+, and FOXP3+ T-cell densities in tumor tissue and colorectal cancer mortality.

Study population

We utilized the databases of two U.S.-nationwide prospective cohort studies, the NHS (121,701 women followed since 1976) and the HPFS (51,529 men followed since 1986; refs. 23, 24). In both cohorts, follow-up questionnaires were sent at baseline and biennially thereafter to collect and update lifestyle and health-related information, and to identify newly diagnosed diseases. Study physicians reviewed medical records and assigned causes of death. The National Death Index was used to ascertain deaths of study participants and identify unreported fatal colorectal cancer cases. Formalin-fixed paraffin-embedded (FFPE) tissue blocks were collected from hospitals where participants with colorectal cancer had undergone tumor resection. Hematoxylin and eosin–stained tissue sections of all colorectal cancer cases were reviewed by a pathologist (S. Ogino), who was unaware of other data. Tumor differentiation was categorized as well, as moderate versus poor (>50% vs. ≤50% glandular area). Based on the availability of follow-up data and adequate tissue specimens for analysis, 795 colorectal cancer cases (diagnosed up to 2008) were included. Written informed consent was obtained from all study participants. Tumor tissue was macrodissected from FFPE specimens in all cases. Tissue collection and analyses were approved by the human subjects committee at the Harvard T.H. Chan School of Public Health and the Brigham and Women's Hospital (Boston, MA).

Quantitative reverse-transcription PCR for let-7a and let-7b

RNA was extracted with the RecoverAll Total Nucleic Acid Isolation Kit (Ambion), and cDNA was synthesized with miScript II RT Kit (Qiagen). Quantitative PCR was performed using the miScript PCR System (Qiagen) with specific miScript Primer Assays for let-7a (catalog number, MS00031220), let-7b (catalog number, MS00003122), and RNU6-2 (catalog number MS00033740). Amplification was performed in duplicate with the StepOnePlus Real-Time PCR Systems (Applied Biosystems) under the following conditions: 15 minutes at 95°C and 40 cycles of 15 seconds at 94°C, 30 seconds at 55°C, and 30 seconds at 70°C. The Spearman rank-correlation coefficient between the duplicate cycle threshold (Ct) values was 0.99 for let-7a, 0.97 for let-7b, and 0.98 for RNU6-2.

In our validation study, the Ct values for let-7a, let-7b, and RNU6-2 increased linearly with serial 10-fold dilution of cDNA from the same tumor specimens (R2 ≥ 0.98; Supplementary Fig. S1). The interassay coefficient of variation of Ct values from the same tumor specimens in four different batches was 1.2% for let-7a, 1.4% for let-7b, and 1.0% for RNU6-2 (Supplementary Table S1). let-7a and let-7b expression was normalized with RNU6-2 using the 2−ΔCt method (where ΔCt = “the average Ct value of let-7a or let-7b” − “the average Ct value of RNU6-2”) (25).

Analysis of T-cell densities in tumors

Tissue microarray was constructed to assess the densities of CD3+, CD8+, CD45RO (PTPRC)+, and FOXP3+ T cells in tumors with immunohistochemistry techniques (5). We used an automated scanning microscope and the Ariol image analysis system (Genetix) to quantify T-cell density in tissue microarray cores as previously described (5). The median value was used to dichotomize the density of CD3+, CD8+, CD45RO+, or FOXP3+ T cells.

Analyses of BRAF, KRAS, and PIK3CA mutations and other molecular features

DNA was extracted from archival FFPE cancer tissue blocks. PCR and pyrosequencing for BRAF (codon 600; ref. 26), KRAS (codons 12, 13, 61 and 146; refs. 27, 28), and PIK3CA (exons 9 and 20; ref. 29) were performed as previously described. MSI status was assessed using 10 markers (D2S123, D5S346, D17S250, BAT25, BAT26, BAT40, D18S55, D18S56, D18S67, and D18S487) as previously (30). We defined MSI-high as the presence of instability in ≥30% of the markers, and MSI-low/microsatellite stable (MSS) as instability in <30% of the markers. Analyses of long interspersed nucleotide element-1 (LINE-1) methylation (31, 32) and eight CpG island methylator phenotype (CIMP)-specific loci (CACNA1G, CDKN2A, CRABP1, IGF2, MLH1, NEUROG1, RUNX3, and SOCS1; ref. 30) were performed.

Laser capture microdissection

To investigate let-7 expression in tumor cells separately from stromal cells, 10 cases were selected, and paired tumor cells and tumor stromal tissues were obtained, using the Arcturus XT LCM system (Applied Biosystems). Sections were cut to 8-μm thickness, and placed on Arcturus polyethylene naphthalate membrane frame slides (catalog number, LCM0521) and allowed to dry. Based on histopathologic review, tumor cells and adjacent stromal areas were separately microdissected and captured onto CapSure Macro LCM Caps (catalog number LCM0211).

Statistical analysis

Neither tumor-associated let-7 expression nor log-transformed values of let-7 expression fit a normal distribution in the Kolmogorov–Smirnov test for normality (P ≤ 0.01). Thus, we tested our primary hypotheses using a statistical trend test across the ordinal quartiles of let-7 expression as a continuous variable. For our first primary hypothesis that let-7 expression might be inversely associated with immune response, we used a logistic regression model to test the association of let-7a or let-7b expression (an ordinal quartile predictor variable) with T-cell densities (a binary outcome variable). The densities of CD3+, CD8+, CD45RO (PTPRC)+, and FOXP3+ T cells in tumors were dichotomized at the median value. Considering multiple comparisons (using four T-cell variables for each of let-7a and let-7b), the α level was adjusted to 0.006 (=0.05/8) by simple Bonferroni correction. To control for confounding, multivariate logistic regression model initially included age (continuous), sex, year of diagnosis (continuous), family history of colorectal cancer, tumor location (proximal colon vs. distal colon vs. rectum), MSI (high vs. MSI-low/MSS), CIMP (high vs. low/negative), KRAS (mutant vs. wild-type), BRAF (mutant vs. wild-type), PIK3CA (mutant vs. wild-type), and LINE-1 methylation level (continuous). A backward stepwise elimination with a threshold of P = 0.05 was used to select covariates in the final models. For cases missing any categorical covariate, such as family history (0.4%), tumor location (0.4%), MSI (3.9%), CIMP (8.6%), KRAS (2.6%), BRAF (2.9%), and PIK3CA (9.7%), we included those cases in a majority category of the covariate.

For the second primary hypothesis that let-7 expression in colorectal cancer tissue might be associated with higher mortality, we used a Cox proportional hazards regression model to assess the association of let-7a or let-7b expression (an ordinal quartile predictor variable) with colorectal cancer mortality. Deaths from causes other than colorectal cancer were censored for cancer-specific mortality. Considering multiple comparisons (cancer-specific mortality and overall mortality for each of let-7a and let-7b), the α level was adjusted to 0.012 (=0.05/4) by simple Bonferroni correction. To control for confounding, the multivariate Cox proportional hazards regression model initially included the same set of covariates as mentioned above, and we used the same backward stepwise elimination procedure to select covariates for the final model.

All other analyses for clinical, pathologic, and molecular associations were secondary exploratory analyses, and we adjusted the two-sided α level to 0.003 (=0.05/15) by simple Bonferroni correction for multiple comparisons. To assess associations between the ordinal categories (first to fourth quartile) of tumor-associated let-7a and let-7b expression and categorical data, the χ2 test was performed. To compare mean age and mean LINE-1 methylation levels, an analysis of variance assuming equal variances was performed. Because of a potential association between let-7 and disease stage, multivariate logistic regression analysis was performed to control for confounding. We used let-7a or let-7b expression as an ordinal quartile predictor variable and disease stage as a binary outcome variable (stages I–II vs. stages III–IV), with the same set of initial covariates and the same model construction procedure as mentioned above. We used the Wilcoxon signed rank test to compare the expression of let-7a or let-7b in colorectal cancer cells versus tumor stromal cells, and in tumor tissues versus adjacent mucosal tissues. We used the SAS program (Version 9.3, SAS Institute) for all statistical analyses. All P values were two-sided.

Expression levels of let-7a and let-7b in colorectal cancer tissue

We measured tumor-associated let-7a and let-7b expression by quantitative reverse-transcription PCR assay on 795 colorectal cancer cases within the NHS and the HPFS. Table 1 and Supplementary Table S2 show clinical, pathologic, and molecular characteristics of the 795 cases according to let-7a and let-7b expression levels, respectively. Levels of let-7a expression might be associated with disease stage, but statistical power was limited when we conducted a 4 × 4 χ2 test (P = 0.10). To assess a statistical trend of this association and control for confounding, we used logistic regression analysis and assessed the association of let-7a or let-7b expression as a quartile predictor variable with disease stage as a binary outcome variable (stage I–II vs. stage III–IV; Supplementary Table S3). Higher levels of let-7a or let-7b expression might be associated with stage III–IV disease (Ptrend = 0.005 or 0.0005, respectively, with an adjusted α level of 0.003).

Table 1.

Clinical, pathologic, and molecular characteristics according to let-7a expression in 795 colorectal cancer tissues

let-7a expression
CharacteristicaTotal (795)Q1 (198)Q2 (199)Q3 (199)Q4 (199)Pb
Age 
 Mean ± SD (year) 68.6 ± 8.7 69.3 ± 8.5 69.1 ± 8.9 67.7 ± 8.6 68.4 ± 8.7 0.26 
Sex      0.49 
 Men 348 (44%) 86 (43%) 94 (47%) 89 (45%) 79 (40%)  
 Women 447 (56%) 112 (57%) 105 (53%) 110 (55%) 120 (60%)  
BMI (kg/m2     0.30 
 <25 317 (40%) 78 (40%) 87 (44%) 83 (42%) 69 (35%)  
 25–30 315 (40%) 75 (38%) 81 (41%) 78 (39%) 81 (41%)  
 >30 161 (20%) 44 (22%) 31 (16%) 37 (19%) 49 (25%)  
Year of diagnosis      0.01 
 Prior to 1998 374 (47%) 85 (43%) 113 (57%) 91 (46%) 85 (43%)  
 1998 to 2008 421 (53%) 113 (57%) 86 (43%) 108 (54%) 114 (57%)  
Family history of colorectal cancer      0.82 
 Absent 631 (80%) 155 (79%) 156 (78%) 157 (80%) 163 (82%)  
 Present 161 (20%) 42 (21%) 43 (22%) 40 (20%) 36 (18%)  
Tumor location      0.38 
 Cecum 139 (17%) 35 (18%) 40 (20%) 33 (17%) 31 (16%)  
 Ascending to transverse colon 251 (32%) 62 (31%) 51 (26%) 72 (36%) 66 (33%)  
 Splenic flexure to sigmoid 223 (28%) 53 (27%) 58 (29%) 48 (24%) 64 (32%)  
 Rectosigmoid and rectum 179 (23%) 47 (24%) 50 (25%) 45 (23%) 37 (19%)  
Number of negative lymph nodes      0.42 
 Median (interquartile range) 7 (0–12) 7.5 (0–14) 7 (2–11) 6 (0–12) 7 (0–12)  
Disease stage      0.10 
 I 183 (24%) 52 (28%) 52 (28%) 41 (22%) 38 (20%)  
 II 242 (32%) 70 (37%) 58 (31%) 60 (32%) 54 (29%)  
 III 221 (30%) 48 (26%) 54 (29%) 61 (33%) 58 (31%)  
 IV 104 (14%) 18 (10%) 24 (13%) 25 (13%) 37 (20%)  
Tumor differentiation      0.65 
 Well to moderate 721 (91%) 183 (93%) 179 (90%) 181 (91%) 178 (89%)  
 Poor 73 (9%) 14 (7%) 20 (10%) 18 (9%) 21 (11%)  
MSI status      0.37 
 MSI-low/MSS 648 (85%) 162 (86%) 164 (84%) 155 (82%) 167 (88%)  
 MSI-high 116 (15%) 27 (14%) 31 (16%) 35 (18%) 23 (12%)  
CIMP status      0.34 
 Low/negative 595 (82%) 144 (84%) 154 (81%) 148 (80%) 149 (82%)  
 High 132 (18%) 28 (16%) 35 (19%) 36 (20%) 33 (18%)  
BRAF mutation      0.04 
 (−) 650 (84%) 166 (88%) 173 (88%) 155 (80%) 156 (81%)  
 (+) 122 (16%) 23 (12%) 23 (12%) 38 (20%) 38 (19%)  
KRAS mutation      0.56 
 (−) 459 (60%) 114 (60%) 109 (56%) 121 (63%) 115 (60%)  
 (+) 308 (40%) 76 (40%) 85 (44%) 70 (37%) 77 (40%)  
PIK3CA mutation      0.52 
 (−) 595 (83%) 155 (86%) 143 (80%) 152 (83%) 145 (82%)  
 (+) 123 (17%) 25 (14%) 35 (20%) 31 (17%) 32 (18%)  
LINE-1 methylation level      0.63 
 Mean ± SD (%) 62.2 ± 9.3 62.0 ± 8.0 61.7 ± 9.6 62.1 ± 9.5 62.9 ± 10.0  
let-7a expression
CharacteristicaTotal (795)Q1 (198)Q2 (199)Q3 (199)Q4 (199)Pb
Age 
 Mean ± SD (year) 68.6 ± 8.7 69.3 ± 8.5 69.1 ± 8.9 67.7 ± 8.6 68.4 ± 8.7 0.26 
Sex      0.49 
 Men 348 (44%) 86 (43%) 94 (47%) 89 (45%) 79 (40%)  
 Women 447 (56%) 112 (57%) 105 (53%) 110 (55%) 120 (60%)  
BMI (kg/m2     0.30 
 <25 317 (40%) 78 (40%) 87 (44%) 83 (42%) 69 (35%)  
 25–30 315 (40%) 75 (38%) 81 (41%) 78 (39%) 81 (41%)  
 >30 161 (20%) 44 (22%) 31 (16%) 37 (19%) 49 (25%)  
Year of diagnosis      0.01 
 Prior to 1998 374 (47%) 85 (43%) 113 (57%) 91 (46%) 85 (43%)  
 1998 to 2008 421 (53%) 113 (57%) 86 (43%) 108 (54%) 114 (57%)  
Family history of colorectal cancer      0.82 
 Absent 631 (80%) 155 (79%) 156 (78%) 157 (80%) 163 (82%)  
 Present 161 (20%) 42 (21%) 43 (22%) 40 (20%) 36 (18%)  
Tumor location      0.38 
 Cecum 139 (17%) 35 (18%) 40 (20%) 33 (17%) 31 (16%)  
 Ascending to transverse colon 251 (32%) 62 (31%) 51 (26%) 72 (36%) 66 (33%)  
 Splenic flexure to sigmoid 223 (28%) 53 (27%) 58 (29%) 48 (24%) 64 (32%)  
 Rectosigmoid and rectum 179 (23%) 47 (24%) 50 (25%) 45 (23%) 37 (19%)  
Number of negative lymph nodes      0.42 
 Median (interquartile range) 7 (0–12) 7.5 (0–14) 7 (2–11) 6 (0–12) 7 (0–12)  
Disease stage      0.10 
 I 183 (24%) 52 (28%) 52 (28%) 41 (22%) 38 (20%)  
 II 242 (32%) 70 (37%) 58 (31%) 60 (32%) 54 (29%)  
 III 221 (30%) 48 (26%) 54 (29%) 61 (33%) 58 (31%)  
 IV 104 (14%) 18 (10%) 24 (13%) 25 (13%) 37 (20%)  
Tumor differentiation      0.65 
 Well to moderate 721 (91%) 183 (93%) 179 (90%) 181 (91%) 178 (89%)  
 Poor 73 (9%) 14 (7%) 20 (10%) 18 (9%) 21 (11%)  
MSI status      0.37 
 MSI-low/MSS 648 (85%) 162 (86%) 164 (84%) 155 (82%) 167 (88%)  
 MSI-high 116 (15%) 27 (14%) 31 (16%) 35 (18%) 23 (12%)  
CIMP status      0.34 
 Low/negative 595 (82%) 144 (84%) 154 (81%) 148 (80%) 149 (82%)  
 High 132 (18%) 28 (16%) 35 (19%) 36 (20%) 33 (18%)  
BRAF mutation      0.04 
 (−) 650 (84%) 166 (88%) 173 (88%) 155 (80%) 156 (81%)  
 (+) 122 (16%) 23 (12%) 23 (12%) 38 (20%) 38 (19%)  
KRAS mutation      0.56 
 (−) 459 (60%) 114 (60%) 109 (56%) 121 (63%) 115 (60%)  
 (+) 308 (40%) 76 (40%) 85 (44%) 70 (37%) 77 (40%)  
PIK3CA mutation      0.52 
 (−) 595 (83%) 155 (86%) 143 (80%) 152 (83%) 145 (82%)  
 (+) 123 (17%) 25 (14%) 35 (20%) 31 (17%) 32 (18%)  
LINE-1 methylation level      0.63 
 Mean ± SD (%) 62.2 ± 9.3 62.0 ± 8.0 61.7 ± 9.6 62.1 ± 9.5 62.9 ± 10.0  

Abbreviations: BMI, body mass index; Q1 to Q4, quartile 1 (lowest) to quartile 4 (highest); SD, standard deviation.

aPercentage indicates the proportion of cases with a specific clinical, pathologic, or tumor molecular feature in patients with each expression level of let-7a. There were cases that had missing values for any of the characteristics except for age, sex, and year of diagnosis.

bTo assess associations between the ordinal categories (first to fourth quartile) of let-7a expression and categorical data, the χ2 test was performed. To compare mean age and mean LINE-1 methylation levels, the analysis of variance was performed. We adjusted the two-sided α level to 0.003 (=0.05/15) by simple Bonferroni correction.

let-7a and let-7b expression and T-cell density in colorectal cancer tissue

We measured the densities of T cells in colorectal cancer tissue by immunohistochemistry and image analysis. Pairwise correlations between the densities of CD3+, CD8+, CD45RO+, and FOXP3+ T cells were significant except between FOXP3+ and CD3+ cells and between FOXP3+ and CD8+ T cells (Supplementary Table S4).

Table 2 shows the distribution of colorectal cancer cases according to the densities of T cells (quartiles) and let-7a or let-7b expression (quartiles) in colorectal cancer tissue. In our primary hypothesis testing, we conducted univariate and multivariate logistic regression analyses to assess the associations of let-7a or let-7b expression as an ordinal quartile predictor variable with the density of CD3+, CD8+, CD45RO+, or FOXP3+ T cells in colorectal cancer tissue as a binary outcome variable (Table 3). let-7a expression was inversely associated with the densities of CD3+ and CD45RO+ T cells in multivariate analyses (multivariate Ptrend = 0.003 and 0.0004, respectively, with an adjusted α level at 0.006). Compared with cases in the lowest quartile of let-7a expression, those in the highest quartile were associated with lower density of CD3+ T cells [multivariate OR, 0.40; 95% confidence interval (CI), 0.23–0.67] and CD45RO+ T cells (multivariate OR, 0.31; 95% CI, 0.17–0.58). In contrast, let-7b expression was not significantly associated with any of the T-cell densities (all multivariate Ptrend ≥ 0.04 with an adjusted α level at 0.006).

Table 2.

Distribution of cases according to let-7a and let-7b expression and T-cell densities in colorectal cancer tissue

let-7a expressionlet-7b expression
T-cell densityTotalQ1Q2Q3Q4PtrendaQ1Q2Q3Q4Ptrenda
CD3+ cell density 0.004     0.18 
 Q1 116 (23%) 19 (16%) 33 (24%) 27 (21%) 37 (28%)  33 (26%) 21 (16%) 31 (23%) 30 (25%)  
 Q2 121 (23%) 24 (20%) 32 (24%) 26 (20%) 39 (30%)  37 (30%) 33 (25%) 23 (17%) 28 (23%)  
 Q3 144 (28%) 41 (34%) 33 (24%) 36 (28%) 34 (26%)  29 (23%) 43 (33%) 38 (29%) 34 (28%)  
 Q4 133 (26%) 35 (29%) 37 (27%) 40 (31%) 21 (16%)  26 (21%) 34 (26%) 42 (31%) 30 (24%)  
CD8+ cell density 0.010     0.008 
 Q1 130 (26%) 30 (25%) 27 (21%) 32 (25%) 41 (32%)  40 (32%) 35 (27%) 32 (25%) 23 (19%)  
 Q2 129 (25%) 26 (22%) 31 (24%) 34 (27%) 38 (29%)  31 (25%) 38 (30%) 30 (23%) 29 (24%)  
 Q3 127 (25%) 34 (28%) 41 (32%) 27 (21%) 25 (19%)  26 (21%) 29 (23%) 34 (26%) 37 (30%)  
 Q4 120 (24%) 30 (25%) 31 (24%) 34 (27%) 25 (19%)  27 (22%) 26 (20%) 34 (26%) 33 (27%)  
CD45RO+ cell density 0.001     0.05 
 Q1 133 (26%) 20 (17%) 34 (25%) 29 (22%) 50 (38%)  28 (22%) 31 (23%) 39 (29%) 34 (28%)  
 Q2 131 (25%) 22 (19%) 38 (28%) 33 (25%) 38 (29%)  32 (26%) 36 (27%) 27 (20%) 36 (30%)  
 Q3 126 (24%) 33 (28%) 33 (24%) 37 (28%) 23 (17%)  34 (27%) 27 (20%) 31 (23%) 34 (28%)  
 Q4 128 (25%) 43 (36%) 30 (22%) 34 (26%) 21 (16%)  32 (25%) 39 (30%) 39 (28%) 17 (14%)  
FOXP3+ cell density 0.88     0.40 
 Q1 126 (26%) 29 (26%) 32 (26%) 30 (24%) 35 (27%)  31 (26%) 32 (26%) 30 (24%) 32 (27%)  
 Q2 130 (26%) 30 (27%) 33 (26%) 36 (29%) 31 (24%)  32 (27%) 30 (24%) 29 (23%) 38 (32%)  
 Q3 121 (25%) 29 (26%) 28 (22%) 31 (25%) 33 (25%)  30 (25%) 29 (23%) 32 (25%) 30 (26%)  
 Q4 114 (23%) 25 (22%) 32 (26%) 26 (21%) 31 (24%)  27 (22%) 34 (27%) 35 (28%) 18 (15%)  
let-7a expressionlet-7b expression
T-cell densityTotalQ1Q2Q3Q4PtrendaQ1Q2Q3Q4Ptrenda
CD3+ cell density 0.004     0.18 
 Q1 116 (23%) 19 (16%) 33 (24%) 27 (21%) 37 (28%)  33 (26%) 21 (16%) 31 (23%) 30 (25%)  
 Q2 121 (23%) 24 (20%) 32 (24%) 26 (20%) 39 (30%)  37 (30%) 33 (25%) 23 (17%) 28 (23%)  
 Q3 144 (28%) 41 (34%) 33 (24%) 36 (28%) 34 (26%)  29 (23%) 43 (33%) 38 (29%) 34 (28%)  
 Q4 133 (26%) 35 (29%) 37 (27%) 40 (31%) 21 (16%)  26 (21%) 34 (26%) 42 (31%) 30 (24%)  
CD8+ cell density 0.010     0.008 
 Q1 130 (26%) 30 (25%) 27 (21%) 32 (25%) 41 (32%)  40 (32%) 35 (27%) 32 (25%) 23 (19%)  
 Q2 129 (25%) 26 (22%) 31 (24%) 34 (27%) 38 (29%)  31 (25%) 38 (30%) 30 (23%) 29 (24%)  
 Q3 127 (25%) 34 (28%) 41 (32%) 27 (21%) 25 (19%)  26 (21%) 29 (23%) 34 (26%) 37 (30%)  
 Q4 120 (24%) 30 (25%) 31 (24%) 34 (27%) 25 (19%)  27 (22%) 26 (20%) 34 (26%) 33 (27%)  
CD45RO+ cell density 0.001     0.05 
 Q1 133 (26%) 20 (17%) 34 (25%) 29 (22%) 50 (38%)  28 (22%) 31 (23%) 39 (29%) 34 (28%)  
 Q2 131 (25%) 22 (19%) 38 (28%) 33 (25%) 38 (29%)  32 (26%) 36 (27%) 27 (20%) 36 (30%)  
 Q3 126 (24%) 33 (28%) 33 (24%) 37 (28%) 23 (17%)  34 (27%) 27 (20%) 31 (23%) 34 (28%)  
 Q4 128 (25%) 43 (36%) 30 (22%) 34 (26%) 21 (16%)  32 (25%) 39 (30%) 39 (28%) 17 (14%)  
FOXP3+ cell density 0.88     0.40 
 Q1 126 (26%) 29 (26%) 32 (26%) 30 (24%) 35 (27%)  31 (26%) 32 (26%) 30 (24%) 32 (27%)  
 Q2 130 (26%) 30 (27%) 33 (26%) 36 (29%) 31 (24%)  32 (27%) 30 (24%) 29 (23%) 38 (32%)  
 Q3 121 (25%) 29 (26%) 28 (22%) 31 (25%) 33 (25%)  30 (25%) 29 (23%) 32 (25%) 30 (26%)  
 Q4 114 (23%) 25 (22%) 32 (26%) 26 (21%) 31 (24%)  27 (22%) 34 (27%) 35 (28%) 18 (15%)  

Abbreviation: Q1 to Q4, quartile 1 (lowest) to quartile 4 (highest).

aPtrend value was calculated by the linear trend test across the ordinal (first to fourth quartile) categories of let-7a or let-7b expression as a continuous variable in the univariate ordinal logistic regression model for the density of CD3+, CD8+, CD45RO+, or FOXP3+ cells (an ordinal quartile outcome variable). We adjusted the two-sided α level to 0.006 (=0.05/8) by simple Bonferroni correction.

Table 3.

let-7a and let-7b expression and T-cell densities in colorectal cancer tissue

let-7a expressionUnivariate OR (95% CI)Multivariate OR (95% CI)let-7b expressionUnivariate OR (95% CI)Multivariate OR (95% CI)
Model of CD3+ cell density as a binary outcome (n = 514)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 0.61 (0.37–1.01) 0.62 (0.37–1.03) Q2 1.82 (1.11–2.98) 1.75 (1.06–2.88) 
 Q3 0.81 (0.49–1.36) 0.82 (0.49–1.39) Q3 1.89 (1.15–3.09) 1.67 (1.01–2.78) 
 Q4 0.41 (0.25–0.68) 0.40 (0.23–0.67) Q4 1.40 (0.85–2.32) 1.30 (0.78–2.18) 
Ptrendb 0.004 0.003 Ptrendb 0.18 0.40 
Model of CD8+ cell density as a binary outcome (n = 506)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 1.08 (0.66–1.79) 1.05 (0.63–1.76) Q2 1.01 (0.61–1.66) 1.03 (0.61–1.72) 
 Q3 0.81 (0.49–1.33) 0.77 (0.46–1.29) Q3 1.47 (0.90–2.41) 1.47 (0.88–2.47) 
 Q4 0.55 (0.34–0.92) 0.54 (0.32–0.91) Q4 1.80 (1.09–2.99) 1.59 (0.94–2.69) 
Ptrendb 0.010 0.008 Ptrendb 0.008 0.04 
Model of CD45RO+ cell density as a binary outcome (n = 518)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 0.50 (0.29–0.87) 0.50 (0.28–0.88) Q2 1.22 (0.71–2.11) 1.34 (0.76–2.36) 
 Q3 0.60 (0.35–1.03) 0.52 (0.30–0.92) Q3 1.18 (0.68–2.04) 1.32 (0.75–2.31) 
 Q4 0.33 (0.18–0.60) 0.31 (0.17–0.58) Q4 0.48 (0.25–0.92) 0.52 (0.27–1.00) 
Ptrendb 0.001 0.0004 Ptrendb 0.05 0.09 
Model of FOXP3+ cell density as a binary outcome (n = 491)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 1.01 (0.61–1.68) 1.07 (0.64–1.81) Q2 1.12 (0.68–1.85) 1.09 (0.65–1.83) 
 Q3 0.94 (0.57–1.57) 0.89 (0.53–1.51) Q3 1.26 (0.76–2.07) 1.17 (0.70–1.96) 
 Q4 1.06 (0.64–1.76) 1.00 (0.60–1.68) Q4 0.76 (0.45–1.27) 0.77 (0.46–1.32) 
Ptrendb 0.88 0.84 Ptrendb 0.40 0.43 
let-7a expressionUnivariate OR (95% CI)Multivariate OR (95% CI)let-7b expressionUnivariate OR (95% CI)Multivariate OR (95% CI)
Model of CD3+ cell density as a binary outcome (n = 514)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 0.61 (0.37–1.01) 0.62 (0.37–1.03) Q2 1.82 (1.11–2.98) 1.75 (1.06–2.88) 
 Q3 0.81 (0.49–1.36) 0.82 (0.49–1.39) Q3 1.89 (1.15–3.09) 1.67 (1.01–2.78) 
 Q4 0.41 (0.25–0.68) 0.40 (0.23–0.67) Q4 1.40 (0.85–2.32) 1.30 (0.78–2.18) 
Ptrendb 0.004 0.003 Ptrendb 0.18 0.40 
Model of CD8+ cell density as a binary outcome (n = 506)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 1.08 (0.66–1.79) 1.05 (0.63–1.76) Q2 1.01 (0.61–1.66) 1.03 (0.61–1.72) 
 Q3 0.81 (0.49–1.33) 0.77 (0.46–1.29) Q3 1.47 (0.90–2.41) 1.47 (0.88–2.47) 
 Q4 0.55 (0.34–0.92) 0.54 (0.32–0.91) Q4 1.80 (1.09–2.99) 1.59 (0.94–2.69) 
Ptrendb 0.010 0.008 Ptrendb 0.008 0.04 
Model of CD45RO+ cell density as a binary outcome (n = 518)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 0.50 (0.29–0.87) 0.50 (0.28–0.88) Q2 1.22 (0.71–2.11) 1.34 (0.76–2.36) 
 Q3 0.60 (0.35–1.03) 0.52 (0.30–0.92) Q3 1.18 (0.68–2.04) 1.32 (0.75–2.31) 
 Q4 0.33 (0.18–0.60) 0.31 (0.17–0.58) Q4 0.48 (0.25–0.92) 0.52 (0.27–1.00) 
Ptrendb 0.001 0.0004 Ptrendb 0.05 0.09 
Model of FOXP3+ cell density as a binary outcome (n = 491)a 
 Q1 1 (reference) 1 (reference) Q1 1 (reference) 1 (reference) 
 Q2 1.01 (0.61–1.68) 1.07 (0.64–1.81) Q2 1.12 (0.68–1.85) 1.09 (0.65–1.83) 
 Q3 0.94 (0.57–1.57) 0.89 (0.53–1.51) Q3 1.26 (0.76–2.07) 1.17 (0.70–1.96) 
 Q4 1.06 (0.64–1.76) 1.00 (0.60–1.68) Q4 0.76 (0.45–1.27) 0.77 (0.46–1.32) 
Ptrendb 0.88 0.84 Ptrendb 0.40 0.43 

Abbreviation: Q1 to Q4, quartile 1 (lowest) to quartile 4 (highest).

aThe multivariate logistic regression model initially included age, sex, year of diagnosis, family history of colorectal cancer in parent or sibling, tumor location, MSI, CpG island methylator phenotype, KRAS, BRAF, and PIK3CA mutations, and LINE-1 methylation level. A backward stepwise elimination with a threshold of P < 0.05 was used to select variables in the final models.

bPtrend was calculated by the linear trend test across the ordinal categories (first to fourth quartile) of let-7a or let-7b expression as a continuous variable in the logistic regression model for the density of CD3+, CD8+, CD45RO+, or FOXP3+ cells as a binary outcome variable. We adjusted the two-sided α level to 0.006 (=0.05/8) by simple Bonferroni correction.

Tumor-associated let-7a and let-7b expression and colorectal cancer mortality

During a median follow-up of 13.5 years for censored cases, 445 deaths, including 242 colorectal cancer-specific deaths, were documented. In the second primary hypothesis test (Table 4), let-7a expression was positively associated with cancer-specific mortality (multivariate Ptrend = 0.001). The multivariate hazard ratio of the highest versus lowest quartile of let-7a for cancer-specific mortality was 1.82 (95% CI, 1.42–3.13). We observed similar findings for the 518 cases with density of CD45RO+ cells available (Supplementary Table S5). In contrast, let-7b expression was not associated with cancer-specific mortality (multivariate Ptrend = 0.44). In Kaplan–Meier analysis (Fig. 1), higher let-7a expression was associated with shorter colorectal cancer-specific survival (P = 0.003).

Table 4.

let-7a and let-7b expression in colorectal cancer tissue and survival

Cancer-specific mortalityOverall mortality
let-7 expressionCases, nEvents, nUnivariate HR (95% CI)Multivariate HRa (95% CI)Events, nUnivariate HR (95% CI)Multivariate HRa (95% CI)
let-7a (n = 795) 
 Q1 198 43 1 (reference) 1 (reference) 96 1 (reference) 1 (reference) 
 Q2 199 57 1.38 (0.93–2.05) 1.38 (0.93–2.05) 120 1.28 (0.98–1.68) 1.34 (1.02–1.75) 
 Q3 199 66 1.63 (1.13–2.45) 1.66 (1.48–3.25) 115 1.31 (1.00–1.71) 1.41 (1.08–1.86) 
 Q4 199 76 1.97 (1.25–2.66) 1.82 (1.42–3.13) 114 1.35 (1.03–1.77) 1.39 (1.06–1.84) 
Ptrendb   0.0002 0.001  0.04 0.02 
let-7b (n = 792) 
 Q1 198 52 1 (reference) 1 (reference) 109 1 (reference) 1 (reference) 
 Q2 198 63 1.26 (0.87–1.82) 1.30 (0.90–1.88) 114 1.31 (0.87–1.47) 1.17 (0.90–1.52) 
 Q3 198 70 1.45 (1.01–2.08) 1.53 (1.07–2.20) 114 1.08 (0.96–1.63) 1.26 (0.97–1.64) 
 Q4 198 56 1.08 (0.74–1.58) 1.11 (0.76–1.62) 105 1.01 (0.77–1.32) 1.00 (0.77–1.32) 
Ptrendb   0.53 0.44  0.76 0.84 
Cancer-specific mortalityOverall mortality
let-7 expressionCases, nEvents, nUnivariate HR (95% CI)Multivariate HRa (95% CI)Events, nUnivariate HR (95% CI)Multivariate HRa (95% CI)
let-7a (n = 795) 
 Q1 198 43 1 (reference) 1 (reference) 96 1 (reference) 1 (reference) 
 Q2 199 57 1.38 (0.93–2.05) 1.38 (0.93–2.05) 120 1.28 (0.98–1.68) 1.34 (1.02–1.75) 
 Q3 199 66 1.63 (1.13–2.45) 1.66 (1.48–3.25) 115 1.31 (1.00–1.71) 1.41 (1.08–1.86) 
 Q4 199 76 1.97 (1.25–2.66) 1.82 (1.42–3.13) 114 1.35 (1.03–1.77) 1.39 (1.06–1.84) 
Ptrendb   0.0002 0.001  0.04 0.02 
let-7b (n = 792) 
 Q1 198 52 1 (reference) 1 (reference) 109 1 (reference) 1 (reference) 
 Q2 198 63 1.26 (0.87–1.82) 1.30 (0.90–1.88) 114 1.31 (0.87–1.47) 1.17 (0.90–1.52) 
 Q3 198 70 1.45 (1.01–2.08) 1.53 (1.07–2.20) 114 1.08 (0.96–1.63) 1.26 (0.97–1.64) 
 Q4 198 56 1.08 (0.74–1.58) 1.11 (0.76–1.62) 105 1.01 (0.77–1.32) 1.00 (0.77–1.32) 
Ptrendb   0.53 0.44  0.76 0.84 

Abbreviations: HR, hazard ratio; Q1 to Q4, quartile 1 (lowest) to quartile 4 (highest).

aThe multivariate Cox regression model initially included sex, age, year of diagnosis, family history of colorectal cancer in parent or sibling, MSI, CpG island methylator phenotype, KRAS, BRAF, and PIK3CA mutations, and LINE-1 methylation level. A backward stepwise elimination with a threshold of P < 0.05 was used to select variables in the final models.

bPtrend was calculated by the linear trend test across the ordinal categories (first to fourth quartile) of let-7a or let-7b expression as a continuous variable in the Cox regression model for cancer-specific mortality and overall mortality. We adjusted the two-sided α level to 0.012 (=0.05/4) by simple Bonferroni correction.

Figure 1.

Kaplan–Meier curve for cancer-specific survival and overall survival classified by quartiles of let-7a (A, B) or let-7b expression (C, D) in colorectal cancer tissue. Expression level increases from quartile 1 to quartile 4. Statistical analyses were performed using the log rank test.

Figure 1.

Kaplan–Meier curve for cancer-specific survival and overall survival classified by quartiles of let-7a (A, B) or let-7b expression (C, D) in colorectal cancer tissue. Expression level increases from quartile 1 to quartile 4. Statistical analyses were performed using the log rank test.

Close modal

let-7a and let-7b in cancer, stromal, and adjacent nontumor cells of colonic mucosa

To determine whether let-7 microRNAs might be expressed in colorectal cancer cells and/or cells in tumor stroma, we microdissected tumor cells and tumor stromal tissues in 10 selected cases, using a laser capture microdissection (LCM) technique. Expression of let-7a and let-7b in tumor stroma was on average approximately 4 times higher than that in paired tumor cells (Supplementary Fig. S2A and S2B; Wilcoxon signed rank test, P = 0.002 and 0.004 for let-7a and let-7b, respectively).

We macrodissected tumor tissue and adjacent nontumor colonic mucosa tissue in 52 colorectal cancer cases. Expression of let-7a and let-7b was generally lower in tumor tissue than in paired adjacent nontumor colonic mucosa (Supplementary Fig. S2C and S2D, Wilcoxon signed rank test, P < 0.0001 and 0.0003 for let-7a and let-7b, respectively).

We conducted this study to test the hypothesis that let-7a or let-7b expression might be inversely associated with T-cell densities in colorectal cancer tissue. We found the inverse association between let-7a expression levels and the densities of CD3+ cells and CD45RO+ T cells in colorectal cancer tissue. We also found that tumor-associated let-7a expression was positively associated with colorectal cancer-specific mortality.

The let-7 miRNA family has been reported to regulate immune activation in various cell types. In human epithelial cells, let-7 suppresses immune responses to pathogens by inhibition of Toll-like receptor 4 (TLR4), and downregulation of let-7 restores immune activation (16, 33). In mammalian macrophages exposed to pathogen challenge, let-7 controls the immune response via inhibition of the NFKB1 pathway or repression of cytokine expression (17, 34, 35). Furthermore, the let-7 family has been shown to regulate T-cell functions. In murine CD4+ T cells receiving a T-cell receptor signal, let-7 represses T-cell activation and facilitates T-cell anergy by targeting Mtor mRNA (36). In addition, let-7a has been reported to inhibit Th17 cell differentiation and increase regulatory T cells in a mouse hepatitis model (37), and suppression of let-7 expression has been associated with the differentiation of central memory T cells (18). Our population-based discovery of the inverse association between expression of let-7a and densities of CD3+ and CD45RO+ cells in colorectal cancer tissues suggests that let-7a might inhibit the lymphocytic immune response against colorectal cancer, which is consistent with experimental evidence for immune suppressive effect of the let-7 family.

Tumor molecular analyses are increasingly important in colorectal cancer research (38, 39). In addition, ample evidence indicates the relevance of immune characterization together with tumor molecular analyses (40–43). In particular, upregulation of the immune checkpoint ligand CD274 (PD-L1) in colorectal cancer has been associated with lower densities of FOXP3+ cells and tumor microsatellite stability in 823 tumors (ref. 44; in contrast to other much smaller studies, refs. 11, 45). As evidence indicates influences of tumor somatic alterations on immune checkpoint pathways (46), tumor molecular alterations may be targeted to augment effects of immunotherapies. Our findings suggest that let-7a may be a potential therapeutic target to improve the efficacy of immunotherapy.

Previous studies have found a controversial relationship between let-7 expression and cancer survival. Expression of let-7a and let-7f has been associated with longer survival in lung cancer (19), and let-7b expression with longer survival in serous ovarian cancer (20). However, MIRLET7A3 hypomethylation, which increases let-7a-3 expression, has been associated with shorter survival in epithelial ovarian cancers (21). To date, two studies using microarrays have investigated the prognostic value of let-7 in colorectal cancer, but no significant prognostic association has been observed (47, 48). Potential reasons for the inconsistent effects of let-7 on cancer survival may include limited sample sizes of those studies, and differences in study populations, cancer types, and measurement methods for let-7 family members. In the current study, we measured let-7a and let-7b that account for the majority of let-7 expressed in human colonic epithelia (22) in 795 patients. As a result, we found a strong positive association of let-7a expression with colorectal cancer–specific mortality.

One limitation of our study is the cross-sectional design, which cannot exclude the possibility of reverse causation. It is possible that CD45RO+ cells might suppress let-7a expression in the tumor microenvironment. Nevertheless, our hypothesis was based on various lines of experimental evidence indicating that let-7 suppresses immune responses (16–18, 33–36). Another limitation is the measurement of let-7 expression in colorectal cancer tissues, which contain a mixture of tumor cells and stromal cells, including immune cells. Our measurement in 10 pairs of tumor cells and tumor stroma suggests higher let-7a expression in stromal cells, although further studies are required to validate this finding and to identify the cell types responsible for let-7 production.

Our study has several strengths. First, this study utilizes our molecular pathologic epidemiology (49, 50) database of 795 colorectal cancer cases in two U.S.-nationwide prospective cohorts, which integrates epidemiological exposures, clinicopathologic features, tumor molecular markers, and immune reaction status. This enabled us to rigorously test the association of let-7 expression with T-cell density and mortality, controlling for potential confounders. Second, we used robust methods, including validated PCR assays, for microRNA expression and objective quantification of T-cell densities by image analysis. Third, our tumor specimens were obtained from many hospitals throughout the United States, which improves the generalizability of our findings.

In conclusion, higher let-7a expression was associated with lower densities of CD3+ and CD45RO+ cells in colorectal cancer tissue and with higher colorectal cancer–specific mortality. Our findings support a possible role for let-7a in downregulating T cell–mediated immunity in colorectal cancer. Further prospective studies are needed to validate these findings from the current hypothesis-generating study. Upon validation, these population-based data may have implications for expanding the benefit of immune therapies through targeting microRNAs.

No potential conflicts of interest were disclosed.

Conception and design: R. Dou, E.L. Giovannucci, C.S. Fuchs, Z.R. Qian, S. Ogino

Development of methodology: K. Mima, C.S. Fuchs, Z.R. Qian, S. Ogino

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R. Dou, R. Nishihara, Y. Cao, T. Hamada, K. Mima, A. Masuda, Y. Masugi, Y. Shi, M. Gu, K. Nosho, E.L. Giovannucci, A.T. Chan, C.S. Fuchs, S. Ogino

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): R. Dou, R. Nishihara, Y. Cao, T. Hamada, K. Mima, A. Masuda, M. Gu, X. Zhang, J.A. Meyerhardt, A.T. Chan, C.S. Fuchs, Z.R. Qian, S. Ogino

Writing, review, and/or revision of the manuscript: R. Dou, Y. Cao, T. Hamada, K. Mima, A. Masuda, A. da Silva, X. Zhang, J.A. Meyerhardt, E.L. Giovannucci, A.T. Chan, C.S. Fuchs, Z.R. Qian, S. Ogino

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Shi, W. Li, S. Ogino

Study supervision: A.T. Chan, C.S. Fuchs, Z.R. Qian, S. Ogino

We would like to thank the participants and staff of the Nurses' Health Study and the Health Professionals Follow-up Study for their valuable contributions as well as the following state cancer registries for their help: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, IN, IA, KY, LA, ME, MD, MA, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, SC, TN, TX, VA, WA, and WY.

This work was supported byNIH grants (P01 CA87969 and UM1 CA186107 to M.J. Stampfer; P01 CA55075 and UM1 CA167552 to W.C. Willett; P50 CA127003 to C.S. Fuchs; K24 DK098311 and R01 CA137178 to A.T. Chan; R01 CA151993 and R35 CA197735 to S. Ogino; and K07 CA190673 to R. Nishihara), by grants from the Project P Fund, The Friends of the Dana-Farber Cancer Institute, Bennett Family Fund, and the Entertainment Industry Foundation through National Colorectal Cancer Research Alliance. R. Dou is supported by the Program of Introducing Talents of Discipline to Universities B12003 and the International S&T Cooperation Program 2011DFA32570. T. Hamada was supported by a fellowship grant from the Uehara Memorial Foundation and by a grant from the Mochida Memorial Foundation for Medical and Pharmaceutical Research. Y. Masugi is supported by a fellowship grant of the Keio Gijuku Fukuzawa Memorial Fund for the Advancement of Education and Research.

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.
Ogino
S
,
Galon
J
,
Fuchs
CS
,
Dranoff
G
. 
Cancer immunology–analysis of host and tumor factors for personalized medicine
.
Nat Rev Clin Oncol
2011
;
8
:
711
9
.
2.
Mlecnik
B
,
Bindea
G
,
Angell
HK
,
Maby
P
,
Angelova
M
,
Tougeron
D
, et al
Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability
.
Immunity
2016
;
44
:
698
711
.
3.
Di Caro
G
,
Marchesi
F
,
Laghi
L
,
Grizzi
F
. 
Immune cells: plastic players along colorectal cancer progression
.
J Cell Mol Med
2013
;
17
:
1088
95
.
4.
Ogino
S
,
Nosho
K
,
Irahara
N
,
Meyerhardt
JA
,
Baba
Y
,
Shima
K
, et al
Lymphocytic reaction to colorectal cancer is associated with longer survival, independent of lymph node count, microsatellite instability, and CpG island methylator phenotype
.
Clin Cancer Res
2009
;
15
:
6412
20
.
5.
Nosho
K
,
Baba
Y
,
Tanaka
N
,
Shima
K
,
Hayashi
M
,
Meyerhardt
JA
, et al
Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review
.
J Pathol
2010
;
222
:
350
66
.
6.
Pages
F
,
Berger
A
,
Camus
M
,
Sanchez-Cabo
F
,
Costes
A
,
Molidor
R
, et al
Effector memory T cells, early metastasis, and survival in colorectal cancer
.
N Engl J Med
2005
;
353
:
2654
66
.
7.
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Berger
A
,
Bindea
G
,
Meatchi
T
, et al
Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction
.
J Clin Oncol
2011
;
29
:
610
8
.
8.
Smyth
MJ
,
Ngiow
SF
,
Ribas
A
,
Teng
MW
. 
Combination cancer immunotherapies tailored to the tumour microenvironment
.
Nat Rev Clin Oncol
2016
;
13
:
143
58
.
9.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
10.
Giannakis
M
,
Mu
XJ
,
Shukla
SA
,
Qian
ZR
,
Cohen
O
,
Nishihara
R
, et al
Genomic correlates of immune-cell infiltrates in colorectal carcinoma
.
Cell Rep
2016
;
15
:
857
65
.
11.
Llosa
NJ
,
Cruise
M
,
Tam
A
,
Wicks
EC
,
Hechenbleikner
EM
,
Taube
JM
, et al
The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints
.
Cancer Discov
2015
;
5
:
43
51
.
12.
Jasinski-Bergner
S
,
Mandelboim
O
,
Seliger
B
. 
The role of microRNAs in the control of innate immune response in cancer
.
J Natl Cancer Inst
2014
;
106
:
dju257
.
13.
Li
J
,
Tan
S
,
Kooger
R
,
Zhang
C
,
Zhang
Y
. 
MicroRNAs as novel biological targets for detection and regulation
.
Chem Soc Rev
2014
;
43
:
506
17
.
14.
Cheng
CJ
,
Bahal
R
,
Babar
IA
,
Pincus
Z
,
Barrera
F
,
Liu
C
, et al
MicroRNA silencing for cancer therapy targeted to the tumour microenvironment
.
Nature
2015
;
518
:
107
10
.
15.
Mima
K
,
Nishihara
R
,
Nowak
JA
,
Kim
SA
,
Song
M
,
Inamura
K
, et al
MicroRNA MIR21 and T cells in colorectal cancer
.
Cancer Immunol Res
2016
;
4
:
33
40
.
16.
Chen
XM
,
Splinter
PL
,
O'Hara
SP
,
LaRusso
NF
. 
A cellular micro-RNA, let-7i, regulates Toll-like receptor 4 expression and contributes to cholangiocyte immune responses against Cryptosporidium parvum infection
.
J Biol Chem
2007
;
282
:
28929
38
.
17.
Androulidaki
A
,
Iliopoulos
D
,
Arranz
A
,
Doxaki
C
,
Schworer
S
,
Zacharioudaki
V
, et al
The kinase Akt1 controls macrophage response to lipopolysaccharide by regulating microRNAs
.
Immunity
2009
;
31
:
220
31
.
18.
Almanza
G
,
Fernandez
A
,
Volinia
S
,
Cortez-Gonzalez
X
,
Croce
CM
,
Zanetti
M
. 
Selected microRNAs define cell fate determination of murine central memory CD8 T cells
.
PLoS One
2010
;
5
:
e11243
.
19.
Takamizawa
J
,
Konishi
H
,
Yanagisawa
K
,
Tomida
S
,
Osada
H
,
Endoh
H
, et al
Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival
.
Cancer Res
2004
;
64
:
3753
6
.
20.
Nam
EJ
,
Yoon
H
,
Kim
SW
,
Kim
H
,
Kim
YT
,
Kim
JH
, et al
MicroRNA expression profiles in serous ovarian carcinoma
.
Clin Cancer Res
2008
;
14
:
2690
5
.
21.
Lu
L
,
Katsaros
D
,
de la Longrais
IA
,
Sochirca
O
,
Yu
H
. 
Hypermethylation of let-7a-3 in epithelial ovarian cancer is associated with low insulin-like growth factor-II expression and favorable prognosis
.
Cancer Res
2007
;
67
:
10117
22
.
22.
King
CE
,
Wang
L
,
Winograd
R
,
Madison
BB
,
Mongroo
PS
,
Johnstone
CN
, et al
LIN28B fosters colon cancer migration, invasion and transformation through let-7-dependent and -independent mechanisms
.
Oncogene
2011
;
30
:
4185
93
.
23.
Liao
X
,
Lochhead
P
,
Nishihara
R
,
Morikawa
T
,
Kuchiba
A
,
Yamauchi
M
, et al
Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival
.
N Engl J Med
2012
;
367
:
1596
606
.
24.
Nishihara
R
,
Lochhead
P
,
Kuchiba
A
,
Jung
S
,
Yamauchi
M
,
Liao
X
, et al
Aspirin use and risk of colorectal cancer according to BRAF mutation status
.
JAMA
2013
;
309
:
2563
71
.
25.
Schmittgen
TD
,
Livak
KJ
. 
Analyzing real-time PCR data by the comparative C(T) method
.
Nat Protoc
2008
;
3
:
1101
8
.
26.
Ogino
S
,
Kawasaki
T
,
Kirkner
GJ
,
Loda
M
,
Fuchs
CS
. 
CpG island methylator phenotype-low (CIMP-low) in colorectal cancer: possible associations with male sex and KRAS mutations
.
J Mol Diagn
2006
;
8
:
582
8
.
27.
Ogino
S
,
Kawasaki
T
,
Brahmandam
M
,
Yan
L
,
Cantor
M
,
Namgyal
C
, et al
Sensitive sequencing method for KRAS mutation detection by Pyrosequencing
.
J Mol Diagn
2005
;
7
:
413
21
.
28.
Imamura
Y
,
Lochhead
P
,
Yamauchi
M
,
Kuchiba
A
,
Qian
ZR
,
Liao
X
, et al
Analyses of clinicopathological, molecular, and prognostic associations of KRAS codon 61 and codon 146 mutations in colorectal cancer: cohort study and literature review
.
Mol Cancer
2014
;
13
:
135
.
29.
Liao
X
,
Morikawa
T
,
Lochhead
P
,
Imamura
Y
,
Kuchiba
A
,
Yamauchi
M
, et al
Prognostic role of PIK3CA mutation in colorectal cancer: cohort study and literature review
.
Clin Cancer Res
2012
;
18
:
2257
68
.
30.
Ogino
S
,
Nosho
K
,
Kirkner
GJ
,
Kawasaki
T
,
Meyerhardt
JA
,
Loda
M
, et al
CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer
.
Gut
2009
;
58
:
90
6
.
31.
Ogino
S
,
Nosho
K
,
Kirkner
GJ
,
Kawasaki
T
,
Chan
AT
,
Schernhammer
ES
, et al
A cohort study of tumoral LINE-1 hypomethylation and prognosis in colon cancer
.
J Natl Cancer Inst
2008
;
100
:
1734
8
.
32.
Irahara
N
,
Nosho
K
,
Baba
Y
,
Shima
K
,
Lindeman
NI
,
Hazra
A
, et al
Precision of pyrosequencing assay to measure LINE-1 methylation in colon cancer, normal colonic mucosa, and peripheral blood cells
.
J Mol Diagn
2010
;
12
:
177
83
.
33.
Hu
G
,
Zhou
R
,
Liu
J
,
Gong
AY
,
Eischeid
AN
,
Dittman
JW
, et al
MicroRNA-98 and let-7 confer cholangiocyte expression of cytokine-inducible Src homology 2-containing protein in response to microbial challenge
.
J Immunol
2009
;
183
:
1617
24
.
34.
Kumar
M
,
Sahu
SK
,
Kumar
R
,
Subuddhi
A
,
Maji
RK
,
Jana
K
, et al
MicroRNA let-7 modulates the immune response to mycobacterium tuberculosis infection via control of A20, an inhibitor of the NF-kappaB pathway
.
Cell Host Microbe
2015
;
17
:
345
56
.
35.
Schulte
LN
,
Eulalio
A
,
Mollenkopf
HJ
,
Reinhardt
R
,
Vogel
J
. 
Analysis of the host microRNA response to Salmonella uncovers the control of major cytokines by the let-7 family
.
EMBO J
2011
;
30
:
1977
89
.
36.
Marcais
A
,
Blevins
R
,
Graumann
J
,
Feytout
A
,
Dharmalingam
G
,
Carroll
T
, et al
microRNA-mediated regulation of mTOR complex components facilitates discrimination between activation and anergy in CD4 T cells
.
J Exp Med
2014
;
211
:
2281
95
.
37.
Zhang
Y
,
Wang
X
,
Zhong
M
,
Zhang
M
,
Suo
Q
,
Lv
K
. 
MicroRNA let-7a ameliorates con A-induced hepatitis by inhibiting IL-6-dependent Th17 cell differentiation
.
J Clin Immunol
2013
;
33
:
630
9
.
38.
Colussi
D
,
Brandi
G
,
Bazzoli
F
,
Ricciardiello
L
. 
Molecular pathways involved in colorectal cancer: implications for disease behavior and prevention
.
Int J Mol Sci
2013
;
14
:
16365
85
.
39.
Kuipers
EJ
,
Grady
WM
,
Lieberman
D
,
Seufferlein
T
,
Sung
JJ
,
Boelens
PG
, et al
Colorectal cancer
.
Nat Rev Dis Primers
2015
;
1
:
15065
.
40.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
41.
Fridman
WH
,
Pages
F
,
Sautes-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
42.
Galon
J
,
Mlecnik
B
,
Bindea
G
,
Angell
HK
,
Berger
A
,
Lagorce
C
, et al
Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours
.
J Pathol
2014
;
232
:
199
209
.
43.
Rozek
LS
,
Schmit
SL
,
Greenson
JK
,
Tomsho
LP
,
Rennert
HS
,
Rennert
G
, et al
Tumor-infiltrating lymphocytes, Crohn's-like lymphoid reaction, and survival from colorectal cancer
.
J Natl Cancer Inst
2016
;
108
:
djw027
.
44.
Masugi
Y
,
Nishihara
R
,
Yang
J
,
Mima
K
,
da Silva
A
,
Shi
Y
, et al
Tumour CD274 (PD-L1) expression and T cells in colorectal cancer
.
Gut
2016
;
DOI 10.1136/gutjnl-2016–311421
.
45.
Rosenbaum
MW
,
Bledsoe
JR
,
Morales-Oyarvide
V
,
Huynh
TG
,
Mino-Kenudson
M
. 
PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes
.
Mod Pathol
2016
;
29
:
1104
12
.
46.
Akbay
EA
,
Koyama
S
,
Carretero
J
,
Altabef
A
,
Tchaicha
JH
,
Christensen
CL
, et al
Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors
.
Cancer Discov
2013
;
3
:
1355
63
.
47.
Schetter
AJ
,
Leung
SY
,
Sohn
JJ
,
Zanetti
KA
,
Bowman
ED
,
Yanaihara
N
, et al
MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma
.
JAMA
2008
;
299
:
425
36
.
48.
Slattery
ML
,
Herrick
JS
,
Mullany
LE
,
Valeri
N
,
Stevens
J
,
Caan
BJ
, et al
An evaluation and replication of miRNAs with disease stage and colorectal cancer-specific mortality
.
Int J Cancer
2015
;
137
:
428
38
.
49.
Ogino
S
,
Chan
AT
,
Fuchs
CS
,
Giovannucci
E
. 
Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field
.
Gut
2011
;
60
:
397
411
.
50.
Ogino
S
,
Nishihara
R
,
VanderWeele
TJ
,
Wang
M
,
Nishi
A
,
Lochhead
P
, et al
Review article: the role of molecular pathological epidemiology in the study of neoplastic and non-neoplastic diseases in the era of precision medicine
.
Epidemiology
2016
;
27
:
602
11
.