Abstract
Background:
Tumor-infiltrating lymphocytes (TILs) are a robust and independent prognostic variable in localized colon cancer. Given reported differences in molecular features and prognosis of right- versus left-sided tumors, we examined the association of TIL densities with patient survival by primary tumor sidedness in stage III cancers, including clinical low- (T1–3, N1) and high-risk (T4 and/or N2) groups.
Patients and methods:
In a phase III trial of FOLFOX-based adjuvant chemotherapy, TIL densities were analyzed and dichotomized in colon carcinomas (N=1532) based on a previously determined cutpoint optimized for disease-free survival (DFS). Right-sided tumors were defined as proximal to the splenic flexure. Associations of TILs and sidedness with 5-year DFS were examined using Kaplan-Meier methodology along with multivariable modeling and relative contribution analysis by Cox regression.
Results:
Lower TIL densities were found in left- vs right-sided tumors (P<0.0001). The association of TIL densities with DFS differed significantly by tumor sidedness (Pinteraction= .045). Overall, patient tumors with low (vs high) TILs had significantly poorer DFS in right-sided [HR=2.02 (95%CI: 1.45–2.82); Padj<0.0001], but not left-sided tumors (Padj=0.1731). Among clinical low-risk patients, low (vs high) TILs were adversely prognostic only in right-sided tumors (Padj=0.0058). Among high-risk patients, low TILs were prognostic independent of sidedness (Padj<.025). The relative contribution of TILs to DFS was substantially greater in right- vs left-sided tumors (24% vs 1.5%). In high-risk tumors, TILs had the highest relative contribution to DFS (42%) of all variables. In low-risk tumors, the contribution of TILs (16%) to DFS was second to KRAS.
Conclusions:
The association of TIL densities with patient survival differed by primary tumor sidedness and clinical risk group, suggesting that TILs should be interpreted in this context among stage III colon cancers.
ClinicalTrials.gov Identifier:
Keywords: Tumor-infiltrating lymphocytes, Colonic Neoplasms, Primary Tumor Site, Prognosis, Adjuvant Chemotherapy
Introduction
Colorectal cancer (CRC) is the second most common cause of cancer-related death in the United States.1 Identifying patients who are more likely to have disease recurrence and metastasis after surgical resection is a major unmet need. Tumor-infiltrating lymphocytes (TILs) reflect the host anti-cancer immune response.2, 3 In multiple studies in nonmetastatic CRC, TIL density in the tumor microenvironment (TME) has been shown to be a statistically significant and independent predictor of patient survival.4, 5 TIL densities are typically increased in tumors with deficient DNA mismatch repair (dMMR) which are more commonly located in the right colon.4, 6, as are tumors harboring BRAFV600E point mutations and the CpG island methylator phenotype.7, 8 Furthermore, differences in immune-related gene expression and tumor transcriptomic profiles, including consensus molecular subtypes (CMS), are observed in relationship to primary tumor sidedness.9 10 Primary tumor sidedness has become of much scientific interest given that it’s a source of biological heterogeneity and the finding that colon cancer prognosis11, 12 and response to treatment can differ by anatomic tumor location.13 In this regard, clinical benefit of anti-epidermal growth factor receptor (EGFR) antibody therapy in patients with metastatic CRC was limited to tumors of the left colon.13
We quantified TIL densities in colon cancers and determined whether their association with patient prognosis differs by primary tumor sidedness. Toward this objective, we examined the interaction between TIL densities and tumor sidedness in relationship to disease-free survival (DFS) in patients with stage III colon cancers treated in a phase 3 clinical trial of adjuvant FOLFOX-based chemotherapy (North Central Cancer Treatment Group [NCCTG] N0147). We also studied the association of TILs with DFS by sidedness in clinical low-risk (T1–3, N1) and high-risk (T4 and/or N2) groups that are used to guide the recommended duration of adjuvant treatment with a fluoropyrimidine plus oxaliplatin in patients with stage III colon cancers.14
Method
Study Population
Patients with resected stage III colonic adenocarcinomas had been treated for 6 months in a phase 3 clinical trial of adjuvant mFOLFOX6 chemotherapy with or without cetuximab (NCCTG N0147).15 No difference in patient outcome was seen by study arm,15 which allowed pooling of the data from both arms. TIL data was generated on a subset (N=1532) of patients based on tissue availability and cases are representative of the parent cohort. Use of tissue specimens and clinical data from the N0147 trial was approved by the Mayo Clinic Institutional Review Board. The parent clinical trial was conducted by the NCCTG (now part of Alliance for Clinical Trials in Oncology), and each participant signed an informed consent.
TILs and Sidedness
TIL densities in tumor tissues were determined by manual counting in routine histologic sections from surgical resections, as previously described.2 TIL densities were dichotomized (≤3/HPF) based on a previously determined cut-off that was shown to best separate patients for DFS in the study cohort.2 Primary tumor site was categorized as right-sided for cancers located proximal to the splenic flexure, or left-sided. Covariates included age (years), sex (female vs male), race (white vs others), Eastern Cooperative Oncology Group performance status (PS; 0 vs 1/2),16 tumor grade (high vs low), T stage (T1/T2 vs T3/T4), number of positive lymph nodes (LNs) (1–3 vs ≥4), risk group (low [T1–3, N1] vs high [T4 and/or N2]), DNA mismatch repair (MMR) (deficient MMR vs proficient MMR), KRAS (nonmutated vs mutated), BRAFV600E (present or absent). Methodologies used to determine MMR status and the mutational status of KRAS and BRAFV600E were described previously.2
Statistical Analysis
The primary study outcome was DFS, which was defined as the time from randomization to the first of either disease recurrence or death. The association of the dichotomous TILs and other covariates with tumor site was evaluated using the chi-square test. A Kaplan-Meier methodology17 and multivariable Cox regression model18 were used to analyze DFS by TILs and sidedness with adjustment for covariates including, age, PS, T/N stage, histologic grade, KRAS, BRAF, MMR and treatment arm. Adjusted hazard ratios (HRadj) and 95% confidence intervals (CI) are reported. Two-sided P-values are reported and P-values <0.05 were considered statistically significant for all analyses. SAS version 9.4 (SAS Institute, Cary NC) was utilized. The proportional hazards (PH) assumption was tested and found to be met in multivariable models from our study cohort (a priori p < 0.01 for variable to violate PH assumption given sample size). Data collection and statistical analyses were conducted by the Alliance Statistics and Data Management Center. All analyses were based on the study database frozen on [August 5, 2015]. The relative contribution of each variable to DFS was calculated using χ2 from Harrell’s rms R package (version 3.2.3; http://biostat.mc.vanderbilt.edu/rms) based on multivariable Cox regression models.
Results
Patient characteristics
Within our study cohort of 1532 patients, 783 (51%) had right-sided tumors, 731 (48%) had left-sided cancers, and 18 (1%) had missing data for tumor site. Mean patient age was 58 years (SD of 11 years) with a range of 19–86 years. Median patient follow-up time was 83 months. Patient characteristics stratified by primary tumor sidedness are shown in Table 1. Low TIL densities were significantly more common in patients with left-sided vs right-sided cancers (80% vs 68%, P<0.0001), which was maintained when the cohort was limited to pMMR tumors (81% vs 75%, P=0.025). A higher prevalence of high-grade tumors (P<0.0001) and those with mutations in KRAS (P=0.0005), BRAFV600E (P<0.0001) or dMMR (P<0.0001) were found in right-vs left-sided tumors. These differences remained statistically significant when restricting the cohort to tumors with pMMR (all P<0.0001). Neither T nor N stage nor risk groups (high, low) differed significantly based on tumor sidedness (Table 1).
Table 1.
Clinicopathological and molecular features of patients stratified by primary tumor site.
| Variablesa,b | Total (N=1532) | Tumor site | P-value | |
|---|---|---|---|---|
| Right-sided (N=783) | Left-sided (N=731) | |||
| Age, years N (%) | <0.0001c | |||
| <50 | 327 (21%) | 129 (16%) | 193 (26%) | |
| ≥50 | 1205 (79%) | 654 (84%) | 538 (74%) | |
| Sex, N (%) | 0.2148c | |||
| Female | 732 (48%) | 387 (49%) | 338 (46%) | |
| Male | 800 (52%) | 396 (51%) | 393 (54%) | |
| Race, N (%) | 0.4181c | |||
| White | 1349 (89%) | 696 (89%) | 639 (87%) | |
| Non-white | 163 (11%) | 78 (11%) | 82 (13%) | |
| T stage, N (%) | 0.0533c | |||
| T1 or T2 | 226 (15%) | 103 (13%) | 122 (17%) | |
| T3 or T4 | 1306 (85%) | 680 (87%) | 609 (83%) | |
| N stage, N (%) | 0.8210c | |||
| N1 (1–3 nodes) | 906 (59%) | 464 (59%) | 429 (59%) | |
| N2 (≥4 nodes) | 626 (41%) | 319 (41%) | 302 (41%) | |
| Risk group, N (%) | 0.9276c | |||
| Low (T1–3, N1) | 804 (53%) | 411 (53%) | 382 (52%) | |
| High (T4 and/or N2) | 728 (47%) | 372 (47%) | 349 (48%) | |
| Histologic grade, N (%) | <0.0001c | |||
| Low | 1152 (75%) | 533 (68%) | 606 (83%) | |
| High | 380 (25%) | 250 (32%) | 125 (17%) | |
| Performance status, N (%) | 0.0884c | |||
| 0 | 1161 (76%) | 579 (74%) | 568 (78%) | |
| 1/2 | 371 (24%) | 204 (26%) | 163 (22%) | |
| KRAS, N (%) | 0.0005c | |||
| Nonmutated | 968 (64%) | 462 (60%) | 493 (69%) | |
| Mutated | 536 (36%) | 307 (40%) | 224 (31%) | |
| BRAFV600E, N (%) | <0.0001c | |||
| Absent | 1262 (86%) | 574 (77%) | 671 (96%) | |
| Present | 198 (14%) | 169 (23%) | 28 (4%) | |
| MMR status, N (%) | <0.0001c | |||
| dMMR | 153 (10%) | 137 (18%) | 13 (2%) | |
| pMMR | 1357 (90%) | 635 (82%) | 708 (98%) | |
| TILs, N (%) | <0.0001c | |||
| Low (≤3/HPF) | 1132 (74%) | 534 (68%) | 584 (80%) | |
| High (>3/HPF) | 400 (26%) | 249 (32%) | 147 (20%) | |
dMMR, deficient mismatch repair; pMMR, proficient mismatch repair; TILs, tumor-infiltrating lymphocytes.
Missing values are not included in the percentages.
Sum of the numbers from each subcategory may not equal the total reported cases due to missing values.
Chi-square test.
Association of TILs and sidedness with disease-free survival (DFS)
In the overall cohort, patients whose tumors showed low vs high TILs had significantly poorer DFS [HRadj 1.74 (95%CI, 1.35–2.24); Padj<0.0001] (Table 2) that was also seen when the cohort was limited to pMMR tumors [HR 1.65 (95%CI, 1.25–2.16); Padj=0.0002]. We found an interaction between TILs and sidedness, but not other variables, for DFS that was statistically significant after adjustment for covariates (Pinteraction(adj) =0.045)[Supplementary Table 1]. Accordingly, we compared DFS rates by TILs and sidedness and found that low (vs high) TILs was significantly associated with poorer DFS in right-sided [HRadj 2.02 (95% CI, 1.45–2.82); Padj<0.0001, 5 yr DFS 57% vs 77%], but not in left-sided tumors [HRadj 1.30 (95%CI, 0.88–1.92); Padj=0.173, 5 yr DFS 69% vs 76%] (Figure 1). Analysis based on clinical risk group revealed similar results whereby low risk tumors had significantly poorer DFS for low vs high TILs that was limited to right-sided cancers (Padj=0.0058). Among high risk tumors, however, low vs high TILs were associated with significantly poorer DFS irrespective of sidedness (right: Padj=0.0004 vs left: Padj=0.0244) (Table 2).
Table 2.
Disease–free survival for colon cancer patients stratified by TILs and sidedness among overall population, high and low risk cases.
| Population | Tumor site | HRadj (95%CI)¥ | 5-year DFS | P adj | |
|---|---|---|---|---|---|
| Low TILs | High TILs | ||||
| Overall | Right | 2.02 (1–45–2.82) | 57% | 77% | <0.0001 |
| Left | 1.30 (0.88–1.92) | 69% | 76% | 0.1731 | |
| Overall | 1.74 (1.35–2.24) | 63% | 77% | <0.0001 | |
| Low risk (T1–3, N1) | Right | 2.07 (1.20–3.56) | 70% | 84% | 0.0058 |
| Left | 1.03 (0.60–1.76) | 78% | 79% | 0.9135 | |
| Overall | 1.52 (1.04–2.23) | 74% | 81% | 0.0264 | |
| High risk (T4 and/or N2) | Right | 2.05 (1.34–3.13) | 44% | 68% | 0.0004 |
| Left | 1.86 (1.04–3.32) | 60% | 71% | 0.0244 | |
| Overall | 2.00 (1.43–2.81) | 53% | 69% | <0.0001 | |
TILs, tumor-infiltrating lymphocytes; DFS, disease-free survival
Age, Performance status, T-stage, N-stage, Histological grade, KRAS, BRAS, Mismatch repair status and treatment arm were used in adjustment of hazard ratio.
Figure 1.

Association of TILs and DFS for stage III colon cancer patients in overall cohort and by clinical risk group. Left-sided tumors in overall cohort (left panel); Right-sided tumors in overall cohort (right panel).
Given the observed interaction between TILs and sidedness, we examined their combined variable (4 groups: TIL low/right-sided, TIL low/left-sided; TIL high/right-sided, TIL high/left-sided) in relationship to DFS. Patients with TIL low/right-sided tumors had the poorest DFS in the overall cohort [HRadj 2.12 (95%CI, 1.53–2.95); Padj<0.0001 vs reference] (Figure 2A) and when the cohort was restricted to pMMR tumors [HRadj 2.05 (95% CI, 1.40–3.01); Padj=0.0003]. Analysis by clinical risk group was then performed. In both low risk and high risk groups, TIL low/right sided tumors had the poorest DFS [low risk: HRadj 2.09 (95%CI, 1.23 –3.55), Padj=0.0402, (Figure 2B); high risk: HRadj 2.14 (95%CI, 1.41–3.27), Padj<0.0001, (Figure 2C)]. Importantly, similar outcomes were seen for low risk tumors that were TIL low/right-sided (5 yr DFS: 70%) and for high risk tumors with TILs high irrespective of sidedness (5-yr DFS: right, 68%; left, 71%).
Figure 2.

Kaplan–Meier estimates of disease–free survival for colon cancer patients with TIL high/right-sided, TIL high/left-sided, TIL low/right-sided, TIL low /left-sided. (A) overall population; (B) low risk patients [T1–3, N1], (C) high risk patients [T4 and/or N2].
A forest plot was constructed based on results of multivariable regression analysis of TIL/sidedness and other tumor-related variables associated with DFS in the overall patient population (Table 3). In the overall cohort, TIL/sidedness as well as T stage, N stage, and KRAS were each significantly and independently associated with patient DFS. We have previously shown that the association of BRAFV600E with clinical outcome is attenuated by the enrichment for BRAFV600E in dMMR tumors in patients.19, 20 A similar multivariable analysis in clinical low and high risk group was performed as shown in Table 4. While low TILs and KRAS mutation were adversely prognostic in both risk groups, dMMR vs pMMR was only prognostic in high risk tumors where it was associated with poorer DFS. Multivariable models were also adjusted for study treatment arm (cetuximab-containing vs control). To further support our findings, we generated minimally adjusted multivariable models that included MMR or TILs along with covariates (age, T stage, N stage, histologic grade, PS) [data not shown]. In these models, we found that in contrast to MMR, TILs were significantly associated with DFS indicating that TILs are a more robust prognostic variable.
Table 3.
Forest plot based on results of multivariable analysis of tumor-related variables associated with DFS in the overall patient population.
|
dMMR, deficient mismatch repair; pMMR, proficient mismatch repair; TILs, tumor-infiltrating lymphocytes; PS, performance status; WT, wild-type.
Table 4.
Forest plot based on results of multivariable analysis of tumor-related variables associated with DFS in clinical low risk (T1–3, N1) and high risk (T4 and/or N2) groups.
|
dMMR, deficient mismatch repair; pMMR, proficient mismatch repair; TILs, tumor-infiltrating lymphocytes; PS, performance status; WT, wild-type.
Analysis of the relative contribution (%) of each variable to DFS was then examined multivariately. In the overall population, the top three contributors to DFS were N stage (44%), TILs (15%), and KRAS mutation (14%). The relative contribution of TILs to DFS was substantially greater in right-sided tumors (right: 24% vs left: 1.5%) and was only exceeded by N stage (Figure 3 A, B). Among low risk tumors, the relative contribution of TILs to DFS (16%) was second only to KRAS status (Figure 3 C). For high risk tumors, TILs had the highest relative contribution to DFS (42%) followed by tumor sidedness (14%) [Figure 3 D].
Figure 3.

Relative contributions (%) of the variables in multivariable Cox models for prediction of DFS in (A) left-sided and (B) right-sided tumors in the overall cohort; and patients categorized into clinical (C) low risk and (D) high risk groups.
Discussion
Based on reported differences in immune cell composition, molecular subtypes, and microbial composition by anatomic tumor location,7, 21–23 we analyzed TIL densities and their association with DFS based on tumor sidedness. Consistent with prior studies by ourselves and others, we found significantly higher TIL densities in right- versus left-sided tumors. Moreover, a low vs high TIL density was significantly and independently associated with poorer DFS in patients with stage III colon cancers.2–4, 24, 25 In contrast to MMR that was not prognostic in the overall cohort, TILs were significantly associated with DFS multivariately indicating that TILs are a more robust prognostic variable. Next, we demonstrated that the association of TIL densities with DFS differed significantly by primary tumor sidedness. Specifically, low TIL densities were predictive of significantly poorer DFS in right-sided, but not left-sided cancers after adjustment for KRAS/BRAF, MMR status, and other covariates. Given the significant interaction found between TILs and sidedness, we then examined their combined variable in relationship to DFS and found that patients with low TIL/right-sided tumors had the poorest DFS in the overall cohort and in both clinical risk groups. Within each clinical risk group, the combined variable was able to prognostically stratify patients. In clinical high (vs low) risk patients, those with TILs low/right-sided tumors had markedly shorter 5-year DFS (44% vs 70%). Furthermore, patient tumors with high TILs of either anatomic location had similarly favorable survival rates. Of note, no difference was observed between TILs and DFS by treatment arm overall or in left-sided tumors from the N0147 or PETACC-8 clinical trial cohorts,26 in contrast to prior data demonstrating the predictive utility of tumor sidedness for response to anti-EGFR therapy in metastatic CRC.13
Our observation that low TIL/right-sided tumors had the poorest DFS in the overall cohort and in clinical risk groups deserves comment. We found higher TIL densities in right- versus left-sided tumors (even after exclusion of dMMR cancers) and in a prior study in the N0147 cohort, increased TILs included CD3+ and cytotoxic CD8+ T lymphocytes.3 Gene expression data have also shown that right- versus left-sided colon cancers have increased immune cells with enhanced cytotoxic function and interferon-Ɣ signatures shown to activate innate immunity and inhibit tumorigenesis.27, 28 Potentially, higher TIL densities including cytotoxic T-cells may be needed to overcome an immunosuppressive TME in the right colon. We found that BRAFV600E and KRAS mutations were more common in the right colon and both of these oncogenes have been shown to promote an immunosuppressive TME.29–31 Interestingly, the association of FOXP3+ regulatory T-cells (Tregs) with poor survival was restricted to colon cancers with low CD4+ or low cytotoxic CD8+ T-cells infiltration.32, 33 While Tregs can suppress inflammation and promote better outcome in colon cancer, they were shown to suppress anti-tumor immunity with expression of RORƳt.34 Another factor shown to differ by tumor sidedness that contributes to biological heterogeneity is the gut flora35–38 which can modulate anti-tumor immunity.21, 22, 39 A potential contributor to immune suppression in the TME of the right colon is the anaerobic bacterium Fusobacterium nucleatum whose DNA content was shown to be significantly increased in right- vs left-sided colon cancers35 where it was inversely related to CD3+ T-cell density.36, 37 F. nucleatum has also been shown to be associated with adverse outcome in patients with CRC.40, 41
We examined the relative contribution of TILs for the prediction of patient DFS. In the overall cohort, TILs were second only to N stage for prediction of DFS and their relative contribution was substantially greater, i.e., 16-fold, in right- vs left-sided cancers. Analysis by clinical risk group revealed that TIL density had the highest relative contribution to DFS among high risk tumors that was followed by sidedness. Among low risk tumors, TILs were second to KRAS then followed by MMR for prediction of DFS.
Strengths of this study include same stage patients from a well-characterized clinical trial cohort with mature survival data. While our findings await validation, we studied a relatively large clinical trial cohort of well annotated cases with meticulous documentation of patient outcomes. Limitations include the fact that all patients received adjuvant chemotherapy for 6 months such that predictive utility of TILs for chemotherapy outcomes could not be studied. In conclusion, patient tumors with low TILs and right-sided location had the poorest DFS in the overall cohort and within clinical risk groups. If validated in a separate clinical trial cohort, these findings suggest that TIL densities should be interpreted in the context of tumor site for prognostication.
Supplementary Material
Highlights.
Association of TIL densities with 5-year DFS differed significantly by primary tumor sidedness.
Overall, low vs high TILs were significantly associated with poorer DFS among right-sided, but not left-sided tumors.
Prognostic impact of TILs was limited to right-sided cancers among clinical low risk tumors.
Relative contribution of TILs to DFS was greater in right- vs left-sided tumors (24% vs 1.5%).
In clinical high-risk tumors, TILs had the highest relative contribution to DFS of all variables.
Support
Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under Awards Numbers R01 CA210509 (to FAS), U10CA180821, U10CA180882, and U24CA196171 (to the Alliance for Clinical Trials in Oncology), and UG1CA232760. Also supported in part by funds from Sanofi. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. https://acknowledgments.alliancefound.org.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ClinicalTrials.gov Identifier: NCT00079274
Conflict of interest statement
The authors report no relevant conflicts of interest related to the content of this manuscript.
References
- 1.Siegel RL, Miller KD, Goding Sauer A et al. Colorectal cancer statistics, 2020. CA Cancer J Clin 2020; 70 (3): 145–164. [DOI] [PubMed] [Google Scholar]
- 2.Lee H, Sha D, Foster NR et al. Analysis of tumor microenvironmental features to refine prognosis by T, N risk group in patients with stage III colon cancer (NCCTG N0147) (Alliance). Ann Oncol 2020; 31 (4): 487–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Yoon HH, Shi Q, Heying EN et al. Intertumoral Heterogeneity of CD3(+) and CD8(+) T-Cell Densities in the Microenvironment of DNA Mismatch-Repair-Deficient Colon Cancers: Implications for Prognosis. Clin Cancer Res 2019; 25 (1): 125–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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 (5795): 1960–1964. [DOI] [PubMed] [Google Scholar]
- 5.Pages F, Mlecnik B, Marliot F et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018; 391 (10135): 2128–2139. [DOI] [PubMed] [Google Scholar]
- 6.Ogino S, Nosho K, Irahara N 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 (20): 6412–6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kim K, Castro EJT, Shim H et al. Differences Regarding the Molecular Features and Gut Microbiota Between Right and Left Colon Cancer. Ann Coloproctol 2018; 34 (6): 280–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Weisenberger DJ, Siegmund KD, Campan M et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 2006; 38 (7): 787–793. [DOI] [PubMed] [Google Scholar]
- 9.Guo JN, Li MQ, Deng SH et al. Prognostic Immune-Related Analysis Based on Differentially Expressed Genes in Left- and Right-Sided Colon Adenocarcinoma. Front Oncol 2021; 11: 640196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kim SR, Song N, Yothers G et al. Tumour sidedness and intrinsic subtypes in patients with stage II/III colon cancer: analysis of NSABP C-07 (NRG Oncology). Br J Cancer 2018; 118 (5): 629–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Akce M, Zakka K, Jiang R et al. Impact of Tumor Side on Clinical Outcomes in Stage II and III Colon Cancer With Known Microsatellite Instability Status. Front Oncol 2021; 11: 592351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Petrelli F, Tomasello G, Borgonovo K et al. Prognostic Survival Associated With Left-Sided vs Right-Sided Colon Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2017; 3 (2): 211–219. [DOI] [PubMed] [Google Scholar]
- 13.Holch JW, Ricard I, Stintzing S et al. The relevance of primary tumour location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur J Cancer 2017; 70: 87–98. [DOI] [PubMed] [Google Scholar]
- 14.Grothey A, Sobrero AF, Shields AF et al. Duration of Adjuvant Chemotherapy for Stage III Colon Cancer. N Engl J Med 2018; 378 (13): 1177–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Alberts SR, Sargent DJ, Nair S et al. Effect of oxaliplatin, fluorouracil, and leucovorin with or without cetuximab on survival among patients with resected stage III colon cancer: a randomized trial. JAMA 2012; 307 (13): 1383–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Oken MM, Creech RH, Tormey DC et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol 1982; 5 (6): 649–655. [PubMed] [Google Scholar]
- 17.Kaplan EL, Meier P. Nonparametric Estimation from Incomplete Observations. Journal of the American Statistical Association 1958; 53 (282): 457–481. [Google Scholar]
- 18.Cox DR. Regression Models and Life-Tables. Journal of the Royal Statistical Society Series B (Methodological) 1972; 34 (2): 187–220. [Google Scholar]
- 19.Sinicrope FA, Shi Q, Smyrk TC et al. Molecular markers identify subtypes of stage III colon cancer associated with patient outcomes. Gastroenterology 2015; 148 (1): 88–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Taieb J, Le Malicot K, Shi Q et al. Prognostic Value of BRAF and KRAS Mutations in MSI and MSS Stage III Colon Cancer. J Natl Cancer Inst 2017; 109 (5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hurtado CG, Wan F, Housseau F et al. Roles for Interleukin 17 and Adaptive Immunity in Pathogenesis of Colorectal Cancer. Gastroenterology 2018; 155 (6): 1706–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Song M, Chan AT, Sun J. Influence of the Gut Microbiome, Diet, and Environment on Risk of Colorectal Cancer. Gastroenterology 2020; 158 (2): 322–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhong M, Xiong Y, Ye Z et al. Microbial Community Profiling Distinguishes Left-Sided and Right-Sided Colon Cancer. Front Cell Infect Microbiol 2020; 10: 498502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Salama P, Phillips M, Grieu F et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 2009; 27 (2): 186–192. [DOI] [PubMed] [Google Scholar]
- 25.Sinicrope FA, Shi Q, Hermitte F et al. Contribution of Immunoscore and Molecular Features to Survival Prediction in Stage III Colon Cancer. JNCI Cancer Spectr 2020; 4 (3): pkaa023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Taieb J, Kourie HR, Emile JF et al. Association of Prognostic Value of Primary Tumor Location in Stage III Colon Cancer With RAS and BRAF Mutational Status. JAMA Oncol 2018; 4 (7): e173695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang L, Zhao Y, Dai Y et al. Immune Landscape of Colorectal Cancer Tumor Microenvironment from Different Primary Tumor Location. Front Immunol 2018; 9: 1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Du W, Frankel TL, Green M et al. IFNgamma signaling integrity in colorectal cancer immunity and immunotherapy. Cell Mol Immunol 2022; 19 (1): 23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hamarsheh S, Gross O, Brummer T et al. Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun 2020; 11 (1): 5439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Petanidis S, Anestakis D, Argyraki M et al. Differential expression of IL-17, 22 and 23 in the progression of colorectal cancer in patients with K-ras mutation: Ras signal inhibition and crosstalk with GM-CSF and IFN-gamma. PLoS One 2013; 8 (9): e73616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Khalili JS, Liu S, Rodriguez-Cruz TG et al. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin Cancer Res 2012; 18 (19): 5329–5340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kuwahara T, Hazama S, Suzuki N et al. Intratumoural-infiltrating CD4 + and FOXP3 + T cells as strong positive predictive markers for the prognosis of resectable colorectal cancer. Br J Cancer 2019; 121 (8): 659–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yoon HH, Orrock JM, Foster NR et al. Prognostic impact of FoxP3+ regulatory T cells in relation to CD8+ T lymphocyte density in human colon carcinomas. PLoS One 2012; 7 (8): e42274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Blatner NR, Mulcahy MF, Dennis KL et al. Expression of RORgammat marks a pathogenic regulatory T cell subset in human colon cancer. Sci Transl Med 2012; 4 (164): 164ra159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mima K, Cao Y, Chan AT et al. Fusobacterium nucleatum in Colorectal Carcinoma Tissue According to Tumor Location. Clin Transl Gastroenterol 2016; 7 (11): e200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mima K, Sukawa Y, Nishihara R et al. Fusobacterium nucleatum and T Cells in Colorectal Carcinoma. JAMA Oncol 2015; 1 (5): 653–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Nosho K, Sukawa Y, Adachi Y et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol 2016; 22 (2): 557–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Flemer B, Lynch DB, Brown JM et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 2017; 66 (4): 633–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Cremonesi E, Governa V, Garzon JFG et al. Gut microbiota modulate T cell trafficking into human colorectal cancer. Gut 2018; 67 (11): 1984–1994. [DOI] [PubMed] [Google Scholar]
- 40.Chen Y, Lu Y, Ke Y et al. Prognostic impact of the Fusobacterium nucleatum status in colorectal cancers. Medicine (Baltimore) 2019; 98 (39): e17221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lee JB, Kim K-A, Cho HY et al. Association between Fusobacterium nucleatum and patient prognosis in metastatic colon cancer. Scientific Reports 2021; 11 (1): 20263. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
