Skip to main content

Correlation and prognostic implications of intratumor and tumor draining lymph node Foxp3+ T regulatory cells in colorectal cancer

Abstract

Background

The prognostic value of intratumor T regulatory cells (Tregs) in colorectal cancer (CRC) was previously reported, but the role of these cells in tumor draining lymph nodes (TDLNs) was less addressed.

Methods

A total of 150 CRC stages I-IV were retrospectively enrolled. Intratumor and TDLN Tregs were examined by immunohistochemical assay. The association of these cells was estimated by Pearson correlation. Survival analyses of subgroups were conducted by Kaplan–Meier curves, and the log-rank test and risk factors for survival were tested by the Cox proportional hazard model.

Results

High accumulation of Tregs in tumors was significant in patients with younger age and good histological grade, where enrichment of these cells in TDLNs was more apparent in those with node-negative disease and early TNM stage disease, both of which were more common in early T stage cases. A significant correlation of intratumoral and TDLN Tregs was detected. Patients with higher intratumoral Tregs displayed significantly better PFS and OS than those with lower Tregs. However, no such differences were found, but a similar prognostic prediction trend was found for these cells in TDLNs. Finally, intratumoral Tregs were an independent prognostic factor for both PFS (HR = 0.97, 95% CI 0.95–0.99, P < 0.01) and OS (HR = 0.98, 95% CI 0.95–1.00, P = 0.04) in the patients.

Conclusions

Higher intratumor Tregs were associated with better survival in CRC. Although no such role was found for these cells in TDLNs, the positive correlation and similar prognostic prediction trend with their intratumoral counterparts may indicate a parallelized function of these cells in CRC.

Peer Review reports

Introduction

Colorectal cancer (CRC) remains one of the main cancers worldwide, accounting for 9.8% of all new cases and 9.2% of all deaths for all cancers combined in 2021 [1]. In addition, the disease has become increasingly popular among young adults over the last 25 years [2]. Searching for a simple, reliable prognostic marker for the disease is thus of pivotal importance in practice.

T regulatory cells (Tregs), which are specifically labeled by forkhead box transcription Factor 3 (Foxp3) [3], are a small heterogeneous subset of CD4 + T cells. The main function of these cells was thought to block antitumor immune responses in cancer patients, and not surprisingly, increased counts of these cells in tumors could not only favor the development or growth of malignant cells but also influence the outcome of the patients [4]. The notorious role of these cells in predicting survival in malignancies was validated by a group of reports in gastric cancer [5], pancreatic cancer [6], breast cancer [7], ovarian cancer [8], and non-small cell lung cancer [9]. Nonetheless, the correlation of these cells with dismal outcome was not robustly established in CRC. Previously, although a series of reports concluded that the accumulation of intratumoral Tregs could predict good survival [10,11,12,13], there were also studies indicating that single Tregs were insufficient to predict prognosis [14,15,16]. To date, the paradox role of Tregs in predicting prognosis in CRC still needs further investigation.

Notably, Tregs were also found to be increased in other locations, such as peripheral blood in CRC [17]. TDLNs, which conventionally referred to pericolic, intermediate and additional main nodes for CRC patients underwent lymphadenectomy [18, 19], are an important sites where lymphocytes encounter tumor-specific antigens and generate antitumor immunity [20]. It was expected that the immune status of these nodes would have a profound effect on the spread of cancer [21]. Previously, the accumulation of Tregs was found to be significantly higher in TDLNs in many cancers [22,23,24,25] and was thought to promote cancer development. Similarly, in CRC, Gai et al. included 20 patients with metastatic TDLNs (mTDLNs) and 32 patients with metastasis-free TDLNs (mfTDLNs) and found that Tregs were significantly enriched in mTDLNs [26]; in line with this, Kazama et al. enrolled 50 patients and found that Tregs were enriched in regional lymph nodes, particularly in those near the lesions [27]. However, Deng et al. collected 10 patients with mTDLNs and 22 cases with mfTDLNs and found no such difference (only reported stage III cases), but they found that Tregs in TDLNs rather than those in tumors and peripheral blood were positively associated with disease stage [28]. However, these studies may be biased by their low number of cases and did not test the correlation of these cells with those in tumors or the individual prognostic value.

In this study, we aimed to detect Tregs in tumors as well as TDLNs and to determine the underlying correlation and their individual prognostic value. Furthermore, we compared their prognostic efficacy with other robust prognostic indicators, including the neutrophil to lymphocyte ratio (NLR), lymphocyte to monocyte ratio (LMR), and prognostic nutritional index (PNI), in CRC.

Patients and methods

Patient enrollments

From December 2012 to January 2018, CRC patients with radical recession of primary lesions in Hainan Hospital of Chinese PLA General Hospital were enrolled, and patients were excluded if they met any one of the following criteria: 1. neoadjuvant therapies; 2. missing laboratory tests within a week before surgery or key information in postoperative pathological reports; 3. multiple or recurrent malignancies or in situ lesions; or 4. loss to follow-up or less than 36 months. Other clinicopathological parameters, including body mass index (BMI) and tumor size (TS), were collected as previously reported [29,30,31]. The study was conducted in accordance with the principles stated in the Declaration of Helsinki and was approved by the ethics committee of Hainan Hospital of Chinese PLA General Hospital (ID: 301HLFYLS15), written informed consent was obtained from the patients or their authorized relatives.

Immunohistochemical staining of Foxp3 in tumors and TDLNs

Resected fresh samples were immediately processed by standard histological methods, and the staining was carried out as follows. In brief, 5-µm slides were cut for both tumors (samples with necrosis and broken areas were excluded) and TDLNs, and one node was randomly selected either in cases with mTDLNs or mfTDLNs (including those with tumor deposits). After deparaffinization, sodium citrate solution was used for antigen retrieval for 20 min and then cooled to room temperature. Subsequently, 3% hydrogen peroxide was used for blocking for 10 min. After washing, nonspecific antigen blocking was then performed with 5% goat serum for 30 min at room temperature (cat. no. C0265; Beyotime Institute of Biotechnology). After washing, the primary rabbit anti-human Foxp3 antibody (dilution 1:50; cat. no. #98377; CST) was incubated at 4 °C overnight, and the negative control was conducted by replacement of the primary antibody with identically diluted 5% nonimmunized rabbit serum (cat. no. A7016, Beyotime Institute of Biotechnology). After washing, goat anti-rabbit IgG H&L (HRP)-preadsorbed secondary antibody (dilution 1:500, cat. no. ab7090; Abcam) were added for 30 min at room temperature. Staining was then achieved by adding 3,3′diaminobenzidine (DAB, cat. no. ab64238; Abcam) according to the manufacturer’s protocol, the slides were then washed and mounted. The results were read under a light microscope at a magnification of × 200 (BX51, Olympus Corporation) by two independent pathologists who were blinded to the clinical information. The absolute number of Tregs in the tumor and in TDLNs was counted as follows: for each slides, five hot spots in 10 high power field (HPF) with the highest number of positive cells were selected, and JPEG images were taken by using a digital camera, then the number of positive cells in the images were counted by using ImageJ software as previous report [32]; patients were then divided into low or high subgroups by the mean counts as previously reported [9, 11].

Calculation of inflammation prognostic indicators

Routine laboratory data were collected as described in our previous report [31], and the NLR [33], LMR [34], and PNI [35] were estimated according to previous reports.

Definition of progression-free survival (PFS) and overall survival (OS)

The follow-up was conducted as scheduled in a previous report [31]. PFS was defined as the date of operation to the point of first recurrence of any location, disease progression according to the RECIST (version 1.1) [36] or death from any cause. OS was defined as the point of operation to the date of any cause of death. The latest follow-up point was in June 2021.

Statistical analysis

Statistical analyses were conducted by SPSS 20.0 (SPSS Inc., Chicago, IL, USA). Differences in clinicopathological parameters in Tregs-low or high in tumors and TDLNs were determined by χ2-test, Student’s t, or nonparametric rank sum test when appropriate. The association of Tregs in tumors and TDLNs, as well as with systemic inflammation markers, was determined by Pearson correlation. Survival differences for Treg-low or Treg-high groups were determined by Kaplan–Meier (K-M) analysis followed by log-rank tests. Risk factors for survival were estimated by a Cox proportional hazards model. Double-sided P < 0.05 was considered statistically significant.

Results

Patient characteristics and the differences in clinicopathological parameters in intratumoral and TDLN high or low Tregs

In total, 290 patients were enrolled, and 150 patients (94 males, 56 females) were included in the final analysis (Fig. 1). The mean age of the patients was 60.36 y (range: 24–85 y), and the mean follow-up was 47.61 m (range 1–102 m). A total of 2183 nodes were harvested with 279 mTDLNs. In addition, three patients with mfTDLNs and 10 patients with mTDLNs presented tumor deposits. Tregs were differently expressed in intratumoral and mTDLNs (Fig. 2), the number of the Tregs ranged from 0 to 110.0 (mean: 20.03), 0 to 260 (mean: 43.49) per HPF respectively. And approximately 1/3 (50/150 and 49/150, respectively) of patients displayed intratumoral Tregs and TDLN-high Tregs. As shown in Table 1, Tregs with high intratumoral frequencies were significantly more obvious in younger and good histological grade patients (both P < 0.01), where TDLNs with high frequencies were more apparent in node-negative (P < 0.01) and early TNM stage (P = 0.02) patients, and both of which were more common in early T stages (P < 0.01 and P = 0.03, respectively).

Fig. 1
figure 1

Flow diagram of the study

Fig. 2
figure 2

Accumulation of Tregs in tumor and in mTDLNs. A High Tregs in tumor; B No obvious Tregs in tumor; C High Tregs in mTDLNs; D Low Tregs in mTDLNs. Positive Tregs are indicated by black arrows with a magnification × 200

Table 1 Comparison of differences for the clinicopathological parameters in low or high Foxp3+ Tregs intratumor or TDLNs

Correlation of intratumor and TDLN Tregs and prognostic inflammation indicators

By Pearson correlation analysis, a significant correlation was found for intratumor and TDLN Tregs. In addition, intratumoral Tregs were also positively correlated with preoperative LMR and TS; however, no such correlation was found for TDLN Tregs (Table 2).

Table 2 Correlation of intratumor and TDLNs Tregs with NLR, LMR, PNI and TS

Survival differences of intratumor or TDLN high- or low-Treg subgroups in PFS and OS

Significant differences in intratumoral high and low Tregs in the 3-year PFS (14.00% vs. 37.00%, P < 0.01) and OS (12.00% vs. 27.00%, P = 0.04) rates could be detected, but no such differences could be found for TDLN Tregs in PFS (24.49% vs. 31.68%, P = 0.45) and OS (16.33% vs. 24.75%, P = 0.30). By K-M analyses, patients with intratumor high Tregs had a significantly better PFS (Tregs-high vs. low: 51.56 ± 18.75 m vs. 43.00 ± 24.61 m, Log rank = 7.68, P < 0.01) and OS (Tregs-high vs. low: 53.58 ± 17.30 m vs. 46.95 ± 21.94 m, Log rank = 4.65, P = 0.03) than those with Tregs-low (Fig. 3A, C). In addition, although patients with Tregs with high or low TDLNs did not show such differences in PFS (Tregs-high vs. low: 49.88 ± 21.72 m vs. 43.90 ± 23.62 m, Log rank = 1.57, P = 0.21) and OS (Tregs-high vs. low: 53.45 ± 18.10 m vs. 47.08 ± 21.62 m, Log rank = 1.72, P = 0.19), a similar survival difference trend could be found (Fig. 3B, D).

Fig. 3
figure 3

Prognostic role of Tregs in tumors and in TDLNs for PFS and OS. A, C. High Tregs in tumors predict superior PFS (A) and OS (C); B, D High or low Tregs in TDLNs display no significant differences in PFS (B) and OS (D)

Univariate and multivariate analyses of the prognostic risks for PFS and OS

Univariate tests indicated that CEA status, invasive depth, node involvement, distant metastasis, TNM stages, preoperative LMR and PNI, and intratumor Tregs were significant prognostic factors for PFS (Table 3) and OS (excluding preoperative LMR) (Table 4), and when all these factors (only those P < 0.05 in Tables 3, 4) were included in multivariate tests, the results indicated that intratumor Tregs were an independent prognostic factor for both PFS (HR = 0.97, 95% CI 0.95–0.99, P < 0.01) and OS (HR = 0.98, 95% CI 0.95–1.00, P = 0.04).

Table 3 Univariate and multivariate analyses of different parameters for PFS
Table 4 Univariate and multivariate analyses of different parameters for OS

Discussion

In the present study, we found that Tregs accumulated differently in tumors and TDLNs. Although only intratumoral Tregs showed a significant prognostic value for patient survival, a positive correlation of these cells was established for those located in tumors and TDLNs; in addition, a similar prognostic trend of these cells in TDLNs as in tumors was observed. To the best of our knowledge, our study includes the largest sample to explore Tregs in TDLNs and examine their prognostic role as well as correlation with their intratumoral counterparts in CRC.

Taking into consideration its negative role in manipulating antitumor immune responses, the accumulation of Tregs is regarded as an indicator of poor survival in many cancers [5,6,7,8,9]. Commonly, enrichment of these cells is also associated with clinicopathological parameters such as node metastasis and vascular, lymphatic, or perineural invasion [37,38,39]. However, the notorious role of Tregs in survival prediction is still a matter of debate in CRC. For example, Katz et al. included 188 patients who underwent resection of liver metastases and found that single Tregs were not sufficient to predict recurrence-free survival [14]. Sinicrope et al. collected 160 stage II-III patients and found that Tregs were not prognostic [16]. However, studies with large samples yield different conclusions. For example, Frey et al. included 1420 patients staged I-III and found that high frequency intratumoral Tregs were associated with early T stage and improved 5-year survival rate and Tregs were independent prognostic factor mismatch-repair (MMR)-proficient cases [40]. Salama et al. enrolled 967 staged II-III patients and found that a high density of Tregs correlated with improved survival [41]. Our study also supports that the accumulation of Tregs in tumors suggests superior survival in CRC, which is in line with previous reports [40, 41]. To date, although studies robustly support the positive role of Tregs in prognosis in CRC, the underlying mechanism is still poorly understood. To this end, Ladoire et al. argued that Tregs could attenuate Th17 cell-dependent proinflammatory and tumor-enhancing responses, the latter of which was important in manipulating cancer cell growth, constituting a possible explanation for their favorable role in CRC prognosis [42]. In addition, Saito et al. showed that Tregs in tumors can be classified into Foxp3hi and Foxp3lo subsets, and the latter could release inflammatory cytokines and correlate with better prognosis in CRC [43]. In addition, Lin et al. found that there are different subsets of Tregs in CRC, named activated Tregs (Foxp3hiCD45RA−), nonsuppressive Tregs (Foxp3loCD45RA−), and resting Tregs (Foxp3loCD45RA+); only activated Tregs correlated with tumor metastases [44]. Nonetheless, these studies are not conclusive, and more studies are needed in the future.

TDLNs play an important role in inhibiting the spread of cancer [20, 21] and have been found to be of pivotal importance in immunotherapy in recent years [45]. Tregs were found to be clustered in TDLNs in previous studies in cancer patients [46, 47]. Theoretically, these cells should be positively correlated with those in tumors in terms of quantity and function [48, 49], such as neutrophils [50]. In fact, some reports in gastric cancer supported this notion; for example, Maruyama et al. found that Tregs were significantly increased in mTDLNs [22], and Lee et al. reported that high Treg density in sentinel lymph nodes (SLNs) was significantly associated with the metastasis of non-SLNs [21]. Interestingly, they also reported that the accumulation of Tregs in N1 was significantly higher than that in N2 and Nc (nonregional control nodes) [21]. Additionally, Kawaida et al. found that Tregs in N1 cases were significantly higher than those in control mesenteric lymph nodes or N2 cases [23]. All these results indicated that Tregs in TDLNs may promote cancer development, which is in line with intratumoral Tregs in prognosis to some extent. Of note, the aforementioned two studies in CRC [26, 28] concerning Tregs in TDLNs indicated a similar role of these cells in gastric cancer [21, 22], which is discordant with reports that support Tregs as a good indicator for survival in tumors in CRC [10,11,12,13]. We speculate that these reports may be biased by the limited sample size [26, 28], and in fact, there was also a report demonstrating that low Tregs in tumor-free SLNs are associated with node metastases in CRC [51]. In our study, we found a positive correlation of Tregs in TDLNs and in tumors, which is in line with the report in neutrophils (R = 0.28, P < 0.01) [50]. Additionally, consistent with Lee et al.’s report [19], we also found that Tregs were significantly enriched in N1 compared to N0 and N2 (P = 0.01 and P = 0.07, respectively, data not shown). More importantly, we detected that these cells presented a parallel prognostic trend to the intratumoral ones, which supports our aforementioned speculation that these cells may functionally correlate in CRC irrespective of their different localization.

It was reported that Tregs could not only release inflammatory cytokines such as interleukin (IL)-10, IL-35, tumor necrosis factor-α (TNF-α), and interferon-γ (INF-γ) [44, 52] but also suppress other cytokine-producing cells such as myeloid dendritic cells [53]. Some of these cytokines would have a profound effect on the control of the development of CRC. For example, it was found that high concentration of IL-35 in CRC could inhibit the cancer cell migration, invasion and proliferation, more importantly, suppress the cancer stem cells [54]; also, the concentration of TNF-α was found to be negatively correlated with the stage in CRC [55]. In addition, it was long-time established that TNF-α and INF-γ have a strong effect in inhibiting colon cancer cell proliferation [56], and a combination of these cytokines could resulted in 30–40% more growth inhibition in CRC cell lines [57]. All these evidence would to some extent support that a high accumulation of Tregs in tumor would have a positive role in prognosis. Except these, the NLR, LMR, and PNI were newly established prognostic indicators in recent years and could be manipulated by inflammatory cytokines [58, 59] released by cancer cells or other cells, such as Tregs. In our study, we found a significant correlation of intratumoral Tregs with LMR, and the latter was also positively associated with prognosis in CRC [60]. Tregs themselves are an important constituent of lymphocytes (which are significantly increased in CRC [17]), and their secretion of IL-10 could manipulate monocytes [59], which could then influence the LMR. However, the exact mechanisms of the relationship of these cells with LMR but no other markers are still poorly understood when taking into consideration the complex cellular/molecular network orchestrated by these cells in tumors or in TDLNs [52].

Notably, we found that 17.33% (26/150), 12% (18/150) cases were blank for Tregs in tumor and TDLNs, respectively. In fact, absence of Tregs in tumor has been registered in previous reports [61,62,63]. As a previous study indicated that hypoxic tumor microenvironment (TME) could result in a significant metabolic reprogramming for Tregs (which are especially susceptible to hypoxic metabolic signaling) and subsequently lead to an abnormal survival and proliferation of these cells [64]. Except this, it was found that certain inflammatory cytokines like IL-6, which was significantly higher in TME and peripheral blood of CRC patients [65, 66], could impact the migration capacity of Tregs to the lesion [67]. Take into account these studies, we speculate that the cases with blank Tregs would have a special TME or significant high IL-6 that could prevent the infiltrating or kill Tregs; however, more studies are still needed to validate this speculation.

Our study has some limitations: first, its retrospective nature cannot completely exclude confounding factors; second, patients with T3 or higher disease with other risk factors or those with mTDLNs would receive subsequent therapies after surgery, which would affect not only survival but also Tregs [68]; third, the information of some important genetic alternations was not available, in particular the RAS mutations, as a report indicated that such changes could affect TNF-induced apoptosis in CRC [69], which could intervention of the role of Tregs in CRC; fourth, although there were no differences in survival for Tregs in MMR-proficient or MMR-deficient patients [40, 70], these results were not validated in patients with mTDLNs or mfTDLNs, and large-sample studies with definite MMR status could resolve these questions in the future.

Conclusion

Overall, our study indicated that higher intratumoral Tregs were associated with better survival in CRC, and a positive correlation of these cells in TDLNs could be found with intratumor Tregs. The similar prognostic prediction trend for these cells in TDLNs as intratumor Tregs suggested that these cells may share a parallelized function in patients; however, more studies are still needed in the future.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its Additional file 1.

Abbreviations

CRC:

Colorectal cancer

Tregs:

T regulatory cells

Foxp3:

Forkhead box transcription Factor 3

TDLNs:

Tumor draining lymph nodes

PFS:

Progression free survival

OS:

Overall survival

NLR:

Neutrophil to lymphocyte ratio

LMR:

Lymphocyte to monocyte ratio

PNI:

Prognostic nutritional index

BMI:

Body mass index

TS:

Tumor size

HPF:

High power field

K-M:

Kaplan–Meier analysis

CEA:

Carcinoembryonic antigen

MMR:

Mismatch-repair

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.

    Article  Google Scholar 

  2. Vuik FE, Nieuwenburg SA, Bardou M, Lansdorp-Vogelaar I, Dinis-Ribeiro M, Bento MJ, et al. Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years. Gut. 2019;68:1820–6.

    PubMed  Google Scholar 

  3. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–61.

    CAS  PubMed  Google Scholar 

  4. Beyer M, Schultze JL. Regulatory T cells in cancer. Blood. 2006;108:804–11.

    CAS  PubMed  Google Scholar 

  5. Wang B, Xu D, Yu X, Ding T, Rao H, Zhan Y, et al. Association of intra-tumoral infiltrating macrophages and regulatory T cells is an independent prognostic factor in gastric cancer after radical resection. Ann Surg Oncol. 2011;18:2585–93.

    PubMed  Google Scholar 

  6. Seifert AM, Eymer A, Heiduk M, Wehner R, Tunger A, von Renesse J, et al. PD-1 expression by lymph node and intratumoral regulatory T cells is associated with lymph node metastasis in pancreatic cancer. Cancers. 2020;24(12):2756.

    Google Scholar 

  7. Zhang L, Wang XI, Ding J, Sun Q, Zhang S. The predictive and prognostic value of Foxp3+/CD25+ regulatory T cells and PD-L1 expression in triple negative breast cancer. Ann Diagn Pathol. 2019;40:143–51.

    PubMed  Google Scholar 

  8. Wolf D, Wolf AM, Rumpold H, Fiegl H, Zeimet AG, Muller-Holzner E, et al. The expression of the regulatory T cell-specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer. Clin Cancer Res. 2005;11:8326–31.

    CAS  PubMed  Google Scholar 

  9. Tao H, Mimura Y, Aoe K, Kobayashi S, Yamamoto H, Matsuda E, et al. Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells. Lung Cancer. 2012;75:95–101.

    PubMed  Google Scholar 

  10. Hu G, Li Z, Wang S. Tumor-infiltrating FoxP3(+) Tregs predict favorable outcome in colorectal cancer patients: a meta-analysis. Oncotarget. 2017;8:75361–71.

    PubMed  PubMed Central  Google Scholar 

  11. Kuwahara T, Hazama S, Suzuki N, Yoshida S, Tomochika S, Nakagami Y, 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;121:659–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Sun X, Feng Z, Wang Y, Qu Y, Gai Y. Expression of Foxp3 and its prognostic significance in colorectal cancer. Int J Immunopathol Pharmacol. 2017;30:201–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Vlad C, Kubelac P, Fetica B, Vlad D, Irimie A, Achimas-Cadariu P. The prognostic value of FOXP3+ T regulatory cells in colorectal cancer. J Buon. 2015;20:114–9.

    PubMed  Google Scholar 

  14. Katz SC, Bamboat ZM, Maker AV, Shia J, Pillarisetty VG, Yopp AC, et al. Regulatory T cell infiltration predicts outcome following resection of colorectal cancer liver metastases. Ann Surg Oncol. 2013;20:946–55.

    PubMed  Google Scholar 

  15. Suzuki H, Chikazawa N, Tasaka T, Wada J, Yamasaki A, Kitaura Y, et al. Intratumoral CD8(+) T/FOXP3 (+) cell ratio is a predictive marker for survival in patients with colorectal cancer. Cancer Immunol Immunother. 2010;59:653–61.

    CAS  PubMed  Google Scholar 

  16. Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, Sargent DJ. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology. 2009;137:1270–9.

    CAS  PubMed  Google Scholar 

  17. Ling KL, Pratap SE, Bates GJ, Singh B, Mortensen NJ, George BD, et al. Increased frequency of regulatory T cells in peripheral blood and tumour infiltrating lymphocytes in colorectal cancer patients. Cancer Immun. 2007;7:7.

    PubMed  PubMed Central  Google Scholar 

  18. Shimada Y, Hamaguchi T, Mizusawa J, Saito N, Kanemitsu Y, Takiguchi N, et al. Randomised phase III trial of adjuvant chemotherapy with oral uracil and tegafur plus leucovorin versus intravenous fluorouracil and levofolinate in patients with stage III colorectal cancer who have undergone Japanese D2/D3 lymph node dissection: final results of JCOG0205. Eur J Cancer. 2014;50:2231–40.

    CAS  PubMed  Google Scholar 

  19. Søndenaa K, Quirke P, Hohenberger W, Sugihara K, Kobayashi H, Kessler H, et al. The rationale behind complete mesocolic excision (CME) and a central vascular ligation for colon cancer in open and laparoscopic surgery: proceedings of a consensus conference. Int J Colorectal Dis. 2014;29:419–28.

    PubMed  Google Scholar 

  20. Takeuchi H, Kitajima M, Kitagawa Y. Sentinel lymph node as a target of molecular diagnosis of lymphatic micrometastasis and local immunoresponse to malignant cells. Cancer Sci. 2008;99:441–50.

    CAS  PubMed  Google Scholar 

  21. Lee HE, Park DJ, Kim WH, Kim HH, Lee HS. High FOXP3+ regulatory T-cell density in the sentinel lymph node is associated with downstream non-sentinel lymph-node metastasis in gastric cancer. Br J Cancer. 2011;105:413–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Maruyama T, Kono K, Mizukami Y, Kawaguchi Y, Mimura K, Watanabe M, et al. Distribution of Th17 cells and FoxP3(+) regulatory T cells in tumor-infiltrating lymphocytes, tumor-draining lymph nodes and peripheral blood lymphocytes in patients with gastric cancer. Cancer Sci. 2010;101:1947–54.

    CAS  PubMed  Google Scholar 

  23. Kawaida H, Kono K, Takahashi A, Sugai H, Mimura K, Miyagawa N, et al. Distribution of CD4+CD25high regulatory T-cells in tumor-draining lymph nodes in patients with gastric cancer. J Surg Res. 2005;124:151–7.

    CAS  PubMed  Google Scholar 

  24. Ariafar A, Vahidi Y, Fakhimi M, Asadollahpour A, Erfani N, Faghih Z. Prognostic significance of CD4-positive regulatory T cells in tumor draining lymph nodes from patients with bladder cancer. Heliyon. 2020;6: e05556.

    PubMed  PubMed Central  Google Scholar 

  25. Faghih Z, Erfani N, Haghshenas MR, Safaei A, Talei AR, Ghaderi A. Immune profiles of CD4+ lymphocyte subsets in breast cancer tumor draining lymph nodes. Immunol Lett. 2014;158:57–65.

    CAS  PubMed  Google Scholar 

  26. Gai XD, Li C, Song Y, Lei YM, Yang BX. In situ analysis of FOXP3(+) regulatory T cells and myeloid dendritic cells in human colorectal cancer tissue and tumor-draining lymph node. Biomed Rep. 2013;1:207–12.

    CAS  PubMed  Google Scholar 

  27. Kazama K, Otake J, Satoyoshi T, Shiozawa M, Sugano N, Sato S, et al. Distribution of regulatory T-cells and other phenotypes of T-cells in tumors and regional lymph nodes of colorectal cancer patients. In Vivo. 2020;34:849–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Deng L, Zhang H, Luan Y, Zhang J, Xing Q, Dong S, et al. Accumulation of foxp3+ T regulatory cells in draining lymph nodes correlates with disease progression and immune suppression in colorectal cancer patients. Clin Cancer Res. 2010;16:4105–12.

    CAS  PubMed  Google Scholar 

  29. Xu RYJ, Li F, Yan B. Postoperative fasting blood glucose predicts prognosis in stage I-III colorectal cancer patients undergoing resection. Gastroenterol Res Pract. 2020;2020:2482409.

    PubMed  PubMed Central  Google Scholar 

  30. Li X, An B, Ma J, He B, Qi J, Wang W, et al. Prognostic value of the tumor size in resectable colorectal cancer with different primary locations: a retrospective study with the propensity score matching. J Cancer. 2019;10:313–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang YC, Liu Y, Qiu XM, Yan B. Concurrent comparison of the prognostic values of tumor budding, tumor stroma ratio, tumor infiltrating pattern and lymphocyte-to-monocyte ratio in colorectal cancer patients. Technol Cancer Res Treat. 2021;20:15330338211045826.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Yoshida N, Kinugasa T, Miyoshi H, Sato K, Yuge K, Ohchi T, et al. A high RORγT/CD3 ratio is a strong prognostic factor for postoperative survival in advanced colorectal cancer: analysis of helper T cell lymphocytes (Th1, Th2, Th17 and regulatory T cells). Ann Surg Oncol. 2016;23:919–27.

    PubMed  Google Scholar 

  33. Inamoto S, Kawada K, Okamura R, Hida K, Sakai Y. Prognostic impact of the combination of neutrophil-to-lymphocyte ratio and Glasgow prognostic score in colorectal cancer: a retrospective cohort study. Int J Colorectal Dis. 2019;34:1303–15.

    PubMed  Google Scholar 

  34. Stotz M, Pichler M, Absenger G, Szkandera J, Arminger F, Schaberl-Moser R, et al. The preoperative lymphocyte to monocyte ratio predicts clinical outcome in patients with stage III colon cancer. Br J Cancer. 2014;110:435–40.

    CAS  PubMed  Google Scholar 

  35. Maruyama T, Shimoda M, Hakoda H, Sako A, Ueda K, Suzuki S. Preoperative prognostic nutritional index predicts risk of recurrence after curative resection for stage IIA colon cancer. Am J Surg. 2021;222:179–85.

    PubMed  Google Scholar 

  36. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.

    CAS  Google Scholar 

  37. Perrone G, Ruffini PA, Catalano V, Spino C, Santini D, Muretto P, et al. Intratumoural FOXP3-positive regulatory T cells are associated with adverse prognosis in radically resected gastric cancer. Eur J Cancer. 2009;44:1875–82.

    Google Scholar 

  38. Solis-Castillo LA, Garcia-Romo GS, Diaz-Rodriguez A, Reyes-Hernandez D, Tellez-Rivera E, Rosales-Garcia VH, et al. Tumor-infiltrating regulatory T cells, CD8/Treg ratio, and cancer stem cells are correlated with lymph node metastasis in patients with early breast cancer. Breast Cancer. 2020;27:837–49.

    PubMed  Google Scholar 

  39. Barnett JC, Bean SM, Whitaker RS, Kondoh E, Baba T, Fujii S, et al. Ovarian cancer tumor infiltrating T-regulatory (T(reg)) cells are associated with a metastatic phenotype. Gynecol Oncol. 2010;116:556–62.

    CAS  PubMed  Google Scholar 

  40. Frey DM, Droeser RA, Viehl CT, Zlobec I, Lugli A, Zingg U, et al. High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int J Cancer. 2010;126:2635–43.

    CAS  PubMed  Google Scholar 

  41. Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 2009;27:186–92.

    PubMed  Google Scholar 

  42. Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother. 2011;60:909–18.

    CAS  PubMed  Google Scholar 

  43. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22:679–84.

    CAS  PubMed  Google Scholar 

  44. Lin YC, Mahalingam J, Chiang JM, Su PJ, Chu YY, Lai HY, et al. Activated but not resting regulatory T cells accumulated in tumor microenvironment and correlated with tumor progression in patients with colorectal cancer. Int J Cancer. 2013;132:1341–50.

    CAS  PubMed  Google Scholar 

  45. Dammeijer F, van Gulijk M, Mulder EE, Lukkes M, Klaase L, van den Bosch T, et al. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell. 2020;38:685-700.e8.

    CAS  PubMed  Google Scholar 

  46. Ghods A, Mehdipour F, Shariat M, Talei AR, Ghaderi A. Regulatory T cells express Tumor Necrosis Factor Receptor 2 with the highest intensity among CD4(+) T cells in the draining lymph nodes of breast cancer. Mol Immunol. 2021;137:52–6.

    CAS  PubMed  Google Scholar 

  47. Nakamura R, Sakakibara M, Nagashima T, Sangai T, Arai M, Fujimori T, et al. Accumulation of regulatory T cells in sentinel lymph nodes is a prognostic predictor in patients with node-negative breast cancer. Eur J Cancer. 2009;45:2123–31.

    CAS  PubMed  Google Scholar 

  48. Knutson KL, Disis ML, Salazar LG. CD4 regulatory T cells in human cancer pathogenesis. Cancer Immunol Immunother. 2007;56:271–85.

    PubMed  Google Scholar 

  49. Yaqub S, Henjum K, Mahic M, Jahnsen FL, Aandahl EM, Bjørnbeth BA, et al. Regulatory T cells in colorectal cancer patients suppress anti-tumor immune activity in a COX-2 dependent manner. Cancer Immunol Immunother. 2008;57:813–21.

    CAS  PubMed  Google Scholar 

  50. Hiramatsu S, Tanaka H, Nishimura J, Sakimura C, Tamura T, Toyokawa T, et al. Neutrophils in primary gastric tumors are correlated with neutrophil infiltration in tumor-draining lymph nodes and the systemic inflammatory response. BMC Immunol. 2018;19:13.

    PubMed  PubMed Central  Google Scholar 

  51. Matera L, Sandrucci S, Mussa A, Boffa C, Castellano I, Cassoni P. Low Foxp3 expression in negative sentinel lymph nodes is associated with node metastases in colorectal cancer. Gut. 2010;59:419–20.

    PubMed  Google Scholar 

  52. Saleh R, Elkord E. FoxP3(+) T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett. 2020;490:174–85.

    CAS  PubMed  Google Scholar 

  53. Nagorsen D, Voigt S, Berg E, Stein H, Thiel E, Loddenkemper C. Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J Transl Med. 2007;5:62.

    PubMed  PubMed Central  Google Scholar 

  54. Jiang Y, Ma Y, Li R, Sun J. Colon cancer-induced interleukin-35 inhibits beta-catenin-mediated pro-oncogenic activity. Oncotarget. 2017;9:11989–98.

    PubMed  PubMed Central  Google Scholar 

  55. Tojek K, Anaszewicz M, Szukay B, Czerniak B, Socha E, Lis K, et al. Circulating leptin, adiponectin, and tumor necrosis factor-alpha in patients undergoing surgery due to colorectal cancer. Digestion. 2021;102(2):246–55.

    CAS  PubMed  Google Scholar 

  56. Schiller JH, Bittner G. Anti-proliferative effects of tumor necrosis factor, gamma interferon and 5-fluorouracil on human colorectal carcinoma cell lines. Int J Cancer. 1990;46:61–6.

    CAS  PubMed  Google Scholar 

  57. Schiller JH, Bittner G, Storer B, Willson JK. Synergistic antitumor effects of tumor necrosis factor and gamma-interferon on human colon carcinoma cell lines. Cancer Res. 1987;47:2809–13.

    CAS  PubMed  Google Scholar 

  58. Ubukata H, Motohashi G, Tabuchi T, Nagata H, Konishi S, Tabuchi T. Evaluations of interferon-γ/interleukin-4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol. 2010;102:742–7.

    PubMed  Google Scholar 

  59. Valdés-Ferrada J, Muñoz-Durango N, Pérez-Sepulveda A, Muñiz S, Coronado-Arrázola I, Acevedo F, Soto JA, et al. Peripheral blood classical monocytes and plasma interleukin 10 are associated to neoadjuvant chemotherapy response in breast cancer patients. Front Immunol. 2020;11:1413.

    PubMed  PubMed Central  Google Scholar 

  60. Tan D, Fu Y, Tong W, Li F. Prognostic significance of lymphocyte to monocyte ratio in colorectal cancer: A meta-analysis. Int J Surg. 2018;55:128–38.

    PubMed  Google Scholar 

  61. Kim KJ, Lee KS, Cho HJ, Kim YH, Yang HK, Kim WH, et al. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Hum Pathol. 2014;45:285–93.

    CAS  PubMed  Google Scholar 

  62. Hasegawa T, Suzuki H, Yamaura T, Muto S, Okabe N, Osugi J, et al. Prognostic value of peripheral and local forkhead box P3+ regulatory T cells in patients with non-small-cell lung cancer. Mol Clin Oncol. 2014;2:685–94.

    PubMed  PubMed Central  Google Scholar 

  63. Perrone G, Ruffini PA, Catalano V, Spino C, Santini D, Muretto P, et al. Intratumoural FOXP3-positive regulatory T cells are associated with adverse prognosis in radically resected gastric cancer. Eur J Cancer. 2008;44:1875–82.

    CAS  PubMed  Google Scholar 

  64. Sasidharan Nair V, Saleh R, Toor SM, Cyprian FS, Elkord E. Metabolic reprogramming of T regulatory cells in the hypoxic tumor microenvironment. Cancer Immunol Immunother. 2021;70:2103–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Kim B, Seo Y, Kwon JH, Shin Y, Kim S, Park SJ, et al. IL-6 and IL-8, secreted by myofibroblasts in the tumor microenvironment, activate HES1 to expand the cancer stem cell population in early colorectal tumor. Mol Carcinog. 2021;60:188–200.

    CAS  PubMed  Google Scholar 

  66. Guthrie GJ, Roxburgh CS, Richards CH, Horgan PG, McMillan DC. Circulating IL-6 concentrations link tumour necrosis and systemic and local inflammatory responses in patients undergoing resection for colorectal cancer. Br J Cancer. 2013;109:131–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. He S, Xue M, Cai G. IL-6 alters migration capacity of CD4+Foxp3+ regulatory T cells in systemic lupus erythematosus. Scand J Immunol. 2021;94: e13099.

    CAS  PubMed  Google Scholar 

  68. Maeda K, Hazama S, Tokuno K, Kan S, Maeda Y, Watanabe Y, et al. Impact of chemotherapy for colorectal cancer on regulatory T-cells and tumor immunity. Anticancer Res. 2011;31:4569–74.

    CAS  PubMed  Google Scholar 

  69. Kreeger PK, Mandhana R, Alford SK, Haigis KM, Lauffenburger DA. RAS mutations affect tumor necrosis factor-induced apoptosis in colon carcinoma cells via ERK-modulatory negative and positive feedback circuits along with non-ERK pathway effects. Cancer Res. 2009;69:8191–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Yoon HH, Orrock JM, Foster NR, Sargent DJ, Smyrk TC, Sinicrope FA. Prognostic impact of FoxP3+ regulatory T cells in relation to CD8+ T lymphocyte density in human colon carcinomas. PLoS ONE. 2012;7: e42274.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

FL and WW were responsible for the conception of the work. BY, JX and QY obtained the data. BY, TX, JX, and MX analyzed the data. BY wrote the manuscript. TX, JX and MX critically revised the manuscript. All authors are accountable for the contents of this work. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Fang Li or Wei Wen.

Ethics declarations

Ethics approval and consent to participate

The study was conducted in accordance with the principles stated in the Declaration of Helsinki and was approved by the ethics committee of Hainan Hospital of Chinese PLA General Hospital (ID: 301HLFYLS15). Written informed consent was obtained from the patients or their authorized relatives.

Consent for publication

Not applicable.

Competing interests

The authors have no conflicts of interest to declare.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Dataset to generate the results of the study.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yan, B., Xiong, J., Ye, Q. et al. Correlation and prognostic implications of intratumor and tumor draining lymph node Foxp3+ T regulatory cells in colorectal cancer. BMC Gastroenterol 22, 122 (2022). https://0-doi-org.brum.beds.ac.uk/10.1186/s12876-022-02205-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12876-022-02205-0

Keywords