Skip to main content

Associations of different immune checkpoints-expressing CD4+ Treg/ T cell subsets with disease-free survival in colorectal cancer patients

Abstract

There are different subsets of T regulatory cells (Tregs), orchestrating critical roles in the regulation of anti-tumor immunity in colorectal cancer (CRC). In this study, we report that a high frequency of circulating CD4+FoxP3+ Tregs was associated with poorer disease-free survival (DFS), while their higher frequencies in tumor-infiltrating CD4+ Tregs was associated with better DFS. We further investigated such associations with four Tregs/T cells expressing or lacking FoxP3 and Helios (FoxP3±Helios±). For the first time, we report that a high frequency of circulating CD4+FoxP3+Helios+ Tregs was associated with poorer DFS, while a high frequency of tumor-infiltrating CD4+FoxP3−Helios− T cells was associated with poorer DFS. In the four FoxP3±Helios± T cell subsets expressing any of the immune checkpoints (ICs) investigated, we found that a high frequency of CD4+FoxP3+Helios−PD-1+ Tregs in circulation was associated with worse DFS. We also found that high frequencies of FoxP3+Helios+CTLA-4+ Tregs, FoxP3+Helios−CTLA-4+ Tregs, and FoxP3−Helios+CTLA-4+ CD4+ T cells in circulation were associated with worse DFS. In contrast, high frequencies of CD4+TIM-3+ T cells, FoxP3+Helios+TIM-3+ Tregs, and FoxP3−Helios+TIM-3+ CD4+ T cells in circulation were associated with longer DFS. Our data show that certain CD4+ Treg/T cell subsets could serve as independent predictive biomarkers in CRC patients. Identification of the exact subpopulations contributing to clinical outcomes is critical for prognoses and therapeutic targeting.

Peer Review reports

Introduction

Colorectal cancer (CRC) is the second most deadly malignancy and the third most common cancer in the world [1]. A total of 1.8 million new cases of colon cancer were diagnosed in 2018, accounting for approximately 10% of all new cancer cases and deaths globally [1, 2]. The gastrointestinal tract is susceptible to persistent immune responses, leading to chronic intestinal inflammation, which has a role in the development of cancer, particularly via the secretion of inflammatory cytokines [3]. T regulatory cells (Tregs) are immunosuppressive cells that are found in many different subsets and serve key functions in the maintenance of immunological homeostasis and self-tolerance [4]. They also have critical roles in the regulation of cancer immunity [4]. In different types of cancer, high levels of Treg infiltration into tumors are usually correlated with poor clinical outcomes [5]. However, the role of Tregs is controversial in CRC. Certain studies have shown that tumor-infiltrating FoxP3+ Tregs are associated with a better prognosis in CRC patients [6,7,8,9]. Conversely, Betts et al., found that high levels of Tregs contribute to disease progression in CRC patients [10]. Helios serves as a marker for T cell activation and proliferation and its expression is required for the maintenance of Treg inhibitory function [11]. In CRC, Helios mRNA level was shown to be higher in tumor tissue in advance stages, suggesting their potential effects in CRC progression [12]. Immune checkpoints (ICs) are critical for immune tolerance and for tissue protection during inflammatory responses [13]. Tumors could modulate the expression of certain ICs as a substantial mechanism of immunological resistance [8, 13].

We have recently reported that some inhibitory ICs, including programmed cell death-1 (PD-1), cytotoxic T lymphocyte-associated antigen (CTLA-4), T cell immunoglobulin and mucin domain-containing protein-3 (TIM-3), and lymphocyte-activation gene-3 (LAG-3), are expressed on CD4+ T cells and play roles in CRC progression [8].

This is the first study to insightfully investigate any potential associations between frequencies of different Treg and CD4+ T cell subsets expressing ICs with DFS in CRC patients.

Materials and methods

Patients and samples

This study was carried out in accordance with ethical approvals (protocol no. MRC-02–18-012) from the Medical Research Center, Hamad Medical Corporation, Doha, Qatar. Peripheral blood samples were collected from thirty-four CRC patients with varying disease stages. Thirty-two patients were eligible and included in the DFS analyses reported in this study. Non-malignant normal tissues (NT) and tumor tissues (TT) from colon were obtained from 22 out of these 32 patients, who underwent surgery at Hamad Medical Corporation, Doha, Qatar. All patients were treatment naive prior to surgery, and they gave written informed consent prior to sample collection.

Clinical and pathological characteristics of these patients are shown in Table 1. All DFS data for the 32 patients were collected in January 2022. Four out of the 32 patients had disease progression, in the form of tumor local recurrence or development of new lymph node and/or distant metastasis. Disease progression was assessed by contrast enhanced chest-abdomen-pelvis computed tomography (CT) scan, that was performed for patients on their clinical follow-up.

Table1 Characteristic features of colorectal cancer patients

Multi-parametric flow cytometry

No extra experiments were performed in this study and flow cytometry data of different CD4+ T cell subsets were collected. Immune staining and flow cytometry analyses have been done as per our previously published article [8].

Statistical analyses

Statistical analyses were performed using GraphPad Prism 9 software (GraphPad Software, California, USA). The Shapiro–Wilk test was used to analyze normality of datasets. All immune cell subsets were categorized into low and high groups as below/above mean for normally distributed data, and below/above median for non-normally distributed data. DFS was estimated using the Kaplan–Meier curves, and log-rank test was used to calculate P values. P value of ≤ 0.05 was considered to be significant.

Results

Association of Tregs with DFS

Tregs are a significant subgroup of CD4+ T cells that are characterized by high levels of the interleukin-2 receptor alpha chain (CD25) and the transcription factor forkhead box P3 (FoxP3) [14]. Helios is a transcription factor that modulates FoxP3+ Treg functional stability and is needed for their inhibitory action [11, 15,16,17]. We have previously measured levels of different CD4+ T cell subsets in PBMCs, NILs, and TILs of CRC patients [8]. Representative flow cytometric plots and percentages of the different CD4+ Treg subsets have already been shown in our previous study [8]. In this study, we further identified the specific Treg subsets contributing to the worse DFS. We found that high frequencies of CD4+CD25+ T cells were not associated with DFS in circulation or in tissues in this cohort of CRC patients (Fig. 1A). When Tregs were defined based on FoxP3 expression as a more Treg-specific marker, a high Treg frequency was significantly associated with shorter DFS in circulation. Interestingly, a high Treg frequency in TILs, but not in NILs as controls, was associated with longer DFS (Fig. 1B). CD4+Helios+ T cells showed similar findings like CD4+FoxP3+ Tregs. More specifically, high frequencies of CD4+Helios+ T cells showed a significant association with shorter DFS in circulation (Fig. 1C). There were no associations between frequencies of these cell subsets in normal tissues, as controls, with DFS (Fig. 1A-C).

Fig. 1
figure 1

Kaplan–Meier curves of DFS based on frequencies of CD25+, FoxP3+, and Helios+ in PBMCs, TILs, and NILs. Patients with high frequencies of CD25+ (A), FoxP3+ (B), and Helios+ (C), in CD3+CD4+ T cells, were compared with those with low frequencies of these subsets. Patient numbers investigated are indicated on the survival curves for each cell population in all figures

Association of FoxP3±Helios± T cell subsets with DFS

We further identified the specific Treg subsets contributing to the worse DFS. We report for the first time, that patients with high frequency of CD4+FoxP3+Helios+ Treg subset in circulation had significantly shorter DFS than patients with low frequency of this Treg subset (Fig. 2A). Additionally, there were no associations between frequencies of both CD4+FoxP3+Helios− Treg subset and CD4+FoxP3−Helios+ T cells in PBMCs, TILs, and NILs with DFS (Fig. 2B, C). High frequencies of CD4+FoxP3−Helios− T cells showed a significant association with worse DFS in TILs, while there was no association between frequencies of this T cell subset with DFS in circulation (Fig. 2D). Interestingly, we found that CD4+FoxP3+Helios+ Treg subset and CD4+FoxP3−Helios− T cell subset in the tumor microenvironment (TME) contributed differently to the DFS in CRC patients (Fig. 2A, D).

Fig. 2
figure 2

Kaplan–Meier curves of DFS based on frequencies of FoxP3±Helios± in PBMCs, TILs, and NILs. Patients with high frequencies of FoxP3+Helios+ (A), FoxP3+Helios− (B), FoxP3−Helios+ (C), and FoxP3−Helios− (D), in CD3+CD4+ T cells, were compared with those with low frequencies of these subsets

Association of immune checkpoints with DFS

During T-cell activation, the immune system uses different checkpoint pathways to maintain co-inhibitory and co-stimulatory signals [8, 18]. Immune checkpoints play critical roles in the inhibition of anti-tumor immune responses in a variety of malignancies, including colorectal cancer [18, 19]. We did not find any association between DFS and frequencies of CD4+PD-1+ (Fig. 3A) and CD4+CTLA-4+ T cells (Fig. 3B) in PBMCs, TILs, and NILs. Interestingly, patients with higher frequencies of CD4+TIM-3+ T cells in circulation showed significantly improved DFS (Fig. 3C). Otherwise, there were no associations between frequencies of CD4+TIM-3+ T cells with DFS in tumor and normal tissues (Fig. 3C). Additionally, there were no associations between frequencies of CD4+LAG-3+ T cells in PBMCs and TILs with DFS (Fig. 3D). We did not see any associations between frequencies of these subsets in normal tissues, as controls, with DFS. Overall, CD4+TIM-3+ T cells seem to be the most critical IC-expressing CD4+ T cell subset contributing to DFS in CRC patients.

Fig. 3
figure 3

Kaplan–Meier curves of DFS based on frequencies of different immune checkpoints in PBMCs, TILs, and NILs. Patients with high frequencies of PD-1+(A), CTLA-4+ (B), TIM-3+(C), and LAG-3+ (D), in CD3+CD4+ T cells, were compared with those with low frequencies of these subsets

Association of different FoxP3±Helios± T cell subsets and ICs with DFS

We then went further and identified the specific IC-expressing Treg and T cell subsets, based on the frequency of FoxP3 and Helios (FoxP3±Helios±), which contribute to worse DFS. We did not find any association between frequencies of CD4+FoxP3+Helios+PD-1+ Tregs in PBMCs, TILs, and NILs with DFS (Fig. 4A). We report, for the first time, that CRC patients with higher frequencies of CD4+FoxP3+Helios+CTLA-4+ Treg subset in circulation, but not in TILs or NILs, had significantly shorter DFS than patients with lower frequencies of this Treg subset (Fig. 4B). Conversely, high frequencies of CD4+FoxP3+Helios+TIM-3+ Treg subset in circulation, but not in TILs or NILs, were associated with better DFS (Fig. 4C). Moreover, we found that CRC patients with high frequencies of CD4+FoxP3+Helios−PD-1+ and CD4+FoxP3+Helios−CTLA-4+ Treg subsets in PBMCs, but not in TILs or NILs, had significantly shorter DFS than patients with lower frequencies of these Treg subsets (Fig. 5A, B). Moreover, there were no associations between frequencies of CD4+FoxP3+Helios−TIM-3+ Treg subset with DFS in PBMCs, NILs and TILs (Fig. 5C).

Fig. 4
figure 4

Kaplan–Meier curves of DFS based on frequencies of CD4+FoxP3+Helios+ cells expressing immune checkpoints in PBMCs, TILs, and NILs. Patients with high frequencies of PD-1+ (A), CTLA-4+ (B), and TIM-3+ (C), in CD3+CD4+FoxP3+Helios+ T cells, were compared with those with low frequencies of these subsets

Fig. 5
figure 5

Kaplan–Meier curves of DFS based on frequencies of CD4+FoxP3+Helios− expressing immune checkpoints in PBMCs, TILs, and NILs. Patients with high frequencies of PD-1+ (A), CTLA-4+ (B), and TIM-3+ (C), in CD3+CD4+FoxP3+Helios− T cells, were compared with those with low frequencies of these subsets

Furthermore, we investigated DFS associations with frequencies of IC-expressing CD4+FoxP3− T cells (with or without Helios expression) (Figs. 6 and 7). We did not find any association between frequencies of CD4+FoxP3−Helios+PD-1+ Tregs in PBMCs, TILs, and NILs with DFS (Fig. 6A). Indeed, patients with high frequencies of CD4+FoxP3−Helios+CTLA-4+ T cell in circulation, but not in TILs or NILs, had significantly shorter DFS (Fig. 6B). Moreover, a high frequency of CD4+FoxP3−Helios+TIM-3+ T cells in circulation showed a significant association with longer DFS (Fig. 6C). However, there were no associations between frequencies of this T cell subset with DFS in TILs and NILs (Fig. 6C). Of note, there were no associations between frequencies of CD4+FoxP3−Helios− T cells expressing PD1, or CTLA-4, or TIM-3 with DFS in PBMCs, TILs, and NILs (Fig. 7). Table 2 summarizes associations of high frequencies of any CD4+ Treg/T cell subsets with DFS.

Fig. 6
figure 6

Kaplan–Meier curves of DFS based on frequencies of CD4+FoxP3−Helios+ expressing immune checkpoints in PBMCs, TILs, and NILs. Patients with high frequencies of PD-1+ (A), CTLA-4+ (B), and TIM-3+ (C), in CD3+CD4+FoxP3−Helios+ T cells, were compared with those with low frequencies of these subsets

Fig. 7
figure 7

Kaplan–Meier curves of DFS based on frequencies of CD4+FoxP3−Helios− expressing immune checkpoints in PBMCs, TILs, and NILs. Patients with high frequencies of PD-1+ (A), CTLA-4+ (B), and TIM-3+ (C) in CD3+CD4+FoxP3−Helios− T cells, were compared with those with low frequencies of these cell subsets

Table 2 Summary of the significant associations of high frequencies of different CD4+ T cell subsets with DFS

Discussion

Immune cells have the ability to recognize and destroy cancer cells. CD4+ T cells may target cancer cells by modulating the tumor microenvironment [20, 21]. CD4+ T lymphocytes are predominantly T regulatory cells in CRC tissues, and they express several IC molecules such as PD-1, CTLA-4, TIM-3, and LAG-3 [8]. High levels of Treg-related markers were observed in the TME in CRC patients, suggesting their potential effects in carcinogenesis [8, 22, 23]. In contrast to other solid tumors, high levels of tumor-infiltrating FoxP3+ Tregs were related with increased survival in CRC patients [9, 24]. Recent studies reported that tumor-infiltrating CD39+ Tregs in CRC patients expressed different markers such as OX-40, CTLA-4 and ICOS, implicating their high immunosuppressive abilities in inhibiting anti-tumor immune responses [25, 26]. Correale et al., showed that a higher level of FoxP3+ T-lymphocyte tumor infiltration in CRC patients receiving chemotherapy or chemo-immunotherapy was a favorable prognostic marker [27]. Moreover, a high frequency of FoxP3+ Tregs within tumor lead to a promising outcome in CRC, suggesting that FoxP3+ Tregs are one of the most useful indicators for predicting the prognosis of CRC [7, 28,29,30]. Another study found that CD8+:FoxP3+ cell ratios were significantly correlated with distant-recurrence-free survival (DRFS) in the CRC TME [31]. Also, they reported that high numbers of FoxP3+ cells were associated with longer overall survival (OS) and DFS, although non-significantly [31]. Interestingly, FoxP3+ T cells only have a positive effect on survival in colon tumors that have low levels of CD8+ T-cell infiltration [32]. Another study found that TGF-β, which is produced by tumors, has been linked to an increase in the number of intratumoral FoxP3+ Tregs [33]. Moreover, they found that intratumoral CD8+ T cell:FoxP3+ Treg ratio positively correlated with longer DFS and OS [33]. Nevertheless, functionally different subgroups of tumor-infiltrating FoxP3+ Tregs contribute in opposing ways to determining CRC disease prognosis [6].

Importantly, it has been demonstrated that circulating Tregs are effective in suppressing antitumor immunity, leading to an adverse outcome of CRC patients [10, 34]. We found that higher frequencies of CD4+FoxP3+ Tregs in circulation were associated with shorter DFS, implicating the harmful effect of these suppressive cells in inhibiting anti-tumor immune responses in circulation. However, high frequencies of this Treg subset in the TME were associated with longer DFS, indicating the beneficial anti-inflammatory role of CD4+FoxP3+ Tregs in the TME of CRC patients.

Expression of FoxP3 was highly and positively correlated with the expression of Helios on T cells within PBMCs and TILs in cancer patients [17]. In tumor tissues, the majority of Tregs co-expressed both FoxP3 and Helios, suggesting higher immunosuppressive potentials than cells with single expressions of FoxP3 or Helios [17]. Classification of FoxP3+ Tregs into subsets helps to investigate Treg cell differentiation in both normal and disease conditions, as well as to alter immune responses by modulating specific FoxP3+ Treg subpopulations [35]. Our group has also recently proposed that FoxP3+Helios+ Tregs constitute a functional subset of Tregs with higher suppressive characteristics [36]. Tumor tissues in CRC patients were characterized by high levels of Helios+ Tregs compared to PBMCs and normal colon tissues [24, 37], suggesting their potential roles in CRC progression [12]. In this study, we found that a high frequency of CD4+FoxP3+Helios+ Tregs in blood was associated with shorter DFS, suggesting the potential role of this highly immunosuppressive Treg subset in inhibiting anti-tumor immune responses, and consequently worsening clinical outcomes. Importantly, it is hypothesized that the TME could enhance the induction of the FoxP3+Helios+ Treg subset from the FoxP3−Helios+ T cell subset [17]. Interestingly, we found that high frequencies of CD4+FoxP3−Helios− TILs were significantly associated with worse DFS, suggesting that these cells could induce inflammation in CRC TME.

In TME, certain tumor ligands bind to inhibitory molecules on T cells, such as CTLA-4, PD-1, TIM-3, and LAG-3 and others, which in turn produce immune-suppressive mediators, leading to the failure of cancer elimination [18, 38]. CD4+PD-1+ T cells were predominantly detectable in tumor tissues of CRC patients [39], which may lead to T-cell exhaustion and cancer progression [40, 41]. Additionally, CRC patients with high expression of PD-1 had worse TNM staging and DFS, compared with those with low expression [42]. In agreement with these studies, we found that high frequencies of FoxP3+Helios−PD-1+ were associated with shorter DFS in circulation. More samples might be required to determine possible associations of these subsets with DFS in tumor tissues.

TIM-3 is frequently overexpressed on exhausted CD4+ T cells in CRC patients, suggesting this could be associated with worse prognoses [43,44,45]. Moreover, TIM-3 was correlated with CRC progression and might be a possible therapeutic target [46]. Arai et al., found that TIM-3 expression on CD4+ T cells was significantly increased after CRC operation [47]. Additionally, they found that the production of IFN-γ was linked to TIM-3 and PD-1 expression on CD4+ and CD8+ T cells, suggesting that TIM-3+PD-1+CD4+ and CD8+ T cells are highly dysfunctional [47]. On the other hand, Zhang et al., found that TIM-3 expression either in the primary or metastatic tumor was associated with better progression-free survival (PFS) in renal cell carcinoma [48]. We found that high frequencies of TIM-3+ and FoxP3+Helios+TIM-3+ CD4+ T cells were associated with longer DFS in circulation. Of note, most studies investigated TIM-3 expression in bulk tumor tissues but not on specific T cell subsets. This is the first study to indicate that TIM-3 expression on T cells is associated with better DFS in CRC.

We have previously reported that mRNA level of CTLA-4 in tumor tissues was increased in advanced stages of CRC, suggesting their possible effects in CRC progression [12]. Moreover, we have shown that there was a significant elevation in levels of CD4+CTLA-4+ T cells only in PBMCs of CRC patients with advanced stages, suggesting that there is a relationship between increased levels of CTLA-4+ Tregs and CRC progression [8]. In this study, high frequencies of CTLA-4 expressed in different Treg subsets were associated with worse DFS in circulation. Therefore, targeting CTLA-4 on these subsets might have beneficial roles in CRC.

A recent study showed that overexpression of LAG-3 on tumor tissues was associated with worse prognosis in patients with microsatellite instability high (MSI-H) colon cancer [49]. In addition, we have previously reported that mRNA level of LAG-3 was higher in PBMCs of CRC patients than those of healthy controls [12]. Moreover, the frequency of LAG-3 in tumor tissues was associated with differentiation, lymph node metastasis, and invasion in CRC patients [50]. In our study, we found that there were no associations between frequencies of CD4+LAG-3+ T cells with DFS. Due to weak overall LAG-3 expression in original study [8], more samples are needed to investigate the role of LAG-3 in DFS in CRC patients.

CRC patients with mismatch repair deficiency (dMMR) usually have worse prognosis than patients without dMMR [51]. Notably, over-expressions of FoxP3, IL1β, IL17, TGF-β and IL6 were associated with the microsatellite stability MSS phenotype [52, 53]. It has been shown that a high frequency of tumor-infiltrating FoxP3+ Tregs predicts improved survival in mismatch repair-proficient CRC patients [53]. Moreover, in stage II MSS CRC, it has been found that low frequencies of both FoxP3+ and CD3+ TILs were associated with the highest progression risk [54]. In this study, we were not able to investigate such association because only 4 CRC patients had dMMR (12.5% of this study cohort, as shown in Table 1); the percentage of dMMR patients was low as expected in CRC patients [55].

Our study highlights the potential of some CD4+ T cell subsets as predictive biomarkers associated with worse DFS in CRC patients. Overall, high frequency of tumor-infiltrating FoxP3−Helios− T cells, and high frequencies of circulating FoxP3+, Helios+, FoxP3+Helios+, FoxP3+Helios−PD-1+, FoxP3+Helios+CTLA-4+, FoxP3+Helios−CTLA-4+ Tregs, and FoxP3−Helios+CTLA-4+ T cells, are associated with shorter DFS. Targeting these immune cell subsets in CRC patients could improve clinical outcomes. On the other hand, high frequencies of CD4+TIM-3+, FoxP3+Helios+TIM-3+ Tregs and FoxP3−Helios+TIM-3+ T cells in circulation are associated with longer DFS in CRC patients, suggesting that T cells expressing TIM-3 could be activated cells with improved anti-tumor activities.

Most available studies investigated expression of ICs in bulk tumor tissues but not on specific CD4+ and CD8+ T cell subsets. To date, this is the first study to investigate the associations of different CD4+ Treg subsets and immune checkpoints-expressing CD4+ T cells with DFS in CRC patients. In addition to CD4+ T cell subsets in this study, we have also investigated the association of CD8+ T cell subsets with DFS in CRC patients (Alsalman et al., submitted for publication). However, multi-center investigations are required to confirm these findings in larger cohorts of patients. Moreover, additional investigations are required to determine the exact function of these cell subsets in the TME and circulation of CRC patients. Our data suggest that different CD4+ Treg/T cell subsets in circulation or in the TME play different roles in DFS of CRC patients. Clearly, identification of the exact subpopulations contributing to clinical outcomes is critical for prognosis purposes and therapeutic approaches.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CRC:

Colorectal cancer

CTLA-4:

Cytotoxic T-lymphocyte-associated antigen 4

DFS:

Disease-free survival

FoxP3:

Forkhead box P3

ICs:

Immune checkpoints

LAG-3:

Lymphocyte-activation gene 3

NILs:

Normal tissue-infiltrating lymphocytes

NT:

Normal tissues

PBMC:

Peripheral blood mononuclear cell

PD-1:

Programmed cell death-1

TILs:

Tumor-infiltrating lymphocytes

TIM-3:

T-cell immunoglobulin and mucin domain-3

TME:

Tumor microenvironment

Treg:

T regulatory cell

TT:

Tumor tissues

References

  1. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  PubMed  Google Scholar 

  2. Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. West NR, et al. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol. 2015;15(10):615–29.

    Article  CAS  PubMed  Google Scholar 

  4. Chaudhary B, Elkord E. Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines (Basel). 2016;4(3):28. https://doi.org/10.3390/vaccines4030028.

  5. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–18.

    Article  CAS  PubMed  Google Scholar 

  6. Saito T, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22(6):679–84.

    Article  CAS  PubMed  Google Scholar 

  7. Hu G, Li Z, Wang S. Tumor-infiltrating FoxP3(+) Tregs predict favorable outcome in colorectal cancer patients: A meta-analysis. Oncotarget. 2017;8(43):75361–71.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Toor SM, et al. Immune Checkpoints in Circulating and Tumor-Infiltrating CD4(+) T Cell Subsets in Colorectal Cancer Patients. Front Immunol. 2019;10:2936.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Salama P, et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 2009;27(2):186–92.

    Article  PubMed  Google Scholar 

  10. Betts G, et al. Suppression of tumour-specific CD4+ T cells by regulatory T cells is associated with progression of human colorectal cancer. Gut. 2012;61(8):1163–71.

    Article  CAS  PubMed  Google Scholar 

  11. Kim HJ, et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. 2015;350(6258):334–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Saleh R, et al. Expression of immune checkpoints and T cell exhaustion markers in early and advanced stages of colorectal cancer. Cancer Immunol Immunother. 2020;69(10):1989–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Elkord E, Sasidharan Nair V. T-Regulatory Cells in Health and Disease. J Immunol Res. 2018;2018:5025238.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Elkord E. Helios Should Not Be Cited as a Marker of Human Thymus-Derived Tregs. Commentary: Helios(+) and Helios(-) Cells Coexist within the Natural FOXP3(+) T Regulatory Cell Subset in Humans. Front Immunol. 2016;7:276.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Elkord E, Al-Ramadi BK. Helios expression in FoxP3(+) T regulatory cells. Expert Opin Biol Ther. 2012;12(11):1423–5.

    Article  CAS  PubMed  Google Scholar 

  17. Syed Khaja AS, et al. Intratumoral FoxP3(+)Helios(+) Regulatory T Cells Upregulating Immunosuppressive Molecules Are Expanded in Human Colorectal Cancer. Front Immunol. 2017;8:619.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Darvin P, et al. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1–11.

    Article  PubMed  CAS  Google Scholar 

  19. Mehdizadeh S, et al. Immune checkpoints and cancer development: Therapeutic implications and future directions. Pathol Res Pract. 2021;223:153485.

    Article  CAS  PubMed  Google Scholar 

  20. Kennedy R, Celis E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol Rev. 2008;222:129–44.

    Article  CAS  PubMed  Google Scholar 

  21. Borst J, et al. CD4(+) T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18(10):635–47.

    Article  CAS  PubMed  Google Scholar 

  22. Liu VC, et al. Tumor evasion of the immune system by converting CD4+CD25- T cells into CD4+CD25+ T regulatory cells: role of tumor-derived TGF-beta. J Immunol. 2007;178(5):2883–92.

    Article  CAS  PubMed  Google Scholar 

  23. Ling KL, 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 

  24. Timperi E, et al. Regulatory T cells with multiple suppressive and potentially pro-tumor activities accumulate in human colorectal cancer. Oncoimmunology. 2016;5(7):e1175800.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Szeponik L, et al. Intratumoral regulatory T cells from colon cancer patients comprise several activated effector populations. BMC Immunol. 2021;22(1):58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ahlmanner F, et al. CD39(+) regulatory T cells accumulate in colon adenocarcinomas and display markers of increased suppressive function. Oncotarget. 2018;9(97):36993–7007.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Correale P, et al. Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J Immunother. 2010;33(4):435–41.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ling A, et al. The intratumoural subsite and relation of CD8(+) and FOXP3(+) T lymphocytes in colorectal cancer provide important prognostic clues. Br J Cancer. 2014;110(10):2551–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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(7):909–18.

    Article  CAS  PubMed  Google Scholar 

  30. Nosho K, et al. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol. 2010;222(4):350–66.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zeestraten EC, et al. FoxP3- and CD8-positive Infiltrating Immune Cells Together Determine Clinical Outcome in Colorectal Cancer. Cancer Microenviron. 2013;6(1):31–9.

    Article  CAS  PubMed  Google Scholar 

  32. Yoon HH, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Suzuki H, et al. Intratumoral CD8(+) T/FOXP3 (+) cell ratio is a predictive marker for survival in patients with colorectal cancer. Cancer Immunol Immunother. 2010;59(5):653–61.

    Article  CAS  PubMed  Google Scholar 

  34. Clarke SL, et al. CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS ONE. 2006;1(1):e129.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Miyara M, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899–911.

    Article  CAS  PubMed  Google Scholar 

  36. Elkord E, Abd Al Samid M, B. Chaudhary, Helios, and not FoxP3, is the marker of activated Tregs expressing GARP/LAP. Oncotarget. 2015;6(24):20026–36.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yu WQ, et al. Coexpression of Helios in Foxp3(+) Regulatory T Cells and Its Role in Human Disease. Dis Markers. 2021;2021:5574472.

    PubMed  PubMed Central  Google Scholar 

  38. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015;6(6):e1792.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gasser M, et al. PD-1/PD-L1 expression in colorectal cancer and its implications for apoptosis and tumor immune evasion. Can Res. 2006;66(8 Supplement):1118.

    Google Scholar 

  40. Cantero-Cid R, et al. PD-L1/PD-1 crosstalk in colorectal cancer: are we targeting the right cells? BMC Cancer. 2018;18(1):945.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Llosa NJ, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5(1):43–51.

    Article  CAS  PubMed  Google Scholar 

  42. Zhou X, et al. Upregulation of TIGIT and PD-1 in Colorectal Cancer with Mismatch-repair Deficiency. Immunol Invest. 2021;50(4):338–55.

    Article  CAS  PubMed  Google Scholar 

  43. Zhou E, et al. Up-regulation of Tim-3 is associated with poor prognosis of patients with colon cancer. Int J Clin Exp Pathol. 2015;8(7):8018–27.

    PubMed  PubMed Central  Google Scholar 

  44. Sillo TO, et al. Mechanisms of immunogenicity in colorectal cancer. Br J Surg. 2019;106(10):1283–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Arora SP, Mahalingam D. Immunotherapy in colorectal cancer: for the select few or all? J Gastrointest Oncol. 2018;9(1):170–9.

    Article  PubMed  PubMed Central  Google Scholar 

  46. He Y, et al. TIM-3, a promising target for cancer immunotherapy. Onco Targets Ther. 2018;11:7005–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Arai Y, Saito H, Ikeguchi M. Upregulation of TIM-3 and PD-1 on CD4+ and CD8+ T Cells Associated with Dysfunction of Cell-Mediated Immunity after Colorectal Cancer Operation. Yonago Acta Med. 2012;55(1):1–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang X, et al. Differential expression of TIM-3 between primary and metastatic sites in renal cell carcinoma. BMC Cancer. 2019;19(1):49.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Lee I, et al. LAG-3 expression in tumor infiltrating immune cells is associated with poor prognosis in patients with microsatellite instability high colon cancer. Ann Oncol. 2017;28:v196.

    Article  Google Scholar 

  50. Chen J, Chen Z. The effect of immune microenvironment on the progression and prognosis of colorectal cancer. Med Oncol. 2014;31(8):82.

    Article  CAS  PubMed  Google Scholar 

  51. Goldstein J, et al. Multicenter retrospective analysis of metastatic colorectal cancer (CRC) with high-level microsatellite instability (MSI-H). Ann Oncol. 2014;25(5):1032–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Le Gouvello S, et al. High prevalence of Foxp3 and IL17 in MMR-proficient colorectal carcinomas. Gut. 2008;57(6):772–9.

    Article  PubMed  Google Scholar 

  53. Frey DM, 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(11):2635–43.

    CAS  PubMed  Google Scholar 

  54. Cavalleri T, et al. Combined Low Densities of FoxP3(+) and CD3(+) Tumor-Infiltrating Lymphocytes Identify Stage II Colorectal Cancer at High Risk of Progression. Cancer Immunol Res. 2019;7(5):751–8.

    Article  CAS  PubMed  Google Scholar 

  55. Kawakami H, Zaanan A, Sinicrope FA. Microsatellite instability testing and its role in the management of colorectal cancer. Curr Treat Options Oncol. 2015;16(7):30.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank all patients for donating their samples.

Funding

This work was supported by University of Nizwa.

Author information

Authors and Affiliations

Authors

Contributions

MAA analyzed data and wrote the manuscript. AA assisted in manuscript writing. KM and AAD provided the clinicopathological data and revised the manuscript. EE conceived the idea, designed the study, supervised the project, analyzed and interpreted data, and wrote and revised the manuscript. All authors were involved in the final approval of the manuscript.

Corresponding author

Correspondence to Eyad Elkord.

Ethics declarations

Ethics approval and consent to participate

This study was executed under ethical approval from the Medical Research Center, Hamad Medical Corporation, Doha, Qatar (Protocol no. MRC-02–18-012). The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Qatar Biomedical Research Institute. Written informed consent was obtained from all subjects and/or their legal guardian(s) for study participation.

Consent for publication

Informed consent for publication was obtained from all subjects involved in the study.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher’s Note

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

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

Al-Mterin, M.A., Murshed, K., Alsalman, A. et al. Associations of different immune checkpoints-expressing CD4+ Treg/ T cell subsets with disease-free survival in colorectal cancer patients. BMC Cancer 22, 601 (2022). https://doi.org/10.1186/s12885-022-09710-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-022-09710-1

Keywords