ISSN: 2455-8591
International Journal of Immunotherapy and Cancer Research
Review Article       Open Access      Peer-Reviewed

The Biomarkers of Cd4+ T Regulatory Cells Associated with Tumour Immune Escape

Nyaribari MC*

Pears Biotechnologies, PO BOX 28412 – 00100, Nairobi, Kenya
*Corresponding author: Nyaribari MC, Pears Biotechnologies, PO BOX 28412 – 00100, Nairobi, Kenya, Tel: +254700092562; E-mail: matundaconradus@gmail.com
Received: 19 October, 2020 | Accepted: 26 October, 2020 | Published: 28 October, 2020
Keywords: CD4+FOXP3+; CD4+CD127-; CD4+CD39+ or CD73+ and CD4+LAP+ cancer immunotherapy; Cancer immunotherapy strategies

Cite this as

Nyaribari MC (2020) The Biomarkers of Cd4+ T Regulatory Cells Associated with Tumour Immune Escape. Int J Immunother Cancer Res 6(1): 028-034. DOI: 10.17352/2455-8591.000028

Objective: In this review, we endeavor to do a literature search mainly focusing on keywords CD4+FOXP3+, CD4+CD127-, CD4+CD39+, CD4+LAP+ cancer immunotherapy, cancer immunotherapy strategies and articles published between 2015 to 2019 to add onto the minimal definition of human Treg, by an international workshop organized by collaborative immunoguiding program.

Methodology: In this review, we highlight the antitumor suppressive biomarkers of CD4+ Treg cells, how they suppress the immune response.

Summary: The biomarkers play a role in designing of cancer immunotherapy to overcome resistance and enhance anti-tumor immune response among late-stage cancer patients who have exhausted the standard of care. There is evidence suggesting that a combination of treatment strategies has enhanced immune responses for some patients who have developed or are resistant to monoclonal immunotherapy treatments.

Introduction

There have been advances made in precision oncology in designing effective immunotherapies against cancer. However, it does not appear to benefit all cancer patients, as some have either developed resistance or do not respond at all to FDA approved immunotherapies. And we are yet to understand the mechanism underlying this resistance and why some patients appear to benefit while others do not. Therefore, it is paramount for us to understand the inhibitory mechanism and synergies between various CD4+ Treg cells associated with tumor immune escape. As such we chose the five subsets as there are several studies done on the post the minimal definition of Tregs. The biomarkers expressed of CD4+ Treg cells do make these regulatory cells more potent in suppressing effector immune responses against cancer cells. It is these biomarkers that have been used to monitor the efficacy of immunotherapy treatments and measure the treatment progress of cancer patients. Therefore, there is a need to design cancer immunotherapies against these antigens to enhance the body’s effector immune response against cancer. Some studies suggest that inhibiting the expression of these biomarkers would improve anti-tumor immune response studies such as CD4+FOXP3+ Treg cells suppress the anti-tumor immune response, and it is an indication of poor patient prognosis [1-7]. 

Mechanism of tumor immune escape via CD4+CD25+FOXP3+ Treg cells

The first subset of CD4+ Treg cells to be described was the CD4+CD25+FOXP3+ cells discovered in several cancer types where it prevents autoimmune diseases, as activated suppressive markers where it hampered the Teff. The Foxp3 is highly expressed in CD4+ T cells, and inhibit the proliferative activity of naïve CD4+ T cells [7], as it is an intracellular transcription factor and the master of all Treg cells as such its important to describe it here. However, reports indicate that the FOXP3 gene mutation may contribute to carcinogenesis [8], or gene promoter demethylation [7], the Foxp3 is an intracellular undruggable protein to date [9].

As such, there is evidence suggesting that CD4+CD25+FOXP3+ cell accumulation correlates with age among the elderly with Lung cancer, where the reduced T-cell mediated anti-tumor immunity contributes to immune evasion at the early stage, finally leading to high morbidity and mortality among the elderly with lung cancer [2]. Urinary bladder cancer [3] gastric cancer [4-6] non-small cell lung cancer [7-11] colorectal cancer [12-15] breast cancer [16] oral squamous cells carcinoma [17,18] cervical cancer in mice5 pediatric B-cell acute lymphocytic leukemia [19] prostate cancer [20] and head and neck squamous cell carcinoma [21]. It is also found in tumor-infiltrating lymphocytes, and abundant in the late stages of gastric cancer [22]. The study demonstrates that thyroid cancer cells express a high level of functional FOXP3+, that the inhibition of the FOXP3+ suppresses the proliferation, migration but promotes apoptosis suggesting that targeting FOXP3+ in thyroid cancer cells may offer a novel therapeutic option for thyroid cancer [23].

The CD4+CD25+FOXP3+ serves as a master prognostic biomarker and a crucial determinant of immunosuppressive microenvironment via recruiting Treg cells by directly trans-activating CCL5. Therefore, FOXP3+ could be used to select patients with better responses to CCL5/CCR5 blockade immunotherapy [24]. In additional they are markers associated with the master FOXP3+ transcription factor, which make it more suppressive such as interleukin-IL-12 [5,25], interleukins IL-10 [26], IL-2 & 35 [24], interferons such as IFN-γ [5,16], transforming growth factor TGF-β [19,25,26], tumor necrosis factor receptor type-11 [14], CCR5, CCR7, and their ligands CCL5, CCL19, and CCL21 [24]. Besides, there are additional anti-tumor suppressor markers associated with FOXP3 including PD-L1 [8,18], PD-L1/CTLA-4, PD-1/CD39 [13], CTLA-4, LAG-3 [8], excessive activation of IL-2/ pSTAT5, and TGF-β/Smad signaling and insufficient pSTAT3 in case of acute lymphocytic leukemia [19], suppression of NKG20- mediated NK cell cytotoxicity and contact-dependent manner [26].

No known immunotherapy directly blocks the CD4+CD25+FOXP3+ Treg cells in humans. There is a suggestion that methionine enkephalin an endogenous neuropeptide inhibited the expression of FOXP3 during the process of TGF-βinduction, which is accompanied by diminishing phosphorylation and nuclear translocation of Smad2/3 in S180 tumor-bearing mice [27]. Another example indicated that the INF-X-2b inhibits cancer cell immune evasion by decreasing levels of CD4+FOXP3+ suppressing TGF-β and IL-10 in the tumor microenvironment [28] Genetic targeting of Usp7 impairs FOXP3 + Treg suppressive functions by stabilizing the expression and promoting the multimerization of histone/protein acetyltransferaseTip60 and FOXP3+ [29]. But these mechanisms do not directly affect biomarkers of Treg cells. There are efforts out there trying to discover or innovate new therapies that may overcome the FOXP3 inhibitors.

In this article, we focus on the markers of Treg cells, since FOXP3 is a transcription factor and difficult to target. We explore other ways to deplete the anti-tumor suppressive effects of FOXP3 by using other markers co-expressed on Treg cells. Therefore, studies on the anti-CTLA-4 mAbs on mice models, indicated that it selectively depleted intratumoral FOXP3+ regulatory T cells via an Fc-dependent mechanism [18,30,31] And a humanized anti-CCR4 monoclonal antibody, which functions in an antibody-dependent cellular cytotoxicity activity depleting CD4+CD25+FOXP3+ increasing survivor rates [32] Further, studies suggest that Mogamulizumab an anti-CCR4 monoclonal antibody with a defucosylated Fc region (Potelligent® Technology), enhances antibody-dependent cellular cytotoxicity by increasing its binding affinity to the Fcx03B3; receptor expressed on effector cells [33,34] Effective on patients with CCR4-positive adult T-cell leukemia and peripheral T-cell lymphoma [35] Whereas Dao and colleagues, generated a T cell receptor mimic antibody, “FOXP3-#32,” recognizing a FOXP3-derived epitope in the context of HLA-A*02:01. Selectively recognizes CD4+CD25+CD127low and FOXP3+ Tregs and depletes these cells via antibody-mediated cellular cytotoxicity in xenografts of PBMCs from a healthy donor and ascites fluid from cancer patient [36]. We suggest that immunotherapy targeting CD4+CD25+FOXP3+ should focus on other co-expressed markers on the CD4+ Treg cells because these markers enhance the anti-tumor suppressive mechanism of CD4+CD25+FOXP3+ T regulatory cells. As indicated on the table below Table 1.

Inhibition mechanism of CD4+ CD127-Treg to enhance tumor immune escape

The CD4+CD25hiCD127low/- Treg cells have been detected in follicular lymphoma,39 childhood acute lymphoblastic leukemia [39] peripheral blood of gastric cancer [40,41] nasopharyngeal carcinomas [45] in peripheral blood of non-small cell lung cancer [20,46] colorectal cancer [47,63] hepatocellular carcinoma [42] breast cancer [43] lung adenocarcinoma cells [44]

The CD127 suppresses anti-tumor responses PD-1+ and Tim3+ elevated in gastric cancer in gastric cancer [40]. Treg cells from late stages of Colitis-Associated Colon Cancer CAC displayed an activated phenotype by expressing PD1, CD127 and Tim-3, suggesting an increased suppressive capacity [64] The cytokines such as IL-35 upregulated in colorectal cancer [47] IL-10, and TGF-β secretion in hepatocellular carcinoma [29]. And increased in both plasma concentrations of IL-2, IL-4, IL-6, IL-10, and proportions of latency-associated peptide LAP/TGF-β [44].

CD4+CD25hiCD127low/- elevated IFN-γ and IL-21 secretion, and it acted a cell-to-cell contact-dependent manner and depended on IL-6 secretion [65]. And activated by three main pathways STAT5, PI3K/Akt/mTOR and MEK/Erk [49]. There is evidence suggesting that CD4+CD25hiCD127low/- host cells are major targets of anti-CD127 that modulate therapeutic CD8+ T cell responses and the outcome of anti-CD127 -assisted [66]. By contrast, excessive CD4+CD25hiCD127low/- mediated signaling can drive lymphoid leukemia development, disease acceleration and resistance to chemotherapy [49] combination therapy relies on the interdependence between IL-7 and IFN-γ signaling it increases CD4+CD25hiCD127low/- the expression on tumor-infiltrating T cells in an IFN-γ/IFN-γR signaling-dependent manner, which could be an effective modality to improve immunotherapeutic efficacy [48].

How treatment is carried out by blockade of PD-1+ and Tim-3+ inhibition as a synergistic effect on IFN-γ secretion [41] blockades of Notch signaling inhibits suppression function of CD127dim/- in gastric cancer [40].

Effects of adenosine mediated immunesuppression by CD4+CD39+Treg

The CD39+ detected in colorectal cancer [53] breast tumour [50,51] gastric cancer [52] Colon cancer [54] colorectal cancer [55] chronic lymphocytic leukemia [55] and hepatocellular carcinoma [56], Ovarian cancer [67] myeloma [68] adult T-cell leukemia/lymphoma [58]. Frequencies of CD39+, PD-1+, and CD39+/PD-1+cells were higher among both CD4+ and CD8+ T cells isolated from NSCLC tumor tissue [57].

The ectonucleotidases CD39 and CD73 hydrolyze extracellular adenosine triphosphate and adenosine diphosphate to generate adenosine, which binds to adenosine receptors and inhibits T-cell and natural killer–cell responses thereby suppressing the immune system [69]. The CD39+ the suppress anti-tumor immune response via the adenosine-mediated pathway but independent of TGF-β or IL-10 and secrete IL-17A and GM-CSF. CD4+CD25hiCD39+ Tregs inhibit the proliferative response and the secretion of IL-17 and IFN-γ of autologous CD4+ T effector cells [68]. Which chemoattract myeloid-derived suppressive cells [53], suppressive capacity of CD39+ Treg on proliferation, and IFN-γ secretion by conventional T cells colon cancer [54]. CD39+CD73+ cancer cells inhibited the proliferation of CD4 and CD8 T cells and the generation of cytotoxic effector CD8 T cells in a CD39- and adenosine-dependent manner [69]. Through cooperation between CD39+ Treg and CD73+ expressing Th1/Th17 subset in breast cancer [70] Adenosine derived from the degradation of ATP via ectonucleotidases CD39 and CD73 is a critical immunosuppressive metabolite in the hypoxic microenvironment of tumor tissue and Adenosine signaling via A2aR can inhibit the antitumor immune response of CD8+ T cells [70-73]. The generation of adenosine by CD73 also suppresses antitumor immune responses through the activation of A2A receptors on T cells and natural killer cells [74-76] They express the Th17-associated surface markers CCR6 and IL-23R and phosphorylate the transcription factor Stat3. Further, suppression of IL-17 by CD4+CD25hiCD39+ Tregs occurs via a Stat3-dependent mechanism as inhibition of Stat3 activation in the CD39+ Tregs reverses their ability to suppress IL-17 [77,78].

The potential of CD73 as a double-edged sword in anti-leukemia immunity and argue strongly for the combinational treatment by adding immune checkpoint inhibitors to the CD73-targeting approaches [59,79] it has high expression of immunosuppressive surface molecules such as ICOS, PD-L1, and CTLA-4 [54]. The anti-CD39+ monoclonal antibody is a selective and potent CD39 enzymatic inhibitor capable of preventing adenosine-mediated immune suppression and increasing T-cell activation in the tumor microenvironment [80]. Anti-CD39 treatment alleviated the tumor-induced inhibition of CD4 and CD8 T-cell proliferation and increased CTL- and NK cell-mediated cytotoxicity [70]. Anti-CD39+ reduced Tregs, increased the CD8 / Treg ratio and reduced CD73 expression on immune suppressive cells, and in combination with radiation resulted in enhanced efficacy when compared to either agent [61].

CD4+LAP+ Treg-mediated suppression of anti-tumor immune response

The latency-associated peptide (LAP) is a recently discovered subset of CD4+ Treg cells. LAP+ Treg cells accumulate in the tumor microenvironment of colorectal cancer, were elevated in liver metastasis from colorectal cancer [81]. It is present as clusters in the tumor stroma of patients with hepatocellular carcinoma [82] in tumor-infiltrating B-cells [83] and gastric carcinoma [60]. And it is 50-fold more potent immunosuppressive ability than traditional CD4+CD25+ T cells [84],

The anti-LAP suppresses anti-tumor immune response through IL-10, TGF-γ [84]. IL-10 in liver metastasis [61,85] Anti-LAP antibody targets the LAP/TGF-β complex on Treg to enhance immune responses and reduces tumor growth by increasing the infiltration of tumors by cytotoxic CD8+ T cells [83]. Anti-LAP decreases LAP+ Tregs, tolerogenic dendritic cells and TGF-β secretion, and is associated with CD8+ T cell activation, with increased expression of CTLA-4 and IL-10 and decreased expression of IFN-γ, TNF-α, and granzymes.79 85 Thus, anti-LAP targets multiple immunoregulatory pathways and represents a potential approach for cancer immunotherapy [83]. LAP+CD4+ T cells showed lower Foxp3 expression but significantly higher levels of CTLA-4, CCR4, and CCR5 [81]. LAP+CD4+ T cells expressed significantly higher amounts of IL-10 and TGF-β but lower levels of IL-2, IL-4, IL-17, and interferon-γ, compared with LAP-CD4+ T cells [81].

Furthermore, within the HCC tissues, LAP CD4 T cells were a gift as clusters within the neoplasm stroma and closely related to CD4 T lymphocytes in contrast, within the peri‑cancer liver tissues and HBV‑infected viscus tissues around benign lesions LAP, CD4 T cells sparsely distributed [83]. LAP+CD4+ T cells have anti-tumor suppressive effects within the peripheral blood of neoplasm tissues, and it is a factor in the suppression of anti‑tumor immunity in the neoplasm cells [83]. Anti-LAP antibodies inhibit the discharge, inhibit neoplasm growth in mouse models [84] and have promise as a novel cancer medicine the situation of the LAP-TGFβ1 advanced is of crucial biological, clinical importance, once the mature TGFβ1 protein, is free, it acts domestically, either in associate degree autocrine or close to paracrine fashion [85]. These results warrant additional analysis to work out the effectiveness of anti-LAP in inhibiting the discharge and its effects on immunological disorder within the neoplasm microenvironment [86]. They compared the suppressive activity of CD4+CD25+ regulatory T cells (conventional Treg) with T cells expressing T cell immunoglobulin-3+ (TIM-3+) and latency-associated peptide (LAP)+ T cells [87]. They found that LAP-expressing T cells were more suppressive than conventional Treg, but TIM-3-expressing T cells were not suppressive [88].

Summary

The biomarkers such as PD-L1, CTLA-4 are markers of CD4 Treg cells commonly found on most cancers are thus far been exploited in designing cancer immunotherapies. Some patients do not benefit from them, either because of resistance or outrightly not effective. In overcoming resistance and the low number of patients who benefit from cancer immunotherapies treatment, we suggest that the combination of antibodies. Several studies are ongoing either on animal models or on clinical trials that have tried the combination or single analysis of antibodies against CD4 Treg cell’s surface markers. The other biomarkers such as CD39, CD127-, FOXP3, or LAP are more potent in suppressing the anti-tumor immune response, and the antibodies developed against these biomarkers as been effective in eliciting immune responses against several cancers. However, it is the anti-PD-L1 and CTLA-4 that have been approved for use. From this review, we can recommend the combination of these biomarkers will be more effective if they are combined to design polyclonal cancer immunotherapies taking into account individual cancer patient’s tumor microenvironment and their status of the immune system. There is a future in precision oncology, whereby polyclonal immunotherapies will be the standard of treatment.

There is a need for a precision oncologist to adopt polyclonal immunotherapy to combat cancer, especially in patients who have exhausted the standard of care. We can personalize and administer a combination of antibodies depending on the number and type of antigen markers present on specific cancer cells to elicit anti-tumor immune responses. In genetically engineered cancer models using mass cytometry, they observed that the immune activation was evident and systemic. However, only peripheral immune cells sustained their proliferation upon tumor rejection. This systemic response was coordinated across tissues and required for tumor medication in several immunotherapy models. But an emerging population of peripheral CD4 T cells conferred protection against new tumors and was significantly expanded in patients responding to immunotherapy. We recommend further research on the dual, triple, and multiple combinations of immunotherapies, chemotherapy, radiation, and surgical to combat tumor immune escape.

We acknowledge the staff or Pears Biotechnologies and Kenyatta University for their support and motivation in writing this review article.

  1. Tanaka A, Sakaguchi S (2017) Regulatory T cells in cancer immunotherapy. Cell Res 27: 109-118. Link: https://bit.ly/3kBR5CX
  2. Hou PF, Zhu LJ, Chen XY, Qiu ZQ (2017) Age-related changes in CD4+CD25+FOXP3+ regulatory T cells and their relationship with lung cancer. PLoS One 12: e0173048. Link: https://bit.ly/37OAMzm
  3. Jóźwicki W, Brozyna AA, Siekiera J, Slominski AT (2016) Frequency of CD4+CD25+Foxp3+ cells in peripheral blood in relation to urinary bladder cancer malignancy indicators before and after surgical removal. Oncotarget 7: 11450-11462. Link: https://bit.ly/3oBeo2n
  4. Chen X, Zhu B, Luo Y, Zhang D, Zhang L, et al. (2015) Interleukin-28B plays a therapeutic role on mouse U14 cervical cancer cells by down-regulating CD4+CD25+FoxP3+regulatory T cells in vivo. Int J Gynecol Cancer 25: 1369-1376. Link: https://bit.ly/34xnqFy
  5. Kindlund B, Sjöling Å, Yakkala C, Adamsson J, Janzon A, et al. (2017) CD4+ regulatory T cells in gastric cancer mucosa are proliferating and express high levels of IL-10 but little TGF-β. Gastric Cancer 20: 116-125. Link: https://bit.ly/2Hy4bTL
  6. Attias M, Al-Aubodah T, Piccirillo CA (2019) Mechanisms of human FoxP3+ Treg cell development and function in health and disease. Clin Exp Immunol 197: 36-51. Link: https://bit.ly/34A8u9J
  7. Hu X, Gu Y, Zhao S, Hua S, Jiang Y (2019) Elevated Circulating CD4+CD25-Foxp3+ Regulatory T Cells in Patients with Nonsmall Cell Lung Cancer. Cancer Biother Radiopharm 34: 325–333. Link: https://bit.ly/2TvEOUL
  8. Wei T, Zhang J, Qin Y, Wu Y, Zhu L, et al. (2015) Increased expression of immunosuppressive molecules on intratumoral and circulating regulatory T cells in non-small-cell lung cancer patients. Am J Cancer Res 5: 2190-201. Link: https://bit.ly/2HDR3ft
  9. Guo J, Liu Z, Zhang Z (2015) Study of CD4+ CD25+ Foxp3+ regulatory T cells in peripheral blood of non-small-cell lung cancer patients. Chinese Journal of Primary Medicine and Pharmacy 22: 510-513. Link: https://bit.ly/3oteokV
  10. Jackute J, Zemaitis M, Pranys D, Sitkauskiene B, Miliauskas S, et al. (2015) The prognostic influence of tumor infiltrating Foxp3+CD4+, CD4+ and CD8+ T cells in resected non-small cell lung cancer. J Inflamm 12: 63. Link: https://bit.ly/34Bmakw
  11. Tøndell A, Wahl SGF, Sponaas AM, Sørhaug S, Børset M, et al. (2020) Ectonucleotidase CD39 and Checkpoint Signalling Receptor Programmed Death 1 are Highly Elevated in Intratumoral Immune Cells in Non–small-cell Lung Cancer. Transl Oncol 13: 17–24. Link: https://bit.ly/2TvEVQb
  12. Zhu XW, Zhu HZ, Zhu YQ, Feng MH, Qi J, et al. (2016) Foxp3 expression in CD4+CD25+Foxp3+ regulatory T cells promotes development of colorectal cancer by inhibiting tumor immunity. J Huazhong Univ Sci Technol Med Sci 36: 677-682. Link: https://bit.ly/31Q5l3J
  13. Khaja ASS, Toor SM, Salhat HE, Ali BR, Elkord E (2017) Intratumoral FoxP3+Helios+ regulatory T Cells upregulating immunosuppressive molecules are expanded in human colorectal cancer. Front Immunol 8: 619. Link: https://bit.ly/35FGvEP
  14. Yan F, Du R, Wei F, Zhao H, Yu J, et al. (2015) Expression of TNFR2 by regulatory T cells in peripheral blood is correlated with clinical pathology of lung cancer patients. Cancer Immunol. Immunother 64: 1475-1485. Link: https://bit.ly/2Tyg7XH 
  15. Jasim BS, Dawood DS, Ali SHM (2015) www. ijarbs. com Research Article Immunohistochemical Expression of FOXP3+ CD4+ CD25+ Regulatory T Cells Suppress Anti-Tumor Immune Responses in Patients with Colorectal Cancer. Int J Adv Res Biol Sci 2: 30-38. Link: https://bit.ly/2G48Wn1  
  16. Jafarinia M, Mehdipour F, Hosseini SV, Ghahramani L, Hosseinzadeh M, et al. (2016) Determination of a CD4+CD25−FoxP3+ T cells subset in tumor-draining lymph nodes of colorectal cancer secreting IL-2 and IFN-γ. Tumor Biol 37: 14659-14666. Link: https://bit.ly/3ostWFo
  17. 89PAbundance of Treg cells in oral cancer patients and effects of their inhibition on growth of cancer cells. Link: https://bit.ly/2TsZmgE
  18. Liu SX, Xiao HR, Wang GB, Chen XW, Li CG, et al. (2018) Preliminary investigation on the abnormal mechanism of cd4+foxp3+cd25high regulatory t cells in pediatric b-cell acute lymphoblastic leukemia. Exp Ther Med 16: 1433-1441. Link: https://bit.ly/34zegrU
  19. Liu Z, McMichael EL, Shayan G, Li J, Chen K, et al. (2018) Novel effector phenotype of TIM-3þ Regulatory T cells leads to enhanced suppressive function in head and neck cancer patients. Clin Cancer Res 24: 4529-4538. Link: https://bit.ly/3jBPP1x
  20. Zhang B, Li G, Ye J, Li Z (2015) Changes of CD4 + CD25 + Foxp3 + regulatory T cells in the peripheral blood and their correlation with insulin resistance in different stages of prostate cancer. Zhonghua Nan Ke Xue 21: 420-423. Link: https://bit.ly/3e75ZyS
  21. Sayapina MS, Bykovskaia SN (2018) The Plasticity of CD4+CD25+FOXP3+CD127low T Cells in Patients with Metastatic Renal Cell Carcinoma in the Course of Interferon-Alpha Immunotherapy. J Oncol 7828735. Link: https://bit.ly/34B9AC1
  22. Nagase H, Takeoka T, Urakawa S, Morimoto-Okazawa A, Kawashima A, et al. (2017) ICOS + Foxp3 + TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with H elicobacter pylori. Int J Cancer 140: 686-695. Link: https://bit.ly/37NtEDl
  23. Chu R, Liu SY, Vlantis AC, van Hasselt CA, Ng EK, et al. (2015) Inhibition of Foxp3 in cancer cells induces apoptosis of thyroid cancer cells. Mol Cell Endocrinol 399: 228-234. Link: https://bit.ly/2TCLmAR
  24. Das SN, Aggarwal S, Sharma SC (2017) Abstract A67: Phenotypic and functional dynamics of CD4+ CD25+ FOXP3+ regulatory T cells in patients with tobacco-related oral squamous cell carcinoma. Link: https://bit.ly/3e5qcVF
  25. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, et al. (2016) Two FOXP3 + CD4 + T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 22: 679–684. Link: https://bit.ly/3mueSWa
  26. Geng X, Li M, Cui B, Lu C, Liu X, et al. (2019) CD4+CD25+Foxp3+ regulatory T cells suppress NKG2D-mediated NK cell cytotoxicity in peripheral blood. Medicine (Baltimore). 98: e15722. Link: https://bit.ly/3mrIH9K
  27. Li, X, Meng Y, Plotnikoff NP, Youkilis G, Griffin N, et al. (2015) Methionine Enkephalin (MENK) inhibits tumor growth through regulating CD4+Foxp3+ regulatory T cells (Tregs) in mice. Cancer Biol Ther 16: 450-459. Link: https://bit.ly/3e0RWL8
  28. Yu Y, Huang R, Zong X, He X, Mo W (2016) INFα-2b inhibitory effects on CD4+CD25+FOXP3+ regulatory T cells in the tumor microenvironment of C57BL/6 J mice with melanoma xenografts. BMC Cancer 16: 397. Link: https://bit.ly/2HG9KPs
  29. Wang J, Yang J (2016) Identification of CD4+CD25+CD127- regulatory t cells and CD14+HLA-DR-/low myeloid-derived suppressor cells and their roles in the prognosis of breast cancer. Biomed Rep 5: 208–212. Link: https://bit.ly/2Jhnm4J
  30. Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, et al. (2019) Anti-CTLA-4 immunotherapy does not deplete Foxp3 þ regulatory T cells (Tregs) in human cancers. Clin Cancer Res 25: 1233-1238. Link: https://bit.ly/2HL54HZ 
  31. Nishikawa H, Sakaguchi S (2014) Regulatory T cells in cancer immunotherapy. Curr Opin Immunol 27: 1-7. Link: https://bit.ly/37NtOKX
  32. Kurose K, Ohue Y, Wada H, Iida S, Ishida T, et al. (2015) Phase Ia Study of FoxP3+ CD4 Treg Depletion by Infusion of a Humanized Anti-CCR4 Antibody, KW-0761, in Cancer Patients. Clin Cancer Res 21: 4327-4336. Link: https://bit.ly/35BgWVo
  33. Ueda R (2015) Clinical Application of Anti-CCR4 Monoclonal Antibody. Oncology 89:16-21. Link: https://bit.ly/3kOkU3z
  34. Ishida T, Joh T, Uike N, Yamamoto K, Utsunomiya A, et al. (2012) Defucosylated anti-CCR4 monoclonal antibody (KW-0761) for relapsed adult T-cell leukemia-lymphoma: A multicenter phase II study. J Clin Oncol 30: 837-842. Link: https://bit.ly/2HJWBVT
  35. Ueda R (2015) Clinical Application of Anti-CCR4 Monoclonal Antibody. Oncology 89: 16-21. Link: https://bit.ly/3kOkU3z
  36. Dao T, Mun SS, Scott AC, Jarvis CA, Korontsvit T, et al. (2019) Depleting T regulatory cells by targeting intracellular Foxp3 with a TCR mimic antibody. Oncoimmunology 8. Link: https://bit.ly/3owhjct
  37. Kannappan V, Butcher K, Trela M, Nicholl I, Wang W, et al. (2017) Interleukin 21 inhibits cancer-mediated FOXP3 induction in naïve human CD4 T cells. Cancer Immunol Immunother 66: 637-645. Link: https://bit.ly/3jy8nzt
  38. Le KS, Thibult ML, Just-Landi S, Pastor S, Gondois-Rey F, et al. (2016) Follicular B lymphomas generate regulatory T cells via the ICOS/ICOSL pathway and are susceptible to treatment by anti-ICOS/ICOSL therapy. Cancer Res 76: 4648-4660. Link: https://bit.ly/3kBpWQI
  39. Niedźwiecki M, Budziło O, Zieliński M, Adamkiewicz-Drożyńska E, Maciejka-Kembłowska L, et al. (2018) CD4+CD25highCD127low/−FoxP3+ Regulatory T Cell Subpopulations in the Bone Marrow and Peripheral Blood of Children with ALL: Brief Report. J Immunol Res 2018: 1292404. Link: https://bit.ly/3oBI530
  40. Yang L, Zhao KL, Qin L, Ji DX, Zhang B, et al. (2019) Notch signaling pathway regulates CD4+CD25+CD127dim/− regulatory T cells and T helper 17 cells function in gastric cancer patients. Biosci Rep 39. Link: https://bit.ly/35GB4FB
  41. Yuan L, Xu B, Yuan P, Zhou J, Qin P, et al. (2017) Tumor-infiltrating CD4+ T cells in patients with gastric cancer. Cancer Cell Int 17: 114. Link: https://bit.ly/37Moszq
  42. Wang J, Lupo KB, Chambers AM, Matosevic S (2018) Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells 11 Medical and Health Sciences 1107 Immunology. J Immunother Cancer 6: 136. Link: https://bit.ly/3jBPaNm
  43. Sharma S, Khosla R, David P, Rastogi A, Vyas A, et al. (2015) CD4+CD25+CD127low Regulatory T Cells Play Predominant Anti-Tumor Suppressive Role in Hepatitis B Virus-Associated Hepatocellular Carcinoma. Front Immunol 6: 49. Link: https://bit.ly/3e1J5J0
  44. LAP TGF-Beta Subset of CD4+CD25+CD127− Treg Cells is Increased and Overexpresses LAP TGF-Beta in Lung Adenocarcinoma Patients. Link: https://bit.ly/31RxKX7
  45. Chen M, Jin F, Ma L (2018) The detection and significance of T cells in nasopharyngeal carcinoma patients. J Cancer Res Ther 14: 331. Link: https://bit.ly/2JdHQLz
  46. Qiu J, Che G, Liu F, Sha X, Ju S, et al. (2019) The detection and clinical significance of peripheral regulatory CD4+CD25hiCD127low T cells in patients with non-small cell lung cancer. Clin Transl Oncol 21: 1343-1347. Link: https://bit.ly/2TvKAWv
  47. Wang S, Yao Y, Yao M, Fu P, Wang W (2018) Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways. Biochem Biophys Res Commun 503: 1605-1609. Link: https://bit.ly/3mp17I0
  48. Shi LZ, Fu T, Guan B, Chen J, Blando JM, et al. (2016) Interdependent IL-7 and IFN-γ signalling in T-cell controls tumour eradication by combined α-CTLA-4+α-PD-1 therapy. Nat Commun 7. Link: https://bit.ly/2Ttn5NR
  49. Oliveira ML, Akkapeddi P, Ribeiro D, Melão A, Barata JT (2019) IL-7R-mediated signaling in T-cell acute lymphoblastic leukemia: An update. Adv Biol Regul 71: 88-96. Link: https://bit.ly/3mr2B4E
  50. Gourdin N, Bossennec M, Rodriguez C, Vigano S, Machon C, et al. (2018) Autocrine adenosine regulates tumor polyfunctional CD73+ CD4+ effector T cells devoid of immune checkpoints. Cancer research 78: 3604-3618. Link: https://bit.ly/35J7CyC
  51. Gourdin N, Bossennec M, Rodriguez C, Vigano S, Machon C, et al. (2018) Autocrine adenosine regulates tumor polyfunctional CD73+CD4+ effector t cells devoid of immune checkpoints. Cancer Res 78: 3604-3618. Link: https://bit.ly/2Tx9Ifq
  52. Cai XY, Wang XF, Li J, Dong JN, Liu JQ, et al. (2015) Overexpression of CD39 and high tumoral CD39+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. Int J Clin Exp Pathol 8: 14757–14764. Link: https://bit.ly/37MgNRD
  53. Hu G, Wu P, Cheng P, Zhang Z, Wang Z, et al. (2017) Tumor-infiltrating CD39+ γδTregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology 6. Link: https://bit.ly/3kBut5E
  54. Ahlmanner F, et al. (2018) CD39 + regulatory T cells accumulate in colon adenocarcinomas and display markers of increased suppressive function. Oncotarget 9: 36993-37007. Link:
  55. Zhulai G, Churov A, Oleinik E, Romanov AA, Semakova VM, et al. (2018) Activation of cd4+cd39+ Т cells in colorectal cancer. Bull Russ State Med Univ 7: 47-53. Link: https://bit.ly/3e1Gv5M
  56. Cai D, Yan LI, Guo R, Zhang T, Chen Z (2016) Correlation of HPV types with Th17 and Treg cells in cervical cancer. Chinese Journal of Clinical Oncology 43: 1099-1102. Link: https://bit.ly/31Paf0U
  57. Tøndell A, Wahl SGF, Sponaas AM, Sørhaug S, Børset M, et al. (2020) Ectonucleotidase CD39 and Checkpoint Signalling Receptor Programmed Death 1 are Highly Elevated in Intratumoral Immune Cells in Non–small-cell Lung Cancer. Transl Oncol 13: 17-24. Link: https://bit.ly/35FAvvF
  58. Nagate Y, Ezoe S, Fujita J, Yokota T, Iochii M, et al. (2018) Ectonucleosidase CD39 Is Highly Expressed on ATLL Cells and Suppresses the Immune Response through the Adenosine Pathway. Blood 132: 3720-3720. Link: https://bit.ly/3jyvH07
  59. De Leve S, Wirsdörfer F, Jendrossek V (2019) Targeting the immunomodulatory CD73/adenosine system to improve the therapeutic gain of radiotherapy. Front Immunol 10: 698. Link: https://bit.ly/3myvH2d
  60. Toor SM, Sasidharan Nair V, Pfister G, Elkord E (2019) Effect of pembrolizumab on CD4+CD25+, CD4+LAP+ and CD4+TIM-3+ T cell subsets. Clin Exp Immunol 196: 345-352. Link: https://bit.ly/31L4bXa
  61. Anti-CD39+ reduced Tregs, increased the CD8 / Treg ratio and reduced CD73 expression on immune suppressive cells, and in combination with radiation resulted in enhanced efficacy when compared to either agent. Link: https://bit.ly/2TsAcyR
  62. Gabriely G, da Cunha AP, Rezende RM, Kenyon B, Madi A, et al. (2017) Targeting latency-associated peptide promotes antitumor immunity. Sci Immunol 2. Link: https://bit.ly/3oyeEz4
  63. Dunne M, Ryan C, Nolan B, Tosetto M, Geraghty R, et al. (2016) Enrichment of inflammatory IL-17 and TNF-a secreting CD4+ T cells within colorectal tumours despite the presence of elevated CD39+ T regulatory cells and increased expression of the immune checkpoint molecule, PD-1. Front Oncol 6: 50. Link: https://bit.ly/3owc9gB
  64. Olguín JE, Medina-Andrade I, Molina E, Vázquez A, Pacheco-Fernández T, et al. (2018) Early and partial reduction in CD4+Foxp3+ regulatory T cells during colitis-associated colon cancer induces CD4+ and CD8+ T cell activation inhibiting tumorigenesis. J Cancer 9: 239-249. Link: https://bit.ly/3e56Gsm
  65. Li S, Wang Z, Zhang G, Fu J, Zhang X (2019) Interleukin-7 promotes lung-resident CD14 + monocytes activity in patients with lung squamous carcinoma. Int Immunopharmacol 67: 202-210. Link: https://bit.ly/2Hzv4GS
  66. Deiser K, Stoycheva D, Bank U, Blankenstein T, Schüler T (2016) Interleukin-7 modulates anti-tullmor CD8+ T cell responses via its action on host cells. PLoS One 11: e0159690. Link: https://bit.ly/31L7mOA
  67. Li, X, Meng Y, Plotnikoff NP, Youkilis G, Griffin N, et al. (2015) Methionine Enkephalin (MENK) inhibits tumor growth through regulating CD4+Foxp3+ regulatory T cells (Tregs) in mice. Cancer Biol Ther 16: 450-459. Link: https://bit.ly/3jBJqDw
  68. Yang R, Elsaadi S, Misund K, Slupphaug G, Menu E, et al. (2018) Abstract LB-117: Role of ectoenzymes CD39 and CD73 in the immune response to multiple myeloma. Link: https://bit.ly/3jBfxD2
  69. Bastid J, Regairaz A, Bonnefoy N, Déjou C, Giustiniani J, et al. (2015) Inhibition of CD39 enzymatic function at the surface of tumor cells alleviates their immunosuppressive activity. Cancer Immunol Res 3: 254-265. Link: https://bit.ly/2HEgbm7
  70. Bossennec M, Rodriguez C, Hubert M, Di-Roio A, Machon C, et al. (1859) Methotrexate Restores CD73 Expression on Th1.17 in Rheumatoid Arthritis and Psoriatic Arthritis Patients and May Contribute to Its Anti-Inflammatory Effect through Ado Production. J Clin Med 8: 1859. Link: https://bit.ly/2J6hdYM
  71. Leone RD, Emens LA (2018) Targeting adenosine for cancer immunotherapy. Journal for ImmunoTherapy of Cancer 6. Link: https://bit.ly/3e5fwqc
  72. Shi L, Feng M, Du S, Wei X, Song H, et al. (2019) Adenosine Generated by Regulatory T Cells Induces CD8+ T Cell Exhaustion in Gastric Cancer through A2aR Pathway. Biomed Res Int 2019: 4093214. Link: https://bit.ly/2G9nDW6
  73. Vigano S, Alatzoglou D, Irving M, Ménétrier-Caux C, Caux C, et al. (2019) Targeting adenosine in cancer immunotherapy to enhance T-Cell function. Front Immunol 10: 925. Link: https://bit.ly/3e0c6Vy
  74. Sek K, Mølck C, Stewart GD, Kats L, Darcy PK, et al. (2018) Targeting adenosine receptor signaling in cancer immunotherapy. Int J Mol Sci 19: 3837. Link: https://bit.ly/3kBJnZx
  75. Leone RD, Sun IM, Oh MH, Sun IH, Wen J,et al. (2018) Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother 67: 1271-1284. Link: https://bit.ly/2TCCQSp
  76. Young A, Ngiow SF, Gao Y, Patch AM, Barkauskas DS, et al. (2018) A2AR adenosine signaling suppresses natural killer cell maturation in the tumor microenvironment. Cancer Res 78: 1003-1016. Link: https://bit.ly/34AdEmc
  77. Magid-Bernstein JR, Rohowsky-Kochan CM (2017) Human CD39+ Treg Cells Express Th17-Associated Surface Markers and Suppress IL-17 via a Stat3-Dependent Mechanism. J Interf Cytokine Res 37: 153-164. Link: https://bit.ly/3ozmPeA
  78. Zhao H, Bo C, Kang Y, Li H (2017) What else can CD39 tell us? Front Immunol 8: 727. Link: https://bit.ly/3msLgIt
  79. Kong Y, Jia B, Zhao C, Claxton DF, Sharma A, et al. (2019) Downregulation of CD73 associates with T cell exhaustion in AML patients. J Hematol Oncol 12: 40. Link: https://bit.ly/3jAu7uL
  80. Lerner (2019) Targeting CD39 with a first-in-class inhibitory antibody prevents ATP processing and increases T-cell activation. The anti-CD39+ monoclonal antibody is a selective and potent CD39 enzymatic inhibitor capable of preventing adenosine-mediated immune suppression and increasing T-cell activation in the tumor microenvironment. Link: https://bit.ly/3kCfKY2
  81. Zhong W, Jiang ZY, Zhang L, Huang JH, Wang SJ, et al. (2017) Role of LAP+CD4+ T cells in the tumor microenvironment of colorectal cancer. World J Gastroenterol 23: 455-463. Link: https://bit.ly/3oBw5P2
  82. Ou X, Guan J, Chen JS, Ying JC, Liu XP, et al. (2018) Lap+cd4+ t cells are elevated among the peripheral blood mononuclear cells and tumor tissue of patients with hepatocellular carcinoma. Exp Ther Med 16: 788–796. Link: https://bit.ly/2HGLhtJ
  83. Zhang Y, Morgan R, Chen C, Cai Y, Clark E, et al. (2016) Mammary-tumor-educated B cells acquire LAP/TGF-β and PD-L1 expression and suppress anti-tumor immune responses. Int Immunol 28: 423-433. Link: https://bit.ly/35IsPZx
  84. Gabriely G, da Cunha AP, Rezende RM, Kenyon B, Madi A, et al. (2017) Targeting latency-associated peptide promotes antitumor immunity. Sci Immunol 2. Link: https://bit.ly/3oyeEz4
  85. Elkord E, Al Samid MA, Chaudhary B (2015) Helios, and not FoxP3, is the marker of activated Tregs expressing GARP/LAP. Oncotarget 6: 20026–20036. Link: https://bit.ly/3jzT8pX
  86. Hargadon K (2016) Dysregulation of TGFβ1 Activity in Cancer and Its Influence on the Quality of Anti-Tumor Immunity. J Clin Med 5: 76. Link: https://bit.ly/3juv3kg
  87. Kopsiaftis S, Rao PE, Burton R, English JM, Fox BS, et al. (2019) Abstract 2794: Expression of LAP, latency-associated peptide of TGFb, on immune cell subsets. Cancer Res 79: 2794-2794. Link: https://bit.ly/3jEuFzI
  88. Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, et al. (2017) Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell 168: 487-502.e15. Link: https://bit.ly/2HJyLtx
© 2020 Nyaribari MC. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
 

Help ?