Citation: Andrew V. Nguyen, Yuan-Yuan Wu, Elaine Y. Lin. The regulatory function of sphingosine-1-phosphate signaling axis on regulatory T cells in colorectal cancer[J]. AIMS Molecular Science, 2015, 1(1): 34-47. doi: 10.3934/molsci.2015.1.34
[1] | Kenji Chiba, Yasuhiro Maeda, Noriyasu Seki, Hirotoshi Kataoka, Kunio Sugahara . Role of sphingosine 1-phosphate (S1P) and effects of fingolimod, an S1P receptor 1 functional antagonist in lymphocyte circulation and autoimmune diseases. AIMS Molecular Science, 2014, 1(4): 162-182. doi: 10.3934/molsci.2014.4.162 |
[2] | Vera Reitsema, Hjalmar Bouma, Jan Willem Kok . Sphingosine-1-phosphate transport and its role in immunology. AIMS Molecular Science, 2014, 1(4): 183-201. doi: 10.3934/molsci.2014.4.183 |
[3] | Kannan Mayuri, Sundaram Vickram, Thirunavukarasou Anand, Konda Mani Saravanan . MicroRNA-mediated regulation of BCL-2 in breast cancer. AIMS Molecular Science, 2025, 12(1): 32-48. doi: 10.3934/molsci.2025003 |
[4] | Aliya I Sani, Zil-e-Rubab, Shumaila Usman, Syed Zaryab Ahmed, Mervyn Hosein . Role of OX40 and its ligand as costimulatory modulators in cancer immunotherapy. AIMS Molecular Science, 2021, 8(3): 161-173. doi: 10.3934/molsci.2021012 |
[5] | Sujit Nair . Current insights into the molecular systems pharmacology of lncRNA-miRNA regulatory interactions and implications in cancer translational medicine. AIMS Molecular Science, 2016, 3(2): 104-124. doi: 10.3934/molsci.2016.2.104 |
[6] | Simone Leggeri, Navid Sobhani . Single nucleotide polymorphisms Rs1045642 C>T genetic alteration in ATP Binding Cassette Subfamily B Member 1 role in increasing everolimus toxicity in metastatic breast cancer. AIMS Molecular Science, 2020, 7(1): 1-11. doi: 10.3934/molsci.2020001 |
[7] | Jade Sternberg, Miriam Wankell, V. Nathan Subramaniam, Lionel W. Hebbard . The functional roles of T-cadherin in mammalian biology. AIMS Molecular Science, 2017, 4(1): 62-81. doi: 10.3934/molsci.2017.1.62 |
[8] | Jin-Yih Low, Helen D. Nicholson . The up-stream regulation of polymerase-1 and transcript release factor(PTRF/Cavin-1) in prostate cancer: an epigenetic analysis. AIMS Molecular Science, 2016, 3(3): 466-478. doi: 10.3934/molsci.2016.3.466 |
[9] | Amena W. Smith, Swapan K. Ray, Arabinda Das, Kenkichi Nozaki, Baerbel Rohrer, Naren L. Banik . Calpain inhibition as a possible new therapeutic target in multiple sclerosis. AIMS Molecular Science, 2017, 4(4): 446-462. doi: 10.3934/molsci.2017.4.446 |
[10] | Hannah P. Priyanka, Rahul S. Nair, Sanjana Kumaraguru, Kirtikesav Saravanaraj, Vasantharekha Ramasamy . Insights on neuroendocrine regulation of immune mediators in female reproductive aging and cancer. AIMS Molecular Science, 2021, 8(2): 127-148. doi: 10.3934/molsci.2021010 |
Regulatory T cells play a key role in maintaining the balance between healthand disease and, in so doing, provide a system of checks and balances by: restraining asthma and allergy, preventing cytotoxic lymphocytes from reacting to self-antigens, and controlling inflammatory response. There are two main types of regulatory T cells; natural Tregs and adaptive/induced Tregs, which function to suppress the immune reaction [1]. Most natural Tregs are found developed in the thymus but they can also be induced in the periphery or in culture [2,3,4]. The natural Treg subset constitutes approximately 5-10% of resident CD4+ T cells and are involved mainly in self-tolerance while adaptive/induced Tregs are important in oral tolerance and inflammation [5,6]. The seminal work by Sakaguchi et al. in 1995 [7] using CD25+ depleted thymic T cells to induce autoimmune disease in nude mice demonstrated that thymic derived CD25+ T cells were the key cells mediating self-tolerance. CD25 is an alpha chain of IL-2 receptor that is expressed in most Tregs; however, a subset of Tregs, such as regulatory T cells 1 (Tr1) may express low levels of CD25 [8]. Signaling from CD25 is required for Treg survival and function [7,9,10]. While CD25 is often used as marker of Tregs, other surface markers such as, CD38, CD62L (L-selectin) and CD103 are sometimes used to identify different subsets of Tregs [11,12,13].
With the discovery of a transcriptional factor Forkhead-box P3 (FoxP3) as a specific Treg marker about a decade ago, the research field in Tregs has exploded. The general idea that FoxP3 regulates immuno-suppressive function of Tregs was formulated after extensive analysis of the scurfy mice [14] and patients with IPEX (Immunodesregulation, Polyendocrinopathy, Enteropathy, X-linked syndrome) [15,16] which confirmed that a loss of FoxP3 as a causative factor of multi-organ autoimmune disease. FoxP3 was subsequently shown to control the development and suppressive function of Tregs [17,18]. In the developing thymocytes, T-cell receptor (TCR)engagement with the peptide-MHCII [19,20,21] induces the expression of FoxP3 [22], suggesting that Tregs have self-antigen specificity consistent with the data by Gershon and Kondo [23], showing that adoptively transferred antigen-exposed T cells retain antigen-specific tolerance in naïve mice. FoxP3 forms a complex with nuclear factor of activated T cells (NF-AT) and inhibits the expression of IL-2, IL-4 and Interferon-γ (IFN-γ) cytokines which are important for adaptive immunity [24,25]. The FoxP3/NF-AT complex also upregulates the expression of CD25, cytotoxic T lymphocyte antigen 4 (CTLA4 also known as CD152) and glucocorticoid-induced TNF receptor family-related gene (GITR)in Tregs [24,25]. GITR signaling is necessary for Treg suppressive activity [26]. In conjunction with cytokines, such as IL-10 and transforming growth factor (TGF-β), FoxP3+ Tregs dampen effective cell activity by direct contact via CTLA4, which interacts with CD80 (B7-1) and CD86 (B7-2) on the target cells and inhibits their cellular function [27,28,29]. With CTLA4, Tregs can form an aggregate around DC thereby blocking the ability of DC to interact with effective cells and activation of adaptive immunity [30]. Tregs can also induce DC to produce indoleamine 2,3-dioxygenase (IDO), an important enzyme required for peripheral tolerance [31]. In short, FoxP3 is a critical intracellular molecule that governs the development and functions of Tregs.
Adaptive/induced Tregs which are generated in the periphery or in culture upon antigen encounter may or may not, however, express FoxP3 [32]. It appears that a suboptimal antigenic stimulation is required as well as signaling from TGF-β to drive the development of adaptive/induced Tregs from conventional CD4+ or CD8+ T cells [4,33,34] (Figure 1). In the gastrointestinal tract, adaptive/induced Tregs are necessary to maintain oral tolerance to enteric flora and food antigens, thus preventing pathological inflammation [6]. FoxP3+ Tregs specific to oral antigens can be induced from mesenteric or enteric lamina-propria CD4+ T cells by gut associated CD103+ dendritic cells [35]. Interestingly, FoxP3+ expression in adaptive/induced Tregs is less stable than in natural Tregs and depends on TGF-β to maintain FoxP3 expression [36].
Two subtypes of T cells, Tr1 and T helper 3 (Th3), are also known to regulate the development and function of FoxP3+ Tregs. These cells are also considered as regulatory T cells since Tr1 can secrete IL10 [8] and Th3 can secrete TGF-β [37] (Figure 1). While secretory products, IL10 and TGF-β are known for their suppressive functions of mainly effective cell [8,37,38], these cytokines also regulate local FoxP3+ Treg development. Indeed, high CD25+ FoxP3+ Tregs were observed in the periphery of transgenic mice engineered to ectopically express TGF-β in Th3 cells [39]. Tr1 and Th3 differ from adaptive/induced Tregs or natural Tregs in that their suppressive function may be driven by antigen-independent signals [40,41,42]. An interesting note is that colonic mesenchymal fibroblasts from patients who underwent colectomy for colon cancer or isolated from IBD patients can stimulate the development of CD4+CD25+FoxP3+ Tregs [43]. Collectively, these data support the idea that adaptive/induced Tregs can be generated in the periphery by local concentration of cytokines or from stimulation by resident cells or/ and recruited immune cells.
Colorectal cancer is the second leading cause of cancer death in the United States [44]. Approximately 15% of all colorectal cancers are related to one of the two inherited forms: Familial Adenomatous Polyposis (FAP), which is caused by a mutation in the adenomatous polyposis coli (APC) gene and Hereditary Nonpolyposis Colorectal Cancer (HNPCC) which is caused by mutation in any of the DNA mismatch repair protein such as: the msh2 gene, Mut homolog 1, mlh1 gene, or hMSH2 gene [45,46,47,48,49]. Other sporadic colorectal carcinomas have been linked to mutations in p53 genes, K-ras, SMAD4, 18q21 allelic loss associated with or without chromosomal or microsatellite instabilities [50]. Numerous studies have indicated that inheritance of the mutated APC gene is not sufficient to cause colorectal cancer; additional genetic alterations or environmental changes are required for tumor formation [51]. In a murine model of APC mutation known as Min mice, the development of polyps, a precursor to CRC development in these mice, is influenced by a mutation in the Mom1 (modifier of Min)gene. Mom1 encodes for a secreted phospholipase, A2, which catalyzes the formation of arachidonic acid, an important mediator of inflammation. Indeed, Min mice with mutated Mom1 gene were shown to have lower polyp and tumor numbers [52,53]. These data suggest that the microenvironment affects polyp development and may genetically and epigenetically alter tumor cell growth in inflamed/immune environments [54]. Collectively, these data established that multiple mutations are necessary for colorectal cancer development, and that inflammation is an important environmental factor modifying gene mutations and tumorigenesis.
The function of FoxP3+ Tregs in colorectal cancer is paradoxical. On one hand, a high density of FoxP3+ Tregs in tumors is associated with a good prognosis [55,56]. As a strong correlation has been shown between inflammation and CRC outcomes [57,58] and when Treg function is to suppress the inflammatory response, CRC tumorigenesis develops slower and the prognosis is most likely be more favorable. However, other studies have shown that FoxP3+ Tregs favor colon tumor growth [59,60,61] consistent with reports in other tumors such as breast cancer and hepatocellular carcinoma where increased numbers of Tregs correlate with reduced overall survival [62,63,64,65]. This is particularly true if Treg function is to suppress effective T cells involved in immuno-surveillance. Tregs may promote tumorigenesis by suppressing the Th1 antitumorigenic response or by interfering with the inhibitory function of cytotoxic T cells [56,66]. An important distinction should be noted, however, between CRC and other epithelial cancers. In many solid tumors, infiltrating T lymphocytes have specificity for tumor-specific antigens or self-antigens that allow them to destroy cancer cells [67,68]. In the case of CRC, in addition to cytotoxic CD8+ T with specificity to tumor antigens, there are many inflammatory T cells with specificity to the commensal miroflora [69,70] which could affect the functions of Tregs and cytotoxic T lymphocytes. These data add to the complexity of our Understanding of the roles of Tregs in CRC.
As such, direct analysis of Tregs in tumors may not be a reliable biomarker for prognosis of CRC [71,72,73,74]. Instead, others have examined the ratio of CD3+ or CD4+ to FoxP3+ Tregs and have determined that this may be a better predictor of clinical outcome in patients with colon carcinoma [75,76,77]. Yet, there are still many facets to Tregs, CRC, and tumor progression that have not been explored in detail. For example, how significant is the ratio of CD3+/FoxP3+ in the stromal vs. tumor region or the ratio of CD8+/FoxP3+ in the surrounding regions of the tumor as predictive values of clinical outcomes? What is the predictive value of the number of FoxP3+ Tregs in association with metastasis? What is the relationship between Tregs and chemotherapy? What are the densities of FoxP3+ and disease outcomes in other types of CRCs such as: DNA mismatch repair (MMR), PIK3CA mutation, and sporadic CRC? Lastly, what specific antigens do CRC-associated Tregs have? These questions need to be addressed further to determine the function of Tregs in CRC in human.
Recent identification of a subtype of Tregs in a mouse model and in some human colon cancer helps to explain the paradoxical functions of these cells [78]. One differentiating factor between distinctive Tregs is the expression of RAR-related orphan receptors (ROR)-γt, which have previously been shown to be vital for Th17 differentiation [79] (Figure 1). Th17 cells were shown to preferentially express IL17, IL-17F and IL-22, and induce inflammation [79]. Using APCΔ468 mice containing a mutation leading to a loss of APC function that develops benign adenomatous polyps, Blatner et al. [78] observed a significant number of CD4+, ROR-gt+, FoxP3+ Tregs surrounding the polyps consistent with their analysis of biopsied samples isolated from colon cancer patients. Interestingly, these ROR-γt+ FoxP3+ Tregs share many features similar to Th17 cells, suggesting that these cells may be derived from either 1)Th17 cells or 2)induced/activated Tregs responding to a polyp-environment rich in cytokines (IL-1β, IL-6 and IL-23)or 3)natural Treg homing into the colonic mucosal layer (Figure 1). Nevertheless, targeted deletion of ROR-γt in Tregs of APCΔ468 mice prevents the development of polyposis and increases mouse antitumor immunity suggesting that these pathologic ROR-γt+ Tregs actively contribute to polyp and tumor growth. It remains necessary to examine what triggers the switch in these cells to become ROR-gt+ Tregs. Dissecting the mechanisms of this transition state could have implications in future immunotherapy targeting Tregs.
While many Tregs are observed in CRC [75,77,80], little is known about the mechanism regulating their entry into CRC, though cytokine-mediated regulation of Tregs in inflammation has been suggested [81]. Recently, several chemokine-signaling axes have been shown to mediate Treg recruitment to inflamed sites and tumors [82,83]. In addition to G protein coupled receptor (GPCR) chemokine receptors, sphingosine-1 phosphate receptors (S1P1-S1P5) are also important regulators of immune cells including Tregs [84,85,86]. Of the five S1P signaling receptors, signaling from S1P1 is most studied and has been shown to regulate immune cell migration and activation of specific T cell subsets [87,88]. S1P, the agonist of these receptors, is a sphingolipid that is formed from sphingosine, a product of ceramide degradation, by two known sphingosine kinases SphK1 and SphK2 [89,90,91]. S1P and S1P1 regulating the differentiation of Th2, Tregs, Th17 and to some extent Th1 cells have been shown [88]. Moreover, experiments using transgenic mice carrying CD4+ T cells overexpressing S1P1 provides a first clue that this pathway may regulate Treg recruitment in extrathymic tissues [92]. The S1P1 transgenic mice expressed high level of IL-4, an important cytokine required for the selective development of CD4+ CD25+ Tregs [86]; thus, high numbers of CD4+CD25+FoxP3+ Tregs were observed in S1P1 transgenic compared to wild type mice [86].
More recentstudies, have shown that S1P1 signaling restrains thymic Treg development, affects their peripheral numbers, and inhibits Treg suppressive functions by interfering with the function of TGF-β [87,93]. Using transgenic mice with Treg overexpression of S1P1 signaling or transgenic mice with S1P1 ablation in T cells, Liu et al. demonstrated S1P1 signaling controls T cell lineage specification [94]. A reduced number of Tregs were observed in the lymphoid tissues and in the colon of mice with T cells over expressing S1P1 while Th1 subset was increased in the same mice [94]. Interestingly, another group had reported that there was an increase of Tregs in the periphery of S1P1-transgenic mice [92]. The discrepancy between these data may be due to the pleiotropic function of S1P which is known to orchestrate many intracellular and extracellular physiological and pathophysiological processes including: cell proliferation, migration and immune regulations [95,96,97,98]. Extracellularly, S1P functions as chemotactic agent, thus, Tregs with overexpression of S1P1 signaling may exhibit an increase in their homing mechanism. At the same time, S1P may function intracellularly to block the differentiation of Tregs [94].
In the context of CRC, we have recently shown that S1P and S1P1 receptor are also important mediators of cancer progression in a conditional knock-out Stat3 mouse model of colitis associated colorectal cancer [99]. Our paper along with a few other publications demonstrated that the S1P pathway mediated a dynamic loop between tumor cells and recruited immune cells. S1P secreted by tumor cells recruited immune cells whereby those cells secreted interleukin-6 (IL-6) which in turn stimulated tumor cells via IL-6 receptor (GP130) and STAT3 to make more S1P. Thus, S1P maintains the IL-6/STAT3/Sphingosine-kinase signaling in tumor cells [99,100,101,102] (Figure 2). We demonstrated that blocking this signaling pathway inhibits tumor progression [99]. However, whether S1P1 regulates Treg development or their function in the inflamed environment remains to be determined. We observed a reciprocal relationship between S1P signaling (tumor progression) and the Treg population [99]. Interestingly, in a recent study published by Liang et al. [100] using SphK2−/− mice (one of the two kinases responsible for making S1P)in a colitis associated colon cancer model, SphK2−/− mice, exhibited severe inflammation and developed larger tumors in greater numbers after AOM/DSS treatment. The phenotype was shown to be due to a compensatory mechanism of upregulation of SphK1 after the loss of SphK2. The group also used FTY720 (Fingolimod), a prodrug that is converted to the active FTY720-P form, to down regulate S1P1 signaling and demonstrated that FTY720 alleviates colitis reducing tumor size and number [100]. While the group did not report on the effect of FTY720 on Treg development, others have shown that FTY720 induces activation of CD4+CD25+ Tregs [94,103]. These findings suggest that modulation of S1P1/IL6/STAT3 signaling in transformed/cancer cells or in immune cells, including Tregs, may affect the tumor microenvironment and thereby alter CRC tumor progression.
Among many immune cells recruited to the CRC tumor site, Tregs appear to orchestrate the immune reaction that affects clinical outcomes. In light of Blatner’s data [78], the identification of ROR-gt+ Tregs provides a new immnunotherapy targeting Tregs. An important consideration should be noted in regards to the time of Treg recruitment during tumor development. Tregs which are ROR-γt- tend to arrive at tumor site very early during the inflammation [104]. These Tregs have specificity to the non-self-antigen and would most likely function in controlling inflammation [35]. High number of ROR-γt- Tregs, especially during early stage of tumor development, may indicate a good prognosis. This hypothesis is consistent with the findings that treatment targeted inflammatory response in CRC would have a positive clinical outcome [105]. As such, in a recent retrospective study of over 900 patients, those with colorectal cancer on a regular regimen of aspirin were significantly likely to have reduced mortality rates [105]. This study demonstrates the crucial role of inflammation, especially the interaction between inflammation and tumor progression, and the benefit of controlling some CRCs at the early stage with anti inflammatory drugs.
As a tumor progresses, ROR-γt+ Tregs were shown to be pro-tumorigenic since they suppress the cytotoxic T cells. Drugs targeting the ROR receptor to interfere with the function of ROR-γt+ Tregs would most likely alter disease progression. Along this line, drugs that target the S1P pathway to prevent Tregs as well as other lymphocyte egression from the lymph nodes are especially important. In our recent paper, we demonstrated that the S1P pathway is associated with IL-6 signaling pathway which maintains the persistently activated STAT3 and sphingosine kinases in tumor cells. The cycle between S1P, immune cell recruitment, IL-6, activated STAT3 and sphingosine kinases and S1P is required for tumor progression and has been demonstrated by other groups [100,101]. Interestingly, we observed that the epithelial STAT3 status in tumor cells correlated with the reciprocal Treg population in the tumor environment of colitis associated CRC; while conditional knock-out mice of epithelial STAT3 showed a significantly higher CD4+CD25+ Tregs [99]. We advocate the usage of FTY720 as well as other classes of drugs to perturb the S1P metabolism in treatment of late stage CRC [106].
Understanding the mechanisms by which Tregs mediated immune suppression would help in designing more appropriate drugs to inhibit Treg activity. As mentioned previously, Tregs mediate the suppression of effective T cells by direct contact via CTLA4. Antibodies targeting the interaction between CTLA4 and CD80 have been developed to inhibit the suppression function of Tregs on cytotoxic T cells; thereby allowing those cells to destroy tumors [107,108,109]. In an animal model of colitis using adoptively transferred of CD4+CD25− T cells into SCID mice infected with the protozoan parasite Leishmania, transferring CD4+CD25+ T cells can reverse colitis. The effect of CD4+CD25+ was blocked if anti-CTLA4 antibody was used [110]. These data suggest that antibody against CTLA4 can block the function of Tregs. Antibodies against CD25 targeting Tregs have also been used to improve immunotherapy responses in cancer [111,112]. In short, different modalities targeting Tregs in CRC should be considered as an additional strategy to the current treatment options.
Since the discovery of FoxP3+, many subsets of regulatory T cells have been identified. Their main function is to suppress the immune system thus preventing the innate and adaptive immune responses from going awry. Functions of Tregs in CRC are beginning to unfold with the identification of non-pathological and pathological Tregs (ROR-γt+). Understanding the development of these cells is pivotal in CRC treatment. Several mechanisms have been proposed for their presence in CRC: 1) they could represent pre-existing Tregs 2) arise from CD4+ effective cells and 3) be recruited from natural Tregs. The general consensus of pathological Tregs in CRC is pro-tumorigenic since they suppress immunosurveillence and antitumor immunity. As such, regulation of these cellular functions with antibodies against CTLA-4 antibodies would provide an attractive alternative to CRC treatment. Another strategy would be to block lymphocytes recruitment to mucosal layer of the colon by FTY720, prodrug of FTY720-P that binds to S1P1. Prolonged exposure of the cells to FTY720-P prevents the recycling of S1P1 to the surface after internalization. Hence, S1P signaling is blocked in the presence of FTY720 and FTY720-P. S1P pathway is unique in that it is not only involved in lymphocyte egression, but also that its intracellular signaling regulates the differentiation of CD4+ T cells into one type of immune cells that favor adaptive immunity, or another type (Tregs) that dampens it. The S1P pathway is also a critical component of the interaction between colon cancer cells and stromal cells. The IL-6/STAT3/SphK/S1P pathway promotes the persistent activation of STAT3 in tumors, which upregulates S1P to recruit immune cells which secrete IL-6. Thus, the development of Treg-centric therapeutics with S1P signaling inhibitor is likely to affect multiple cells and would add a novel and more effective therapeutic strategy against colorectal cancer especially those cancers associated with inflammation.
This work was supported in part by Professional Staff Congress of the City University of New York and Chancellor’s Research Fellowship to A.V.N.
All authors declare no conflicts of interest in this paper.
[1] |
Curotto de Lafaille MA, Lafaille JJ (2009) Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 30: 626-635. doi: 10.1016/j.immuni.2009.05.002
![]() |
[2] | Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6: 345-352. |
[3] |
Maggi E, Cosmi L, Liotta F, et al. (2005) Thymic regulatory T cells. Autoimmun Rev 4: 579-586. doi: 10.1016/j.autrev.2005.04.010
![]() |
[4] |
Mougiakakos D, Choudhury A, Lladser A, et al. (2010) Regulatory T cells in cancer. Adv Cancer Res 107: 57-117. doi: 10.1016/S0065-230X(10)07003-X
![]() |
[5] |
Jonuleit H, Schmitt E (2003) The regulatory T cell family: distinct subsets and their interrelations. J Immunol 171: 6323-6327. doi: 10.4049/jimmunol.171.12.6323
![]() |
[6] |
Mucida D, Kutchukhidze N, Erazo A, et al. (2005) Oral tolerance in the absence of naturally occurring Tregs. J Clini Invest 115: 1923-1933. doi: 10.1172/JCI24487
![]() |
[7] | Sakaguchi S, Sakaguchi N, Asano M, et al. (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155: 1151-1164. |
[8] |
Roncarolo MG, Gregori S, Battaglia M, et al. (2006) Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev 212: 28-50. doi: 10.1111/j.0105-2896.2006.00420.x
![]() |
[9] |
Setoguchi R, Hori S, Takahashi T, et al. (2005) Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med 201: 723-735. doi: 10.1084/jem.20041982
![]() |
[10] |
Barron L, Dooms H, Hoyer KK, et al. (2010) Cutting edge: mechanisms of IL-2-dependent maintenance of functional regulatory T cells. J Immunol 185: 6426-6430. doi: 10.4049/jimmunol.0903940
![]() |
[11] |
Read S, Mauze S, Asseman C, et al. (1998) CD38+ CD45RB(low) CD4+ T cells: a population of T cells with immune regulatory activities in vitro. Eur J Immunol 28: 3435-3447. doi: 10.1002/(SICI)1521-4141(199811)28:11<3435::AID-IMMU3435>3.0.CO;2-P
![]() |
[12] |
Fu S, Yopp AC, Mao X, et al. (2004) CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential. Am J Transplant 4: 65-78. doi: 10.1046/j.1600-6143.2003.00293.x
![]() |
[13] |
Allakhverdi Z, Fitzpatrick D, Boisvert A, et al. (2006) Expression of CD103 identifies human regulatory T-cell subsets. J Allergy Clin Immunol 118: 1342-1349. doi: 10.1016/j.jaci.2006.07.034
![]() |
[14] | Brunkow ME, Jeffery EW, Hjerrild KA, et al. (2001) Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27: 68-73. |
[15] |
Chatila TA, Blaeser F, Ho N, et al. (2000) JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clini Invest 106: R75-81. doi: 10.1172/JCI11679
![]() |
[16] |
Bennett CL, Christie J, Ramsdell F, et al. (2001) The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 27: 20-21. doi: 10.1038/83713
![]() |
[17] |
Allan SE, Passerini L, Bacchetta R, et al. (2005) The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clini Invest 115: 3276-3284. doi: 10.1172/JCI24685
![]() |
[18] |
Tang Q, Bluestone JA (2008) The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol 9: 239-244. doi: 10.1038/ni1572
![]() |
[19] |
LeGuern C (2003) Regulation of T-cell functions by MHC class II self-presentation. Trends Immunol 24: 633-638. doi: 10.1016/j.it.2003.10.010
![]() |
[20] | Coutinho A, Caramalho I, Seixas E, et al. (2005) Thymic commitment of regulatory T cells is a pathway of TCR-dependent selection that isolates repertoires undergoing positive or negative selection. Curr Top Microbiol 293: 43-71. |
[21] |
Burchill MA, Yang J, Vang KB, et al. (2008) Linked T cell receptor and cytokine signaling govern the development of the regulatory T cell repertoire. Immunity 28: 112-121. doi: 10.1016/j.immuni.2007.11.022
![]() |
[22] |
Liston A, Nutsch KM, Farr AG, et al. (2008) Differentiation of regulatory Foxp3+ T cells in the thymic cortex. P Natl Acad Sci USA 105: 11903-11908. doi: 10.1073/pnas.0801506105
![]() |
[23] | Gershon RK, Kondo K (1970) Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology 18: 723-737. |
[24] |
Lopes JE, Torgerson TR, Schubert LA, et al. (2006) Analysis of FOXP3 reveals multiple domains required for its function as a transcriptional repressor. J Immunol 177: 3133-3142. doi: 10.4049/jimmunol.177.5.3133
![]() |
[25] |
Wu Y, Borde M, Heissmeyer V, et al. (2006) FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell 126: 375-387. doi: 10.1016/j.cell.2006.05.042
![]() |
[26] |
Shimizu J, Yamazaki S, Takahashi T, et al. (2002) Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol 3: 135-142. doi: 10.1038/ni759
![]() |
[27] |
Sakaguchi S (2004) Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol 22: 531-562. doi: 10.1146/annurev.immunol.21.120601.141122
![]() |
[28] |
Krummel MF, Allison JP (1995) CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 182: 459-465. doi: 10.1084/jem.182.2.459
![]() |
[29] |
Walunas TL, Lenschow DJ, Bakker CY, et al. (1994) CTLA-4 can function as a negative regulator of T cell activation. Immunity 1: 405-413. doi: 10.1016/1074-7613(94)90071-X
![]() |
[30] |
Onishi Y, Fehervari Z, Yamaguchi T, et al. (2008) Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. P Natl Acad Sci USA 105: 10113-10118. doi: 10.1073/pnas.0711106105
![]() |
[31] | Belladonna ML, Puccetti P, Orabona C, et al. (2007) Immunosuppression via tryptophan catabolism: the role of kynurenine pathway enzymes. Transplantation 84: S17-20. |
[32] |
Horwitz DA, Zheng SG, Gray JD (2008) Natural and TGF-beta-induced Foxp3(+)CD4(+) CD25(+) regulatory T cells are not mirror images of each other. Trends Immunol 29: 429-435. doi: 10.1016/j.it.2008.06.005
![]() |
[33] |
Apostolou I, von Boehmer H (2004) In vivo instruction of suppressor commitment in naive T cells. J Exp Med 199: 1401-1408. doi: 10.1084/jem.20040249
![]() |
[34] |
Kretschmer K, Apostolou I, Hawiger D, et al. (2005) Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol 6: 1219-1227. doi: 10.1038/ni1265
![]() |
[35] |
Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. (2007) A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 204: 1757-1764. doi: 10.1084/jem.20070590
![]() |
[36] |
Selvaraj RK, Geiger TL (2007) A kinetic and dynamic analysis of Foxp3 induced in T cells by TGF-beta. J Iimmunol 178: 7667-7677. doi: 10.4049/jimmunol.178.12.7667
![]() |
[37] |
Faria AM, Weiner HL (2006) Oral tolerance and TGF-beta-producing cells. Inflamm Allergy Drug Targets 5: 179-190. doi: 10.2174/187152806778256034
![]() |
[38] |
Weiner HL (2001) Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunol Rev 182: 207-214. doi: 10.1034/j.1600-065X.2001.1820117.x
![]() |
[39] | Carrier Y, Yuan J, Kuchroo VK, et al. (2007) Th3 cells in peripheral tolerance. II. TGF-beta-transgenic Th3 cells rescue IL-2-deficient mice from autoimmunity. J Immunol 178: 172-178. |
[40] |
Walker MR, Carson BD, Nepom GT, et al. (2005) De novo generation of antigen-specific CD4+CD25+ regulatory T cells from human CD4+CD25- cells. P Natl Acad Sci USA 102: 4103-4108. doi: 10.1073/pnas.0407691102
![]() |
[41] |
Wilczynski JR, Radwan M, Kalinka J (2008) The characterization and role of regulatory T cells in immune reactions. Front Biosci 13: 2266-2274. doi: 10.2741/2840
![]() |
[42] |
Mazza G, Sabatos-Peyton CA, Protheroe RE, et al. (2010) Isolation and characterization of human interleukin-10-secreting T cells from peripheral blood. Hum Immunol 71: 225-234. doi: 10.1016/j.humimm.2009.12.003
![]() |
[43] |
Pinchuk IV, Beswick EJ, Saada JI, et al. (2011) Human colonic myofibroblasts promote expansion of CD4+ CD25high Foxp3+ regulatory T cells. Gastroenterology 140: 2019-2030. doi: 10.1053/j.gastro.2011.02.059
![]() |
[44] | Wingo PA, Ries LA, Parker SL, et al. (1998) Long-term cancer patient survival in the United States. Cancer Epidemiol Biomarkers Prev 7: 271-282. |
[45] |
Cannon-Albright LA, Skolnick MH, Bishop DT, et al. (1988) Common inheritance of susceptibility to colonic adenomatous polyps and associated colorectal cancers. N Engl J Med 319: 533-537. doi: 10.1056/NEJM198809013190902
![]() |
[46] |
Peltomaki P, de la Chapelle A (1997) Mutations predisposing to hereditary nonpolyposis colorectal cancer. Adv Cancer Res 71: 93-119. doi: 10.1016/S0065-230X(08)60097-4
![]() |
[47] |
Hawkins NJ, Ward RL (2001) Sporadic colorectal cancers with microsatellite instability and their possible origin in hyperplastic polyps and serrated adenomas. J Natl Cancer I 93: 1307-1313. doi: 10.1093/jnci/93.17.1307
![]() |
[48] |
Fishel R, Lescoe MK, Rao MR, et al. (1993) The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 75: 1027-1038. doi: 10.1016/0092-8674(93)90546-3
![]() |
[49] |
Al-Tassan N, Chmiel NH, Maynard J, et al. (2002) Inherited variants of MYH associated with somatic G:C-->T:A mutations in colorectal tumors. Nat Genet 30: 227-232. doi: 10.1038/ng828
![]() |
[50] |
Grady WM, Carethers JM (2008) Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135: 1079-1099. doi: 10.1053/j.gastro.2008.07.076
![]() |
[51] |
Kinzler KW, Vogelstein B (1996) Lessons from hereditary colorectal cancer. Cell 87: 159-170. doi: 10.1016/S0092-8674(00)81333-1
![]() |
[52] |
MacPhee M, Chepenik KP, Liddell RA, et al. (1995) The secretory phospholipase A2 gene is a candidate for the Mom1 locus, a major modifier of ApcMin-induced intestinal neoplasia. Cell 81: 957-966. doi: 10.1016/0092-8674(95)90015-2
![]() |
[53] | Shoemaker AR, Gould KA, Luongo C, et al. (1997) Studies of neoplasia in the Min mouse. Biochimica et biophysica acta 1332: F25-48. |
[54] |
de Miranda NF, Hes FJ, van Wezel T, et al. (2012) Role of the microenvironment in the tumourigenesis of microsatellite unstable and MUTYH-associated polyposis colorectal cancers. Mutagenesis 27: 247-253. doi: 10.1093/mutage/ger077
![]() |
[55] |
Salama P, Phillips M, Grieu F, et al. (2009) Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 27: 186-192. doi: 10.1200/JCO.2008.18.7229
![]() |
[56] |
Ladoire S, Martin F, Ghiringhelli F (2011) Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother 60: 909-918. doi: 10.1007/s00262-011-1046-y
![]() |
[57] |
Boland CR (2010) Chronic inflammation, colorectal cancer and gene polymorphisms. Digestive diseases 28: 590-595. doi: 10.1159/000320053
![]() |
[58] | Danese S, Mantovani A Inflammatory bowel disease and intestinal cancer: a paradigm of the Yin-Yang interplay between inflammation and cancer. Oncogene 29: 3313-3323. |
[59] |
Chaput N, Louafi S, Bardier A, et al. (2009) Identification of CD8+CD25+Foxp3+ suppressive T cells in colorectal cancer tissue. Gut 58: 520-529. doi: 10.1136/gut.2008.158824
![]() |
[60] |
Brudvik KW, Henjum K, Aandahl EM, et al. (2012) Regulatory T-cell-mediated inhibition of antitumor immune responses is associated with clinical outcome in patients with liver metastasis from colorectal cancer. Cancer Immunol Immunother 61: 1045-1053. doi: 10.1007/s00262-011-1174-4
![]() |
[61] | Zeng JC, Zhang Z, Li TY, et al. (2013) Assessing the role of IL-35 in colorectal cancer progression and prognosis. Int J Clin Exp Pathol 6: 1806-1816. |
[62] |
Bates GJ, Fox SB, Han C, et al. (2006) Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol 24: 5373-5380. doi: 10.1200/JCO.2006.05.9584
![]() |
[63] |
Fu J, Xu D, Liu Z, et al. (2007) Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 132: 2328-2339. doi: 10.1053/j.gastro.2007.03.102
![]() |
[64] |
Betts G, Jones E, Junaid S, et al. (2012) Suppression of tumour-specific CD4(+) T cells by regulatory T cells is associated with progression of human colorectal cancer. Gut 61: 1163-1171. doi: 10.1136/gutjnl-2011-300970
![]() |
[65] | Bacic D, Uravic M, Bacic R, et al. (2011) Augmentation of regulatory T cells (CD4+CD25+Foxp3+) correlates with tumor stage in patients with colorectal cancer. Coll Antropol 35 Suppl 2: 65-68. |
[66] |
Mempel TR, Pittet MJ, Khazaie K, et al. (2006) Regulatory T cells reversibly suppress cytotoxic T cell function independent of effector differentiation. Immunity 25: 129-141. doi: 10.1016/j.immuni.2006.04.015
![]() |
[67] |
Slaney CY, Rautela J, Parker BS (2013) The emerging role of immunosurveillance in dictating metastatic spread in breast cancer. Cancer Res 73: 5852-5857. doi: 10.1158/0008-5472.CAN-13-1642
![]() |
[68] |
Koebel CM, Vermi W, Swann JB, et al. (2007) Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450: 903-907. doi: 10.1038/nature06309
![]() |
[69] | Bonertz A, Weitz J, Pietsch DH, et al. (2009) Antigen-specific Tregs control T cell responses against a limited repertoire of tumor antigens in patients with colorectal carcinoma. J Clini Invest 119: 3311-3321. |
[70] |
Duchmann R, May E, Heike M, et al. (1999) T cell specificity and cross reactivity towards enterobacteria, bacteroides, bifidobacterium, and antigens from resident intestinal flora in humans. Gut 44: 812-818. doi: 10.1136/gut.44.6.812
![]() |
[71] | Naito Y, Saito K, Shiiba K, et al. (1998) CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 58: 3491-3494. |
[72] |
Golby SJ, Chinyama C, Spencer J (2002) Proliferation of T-cell subsets that contact tumour cells in colorectal cancer. Clin Exp Immunol 127: 85-91. doi: 10.1046/j.1365-2249.2002.01730.x
![]() |
[73] |
Le Gouvello S, Bastuji-Garin S, Aloulou N, et al. (2008) High prevalence of Foxp3 and IL17 in MMR-proficient colorectal carcinomas. Gut 57: 772-779. doi: 10.1136/gut.2007.123794
![]() |
[74] |
Nosho K, Baba Y, Tanaka N, et al. (2010) Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol 222: 350-366. doi: 10.1002/path.2774
![]() |
[75] |
Gooden MJ, de Bock GH, Leffers N, et al. (2011) The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Brit J Cancer 105: 93-103. doi: 10.1038/bjc.2011.189
![]() |
[76] |
Sinicrope FA, Rego RL, Ansell SM, et al. (2009) Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology 137: 1270-1279. doi: 10.1053/j.gastro.2009.06.053
![]() |
[77] |
Katz SC, Bamboat ZM, Maker AV, et al. (2013) Regulatory T cell infiltration predicts outcome following resection of colorectal cancer liver metastases. Ann Surg Oncol 20: 946-955. doi: 10.1245/s10434-012-2668-9
![]() |
[78] | Blatner NR, Mulcahy MF, Dennis KL, et al. (2012) Expression of RORgammat marks a pathogenic regulatory T cell subset in human colon cancer. Sci Transl Med 4: 164ra159. |
[79] |
Awasthi A, Kuchroo VK (2009) Th17 cells: from precursors to players in inflammation and infection. Int Immunol 21: 489-498. doi: 10.1093/intimm/dxp021
![]() |
[80] | Liu Z, Huang Q, Liu G, et al. (2014) Presence of FOXP3(+)Treg cells is correlated with colorectal cancer progression. Int J Clin Exp Med 7: 1781-1785. |
[81] |
Sojka DK, Huang YH, Fowell DJ (2008) Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunology 124: 13-22. doi: 10.1111/j.1365-2567.2008.02813.x
![]() |
[82] |
Ishimaru N, Nitta T, Arakaki R, et al. (2010) In situ patrolling of regulatory T cells is essential for protecting autoimmune exocrinopathy. PloS One 5: e8588. doi: 10.1371/journal.pone.0008588
![]() |
[83] |
Akeus P, Langenes V, von Mentzer A, et al. (2014) Altered chemokine production and accumulation of regulatory T cells in intestinal adenomas of APC(Min/+) mice. Cancer Immunol Immunother 63: 807-819. doi: 10.1007/s00262-014-1555-6
![]() |
[84] |
Michaud J, Im DS, Hla T (2010) Inhibitory role of sphingosine 1-phosphate receptor 2 in macrophage recruitment during inflammation. J Immunol 184: 1475-1483. doi: 10.4049/jimmunol.0901586
![]() |
[85] |
Ishimaru N, Yamada A, Nitta T, et al. (2012) CCR7 with S1P1 signaling through AP-1 for migration of Foxp3+ regulatory T-cells controls autoimmune exocrinopathy. Am J Pathol 180: 199-208. doi: 10.1016/j.ajpath.2011.09.027
![]() |
[86] |
Goetzl EJ, Liao JJ, Huang MC (2008) Regulation of the roles of sphingosine 1-phosphate and its type 1 G protein-coupled receptor in T cell immunity and autoimmunity. Biochimica et biophysica acta 1781: 503-507. doi: 10.1016/j.bbalip.2008.03.001
![]() |
[87] | Wang W, Graeler MH, Goetzl EJ (2004) Physiological sphingosine 1-phosphate requirement for optimal activity of mouse CD4+ regulatory T Cells. FASEB J 18: 1043-1045. |
[88] |
Rosen H, Goetzl EJ (2005) Sphingosine 1-phosphate and its receptors: an autocrine and paracrine network. Nat Rev Immunol 5: 560-570. doi: 10.1038/nri1650
![]() |
[89] |
Pyne S, Pyne NJ (2011) Translational aspects of sphingosine 1-phosphate biology. Trends Mol Med 17: 463-472. doi: 10.1016/j.molmed.2011.03.002
![]() |
[90] |
Kohama T, Olivera A, Edsall L, et al. (1998) Molecular cloning and functional characterization of murine sphingosine kinase. J Biol Chem 273: 23722-23728. doi: 10.1074/jbc.273.37.23722
![]() |
[91] |
Liu H, Chakravarty D, Maceyka M, et al. (2002) Sphingosine kinases: a novel family of lipid kinases. Prog Nucleic Acid Res Mol Biol 71: 493-511. doi: 10.1016/S0079-6603(02)71049-0
![]() |
[92] |
Wang W, Huang MC, Goetzl EJ (2007) Type 1 sphingosine 1-phosphate G protein-coupled receptor (S1P1) mediation of enhanced IL-4 generation by CD4 T cells from S1P1 transgenic mice. J Immunol 178: 4885-4890. doi: 10.4049/jimmunol.178.8.4885
![]() |
[93] |
Sawicka E, Dubois G, Jarai G, et al. (2005) The sphingosine 1-phosphate receptor agonist FTY720 differentially affects the sequestration of CD4+/CD25+ T-regulatory cells and enhances their functional activity. J Immunol 175: 7973-7980. doi: 10.4049/jimmunol.175.12.7973
![]() |
[94] |
Liu G, Yang K, Burns S, et al. (2010) The S1P(1)-mTOR axis directs the reciprocal differentiation of T(H)1 and T(reg) cells. Nat Immunol 11: 1047-1056. doi: 10.1038/ni.1939
![]() |
[95] |
Snider AJ, Orr Gandy KA, Obeid LM (2010) Sphingosine kinase: Role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie 92: 707-715. doi: 10.1016/j.biochi.2010.02.008
![]() |
[96] |
Spiegel S, Milstien S (2011) The outs and the ins of sphingosine-1-phosphate in immunity. Nat Rev Immunol 11: 403-415. doi: 10.1038/nri2974
![]() |
[97] |
Maceyka M, Harikumar KB, Milstien S, et al. (2012) Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol 22: 50-60. doi: 10.1016/j.tcb.2011.09.003
![]() |
[98] |
Schwalm S, Pfeilschifter J, Huwiler A (2013) Sphingosine-1-phosphate: a Janus-faced mediator of fibrotic diseases. Biochim Biophys Acta 1831: 239-250. doi: 10.1016/j.bbalip.2012.07.022
![]() |
[99] |
Nguyen AV, Wu YY, Liu Q, et al. (2013) STAT3 in Epithelial Cells Regulates Inflammation and Tumor Progression to Malignant State in Colon. Neoplasia 15: 998-1008. doi: 10.1593/neo.13952
![]() |
[100] |
Liang J, Nagahashi M, Kim EY, et al. (2013) Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer. Cancer Cell 23: 107-120. doi: 10.1016/j.ccr.2012.11.013
![]() |
[101] |
Lee H, Herrmann A, Deng JH, et al. (2009) Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell 15: 283-293. doi: 10.1016/j.ccr.2009.02.015
![]() |
[102] |
Nagahashi M, Hait NC, Maceyka M, et al. (2014) Sphingosine-1-phosphate in chronic intestinal inflammation and cancer. Adv Biol Regul 54: 112-120. doi: 10.1016/j.jbior.2013.10.001
![]() |
[103] |
Daniel C, Sartory N, Zahn N, et al. (2007) FTY720 ameliorates Th1-mediated colitis in mice by directly affecting the functional activity of CD4+CD25+ regulatory T cells. J Immunol 178: 2458-2468. doi: 10.4049/jimmunol.178.4.2458
![]() |
[104] | Bos PD, Rudensky AY (2012) Treg cells in cancer: a case of multiple personality disorder. Sci Transl Med 4: 164fs144. |
[105] | Liao X, Lochhead P, Nishihara R, et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med 367: 1596-1606. |
[106] |
Nguyen AV, Wu YY, Lin EY (2014) STAT3 and sphingosine-1-phosphate in inflammation-associated colorectal cancer. World J Gastroenterol 20: 10279-10287. doi: 10.3748/wjg.v20.i30.10279
![]() |
[107] |
Khan S, Burt DJ, Ralph C, et al. (2011) Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells. Clin Immunol 138: 85-96. doi: 10.1016/j.clim.2010.09.011
![]() |
[108] |
Saha A, Chatterjee SK (2010) Combination of CTL-associated antigen-4 blockade and depletion of CD25 regulatory T cells enhance tumour immunity of dendritic cell-based vaccine in a mouse model of colon cancer. Scand J Immunol 71: 70-82. doi: 10.1111/j.1365-3083.2009.02355.x
![]() |
[109] |
Yano H, Thakur A, Tomaszewski EN, et al. (2014) Ipilimumab augments antitumor activity of bispecific antibody-armed T cells. J Transl Med 12: 191. doi: 10.1186/1479-5876-12-191
![]() |
[110] |
Liu H, Hu B, Xu D, et al. (2003) CD4+CD25+ regulatory T cells cure murine colitis: the role of IL-10, TGF-beta, and CTLA4. J Immunol 171: 5012-5017. doi: 10.4049/jimmunol.171.10.5012
![]() |
[111] | von Boehmer H, Daniel C (2013) Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer. Nat Rev Drug Discov 12: 51-63. |
[112] | Rech AJ, Mick R, Martin S, et al. (2012) CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med 4: 134ra162. |