Regulatory T cell

Source: Wikipedia, the free encyclopedia.
(Redirected from
Regulatory T cells
)

The regulatory T cells (Tregs

CD25 and are thought to be derived from the same lineage as naïve CD4+ cells.[2] Because effector T cells also express CD4 and CD25, Treg cells are very difficult to effectively discern from effector CD4+, making them difficult to study. Research has found that the cytokine transforming growth factor beta (TGF-β) is essential for Treg cells to differentiate from naïve CD4+ cells and is important in maintaining Treg cell homeostasis.[3]

Mouse models have suggested that modulation of Treg cells can treat autoimmune disease and cancer and can facilitate organ transplantation[4] and wound healing.[5] Their implications for cancer are complicated. Treg cells tend to be upregulated in individuals with cancer, and they seem to be recruited to the site of many tumors. Studies in both humans and animal models have implicated that high numbers of Treg cells in the tumor microenvironment is indicative of a poor prognosis, and Treg cells are thought to suppress tumor immunity, thus hindering the body's innate ability to control the growth of cancerous cells.[6] Immunotherapy research is studying how regulation of T cells could possibly be utilized in the treatment of cancer.[7]

Populations

T regulatory cells are a component of the immune system that suppress immune responses of other cells. This is an important "self-check" built into the immune system to prevent excessive reactions. Regulatory T cells come in many forms with the most well-understood being those that express CD4, CD25, and FOXP3 (CD4+CD25+ regulatory T cells). These Treg cells are different from helper T cells.[8] Another regulatory T cell subset is Treg17 cells.[9] Regulatory T cells are involved in shutting down immune responses after they have successfully eliminated invading organisms, and also in preventing autoimmunity.[10]

CD4+ FOXP3+ CD25(high) regulatory T cells have been called "naturally occurring" regulatory T cells[11] to distinguish them from "suppressor" T cell populations that are generated in vitro. Additional regulatory T cell populations include Tr1, Th3, CD8+CD28, and Qa-1 restricted T cells. The contribution of these populations to self-tolerance and immune homeostasis is less well defined. FOXP3 can be used as a good marker for mouse CD4+CD25+ T cells, although recent studies have also shown evidence for FOXP3 expression in CD4+CD25 T cells. In humans, FOXP3 is also expressed by recently activated conventional T cells and thus does not specifically identify human Tregs.[12]

Development

All T cells derive from progenitor cells in the

TCR
expression of NKT or γδ T cells; Tregs have a larger TCR diversity than effector T cells, biased towards self-peptides.

The process of Treg selection is determined by the affinity of interaction with the self-peptide MHC complex. Selection to become a Treg is a "

Goldilocks" process - i.e. not too high, not too low, but just right;[13] a T cell that receives very strong signals will undergo apoptotic death; a cell that receives a weak signal will survive and be selected to become an effector cell. If a T cell receives an intermediate signal, then it will become a regulatory cell. Due to the stochastic
nature of the process of T cell activation, all T cell populations with a given TCR will end up with a mixture of Teff and Treg – the relative proportions determined by the affinities of the T cell for the self-peptide-MHC. Even in mouse models with TCR-transgenic cells selected on specific-antigen-secreting stroma, deletion or conversion is not complete.

After interaction with self-peptide MHC complex, T cell has to upregulate IL-2R, CD25 and TNFR superfamily members GITR, OX40 and TNFR2 to become CD25+FOXP3- Treg cell progenitor. To become mature Treg, FOXP3 transcription factor has to be upregulated which is driven by γ-chain (CD132) dependent cytokines, in particular IL-2 and/or IL-15.[14][15] Only IL-2 alone is not sufficient to stimulate Foxp3 expression, other cytokines are needed. Whereas IL-2 is produced by self-reactive thymocytes, IL-15 is produced by stromal cells of the thymus, mainly mTECs and cTECs.[14]

Recently, other subset of Treg precursors was identified. This subset lacks CD25 and has low expression of Foxp3. Its development is mainly dependent on IL-15. This subset has lower affinity for self antigens than CD25+Foxp3high subset.  Both subsets generate mature Treg cells after stimulation with IL-2 with comparable efficiency both in vitro and in vivo. CD25+Foxp3high progenitors exhibit increased apoptosis and develop into mature Treg cells with faster kinetics than Foxp3low progenitors.[16] Tregs derived from CD25+Foxp3high progenitors protect from experimental auto-immune encephalomyelitis, whereas those derived from CD25+Foxp3low progenitors protect from T-cell induced colitis.[14]

Mature CD25+Foxp3+ Tregs can be also divided into two different subsets based on the expression level of CD25, GITR, and PD-1. Tregs expressing low amounts of CD25, GITR and PD-1 limit the development of colitis by promoting the conversion of conventional CD4+ T cells into pTreg. Tregs highly expressing CD25, GITR and PD-1 are more self-reactive and control lymphoproliferation in peripheral lymph nodes - they may have the ability to protect against autoimmune disorders.[14]

Foxp3+ Treg generation in the thymus is delayed by several days compared to Teff cells and does not reach adult levels in either the thymus or periphery until around three weeks post-partum. Treg cells require

TGF-β
insensitive TGFβRII-DN mice are functional.

Thymic recirculation

There was observed, that some FOXP3+ Treg cells are recirculating back to thymus, where they have developed. This Treg were mainly present in thymic medulla, which is the main site of Treg cells differentiation.[17] The presence of this cells in thymus or addition into fetal thymic tissue culture suppress development of new Treg cells by 34–60%,[17] but Tconv cells are not affected. That means, that recirculating Treg to thymus inhibited just de novo development of Treg cells. Molecular mechanism of this process works due to the ability of Treg to adsorb IL-2 from the microenvironments, thus being able to induce apoptosis of other T cells which need IL-2 as main growth factor.

CD25) encoded by Il2ra gene which gather IL-2 from thymic medulla, and decrease its concentration. New generated FOXP3+ Treg cells in thymus have not so high amount of Il2ra expression.[17] IL-2 is a cytokine necessary for the development of Treg cells in the thymus. It is important for T cells proliferation and survival, but in the case of its deficiency, IL-15 may be replaced. However, Treg cells' development is dependent on IL-2.[19] In humans, there was found population of CD31 negative Treg cells in thymus.[17] CD31 could be used as a marker of new generated Treg cells as same as other T lymphocytes. Mature and peripheral Treg cells have decreased its expression.[20]
So it is possible that this regulatory mechanism of thymic Treg cells development is also functional in humans.

There is probably also positive regulation of thymic Treg cells development caused by recirculating Treg cells into thymus. There was found population of

TIR) Toll interleukin-1 receptor domain, which is normally present in innate immune cells.[23]

Function

The

autoimmune diseases. Regulatory T cells actively suppress activation of the immune system and prevent pathological self-reactivity, i.e. autoimmune disease. The critical role regulatory T cells play within the immune system is evidenced by the severe autoimmune syndrome that results from a genetic deficiency in regulatory T cells (IPEX syndrome
– see also below).

Diagram of regulatory T cell, effector T cells and dendritic cell showing putative mechanisms of suppression by regulatory T cells.

The molecular mechanism by which regulatory T cells exert their suppressor/regulatory activity has not been definitively characterized and is the subject of intense research. In vitro experiments have given mixed results regarding the requirement of cell-to-cell contact with the cell being suppressed. The following represent some of the proposed mechanisms of immune suppression:

Natural and induced regulatory T cells

T regulatory lymphocytes develop during ontogeny either in the thymus or in the periphery. Accordingly, they are divided into natural and induced T regulatory cells.[37]

Natural T regulatory lymphocytes (tTregs, nTregs) are characterized by continuous expression of FoxP3 and T cell receptor (TCR) with relatively high autoaffinity. These cells are predominantly found in the body in the bloodstream or lymph nodes and serve mainly to confer tolerance to autoantigens.[37]

Induced (peripheral) T regulatory cells (iTregs, pTregs) arise under certain situations in the presence of IL-2 and TGF-b in the periphery and begin to express FoxP3 inducibly, thus becoming the functional equivalent of tTreg cells. iTregs, however, are found primarily in peripheral barrier tissues, where they are primarily involved in preventing inflammation in the presence of external antigens.[37]

The main features that differentiate tTreg and iTreg cells include Helios and Neuropilin-1, the presence of which suggests origin in the thymus. Another feature distinguishing these two Treg cell populations is the stability of FoxP3 expression in different settings.[37]

Induced T regulatory cells

Induced regulatory T (iTreg) cells (CD4+ CD25+ FOXP3+) are suppressive cells involved in tolerance. iTreg cells have been shown to suppress T cell proliferation and experimental autoimmune diseases. These cells include Treg17 cells. iTreg cells develop from mature CD4+ conventional T cells outside of the thymus: a defining distinction between natural regulatory T (nTreg) cells and iTreg cells. Though iTreg and nTreg cells share a similar function iTreg cells have recently been shown to be "an essential non-redundant regulatory subset that supplements nTreg cells, in part by expanding TCR diversity within regulatory responses".[38] Acute depletion of the iTreg cell pool in mouse models has resulted in inflammation and weight loss. The contribution of nTreg cells versus iTreg cells in maintaining tolerance is unknown, but both are important. Epigenetic differences have been observed between nTreg and iTreg cells, with the former having more stable FOXP3 expression and wider demethylation.

The small intestinal environment is high in vitamin A and is a location where retinoic acid is produced.

CTLA4 and decreased methylation in the FOXP3 Treg-Specific demethylated region compared to TGF-β-induced Tregs.[45]

RORγt+ regulatory T lymphocytes

Approximately 30%–40% of colonic FoxP3+ Treg cells express the transcription factor RORγt.[49] The iTregs are able to differentiate into RORγt-expressing cells and thus acquire the phenotype of Th17 cells. These cells are associated with the functions of mucosal lymphoid tissues such as the intestinal barrier. In the intestinal lamina propria, 20-30% of Foxp3+ T regulatory cells expressing RORyt are found and this high proportion is strongly dependent on the presence of a complex gut microbiome. In germ-free (GF) mice, the population of RORγt+ T regulatory cells is strongly reduced, whereas recolonization by the specific pathogen-free (SPF) microbiota restores normal numbers of these lymphocytes in the gut. The mechanism by which the gut microbiota induces the formation of RORγt+ Treg cells involves the production of short-chain fatty acids (SCFAs), on which this induction is dependent. SCFAs are a by-product of fermentation and digestion of dietary fiber, therefore, microbial-free mice have very low concentrations of both SCFAs and RORγt Treg cells. Induction of RORγt Treg cells is also dependent on the presence of dendritic cells in adults, Thetis cells in neonatal and antigen presentation by MHC II.[50][51]

RORγt+ Treg cells are not present in the thymus and do not express Helios or

CTLA-4, and the nucleotidases CD39 and CD73, suggesting a strong regulatory function.[50]

Function of RORγt+ regulatory T lymphocytes

Induction of RORγt+ Treg cells in lymph nodes of the small intestine is crucial for the establishment of intestinal luminal antigen tolerance. These cells are particularly important in the prevention of food allergies. One mechanism is the production of suppressive molecules such as the cytokine IL-10. These cells also suppress the Th17 cell population and inhibit the production of IL-17, thus suppressing the pro-inflammatory response.[50]

In mice, colonic RORγt+ Tregs are absent during the first two weeks after birth. Generation of RORγt+ Treg early after birth is essential to prevent the development of various intestinal immunopathologies later in life. Particularly crucial is a time period of gradual transition from relying solely on maternal milk to incorporating solid food, between 15 and 20 days of age, when a large number of microbial antigens is introduced and commensal microbiota are settling in the intestine. During this time, protective RORγt+ Treg cells are induced by the microbial antigens and normal intestinal homeostasis is sustained by induction of tolerance to commensal microbiota. Lack of RORγt+ Treg cell induction led in mice to the development of severe colitis.[52] The quantity of early-life-induced RORγt+ Tregs is influenced by maternal milk, particularly by the amount of IgA antibodies present in the maternal milk. In adult mice, RORγt+ Tregs and IgA exhibit mutual inhibition. Similarly, mice nursed by foster mothers with higher IgA titers in their milk will develop fewer RORγt+ Tregs compared to those fed with milk containing lower IgA titers.[53]

RORγt+ Tregs were also shown for their importance in oral tolerance and prevention of food allergies. Infants with developed food allergies have different composition of fecal microbiota in comparison to healthy infants and have increased

Clostridiales and Bacteroidales species. Upon their introduction, expansion of gut RORγt+ Treg cells in favor of GATA3+ Treg occurs,  mediating the protection against allergies.[54]

Deficiency of tryptophan, an essential amino acid, alters commensal microbiota metabolism which results in expansion of RORγt+ Treg cells and reduction of Gata3+ Treg cells. This induction is possibly regulated by stimulation of Aryl hydrocarbon receptor by metabolites produced by commensal bacteria using tryptophan as an energy source.[55]

Lower number of RORγt+ Treg cells is present in germ free mice colonized with microbiota associated with

Th2 cells and lower number of RORγt+ Treg cells is compensated by increased Helios+ Treg cells. How exactly may RORγt+ Tregs protect from colitis is not yet known.[56]

RORγt+ regulatory T lymphocytes in cancer

Pathological may be involvement of RORγt+ regulatory T cells in colorectal cancer. It was found, that RORγt+ Tregs which are able to express IL-17 are expanded in colorectal cancer and as cancer develops, they lose the ability to express anti-inflammatory IL-10. Similarly such RORγt+ Tregs expressing IL-17 are expanded in mucosa of patients with Crohn´s disease.[57][58] Depletion of RORγt+ Tregs in mice with colorectal cancer caused enhancement of reactivity of tumor-specific T cells and improved cancer immune surveillance. This improvement is not caused by the loss of IL-17 as that was proved to promote cancer progression.[58] In tumors of mice with conditional knockout of RORγt+ Tregs was confirmed downregulation of IL-6, reduction of IL-6 expressing CD11c+ dendritic cells and overexpression of CTLA-4. IL-6 mediates activation of STAT3 transcription factor which is critical for proliferation of cancer cells.[59]

Gata3+ regulatory T lymphocytes

Another important subset of Treg cells are Gata3+ Treg cells, which respond to IL-33 in the gut and influence the regulation of effector T cells during inflammation. Unlike RORγt+ Treg cells, these cells express Helios and are not dependent on the microbiome.[51][60]

Gata3+ T regs are major

IL-17a transcript.[61]

Disease

An important question in the field of immunology is how the immunosuppressive activity of regulatory T cells is modulated during the course of an ongoing immune response. While the immunosuppressive function of regulatory T cells prevents the development of autoimmune disease, it is not desirable during immune responses to infectious microorganisms. Current hypotheses suggest that, upon encounter with infectious microorganisms, the activity of regulatory T cells may be downregulated, either directly or indirectly, by other cells to facilitate elimination of the infection. Experimental evidence from mouse models suggests that some pathogens may have evolved to manipulate regulatory T cells to immunosuppress the host and so potentiate their own survival. For example, regulatory T cell activity has been reported to increase in several infectious contexts, such as

retroviral infections (the most well-known of which is HIV), mycobacterial infections (e.g., tuberculosis[62]), and various parasitic infections including Leishmania and malaria
.

Treg cells play major roles during HIV infection. They suppress the immune system, thus limiting target cells and reducing inflammation, but this simultaneously disrupts the clearance of virus by the cell-mediated immune response and enhances the reservoir by pushing CD4+ T cells to a resting state, including infected cells. Additionally, Treg cells can be infected by HIV, increasing the size of the HIV reservoir directly. Thus, Treg cells are being investigated as targets for HIV cure research.[63] Some Treg cell depletion strategies have been tested in SIV infected nonhuman primates, and shown to cause viral reactivation and enhanced SIV specific CD8+ T cell responses.[64]

Regulatory T cells have a large role in the pathology of visceral leishmaniasis and in preventing excess inflammation in patients cured of visceral leishmaniasis.

CD4+ regulatory T cells are often associated with solid tumours in both humans and murine models. Increased numbers of regulatory T cells in breast, colorectal and ovarian cancers is associated with a poorer prognosis.[65]

CD70+ non-Hodgkin lymphoma B cells induce FOXP3 expression and regulatory function in intratumoral CD4+CD25 T cells.[66]

There is some evidence that Treg cells may be dysfunctional and driving neuroinflammation in

amyotrophic lateral sclerosis due to lower expression of FOXP3.[67] Ex vivo expansion of Treg cells for subsequent autologous transplant is currently being investigated after promising results were obtained in a phase I clinical trial.[68]

Additionally, while regulatory T cells have been shown to increase via polyclonal expansion both systemically and locally during healthy pregnancies to protect the fetus from the maternal immune response (a process called maternal immune tolerance), there is evidence that this polyclonal expansion is impaired in preeclamptic mothers and their offspring.[69] Research suggests reduced production and development of regulatory T cells during preeclampsia may degrade maternal immune tolerance, leading to the hyperactive immune response characteristic of preeclampsia.[70]

Cancer

The recruitment and maintenance of Treg cells in the tumor microenvironment

Most tumors elicit an immune response in the host that is mediated by tumor antigens, thus distinguishing the tumor from other non-cancerous cells. This causes large numbers of

tumor-infiltrating lymphocytes (TILs) to be found in the tumor microenvironment.[71] Although it is not entirely understood, it is thought that these lymphocytes target cancerous cells and therefore slow or terminate the development of the tumor. However, this process is complicated because Treg cells seem to be preferentially trafficked to the tumor microenvironment. While Treg cells normally make up only about 4% of CD4+ T cells, they can make up as much as 20–30% of the total CD4+ population around the tumor microenvironment.[72]

Although high levels of TILs were initially thought to be important in determining an immune response against cancer, it is now widely recognized that the ratio of Treg to effector T cells in the tumor microenvironment is a determining factor in the success of the immune response against the cancer. High levels of Treg cells in the tumor microenvironment are associated with poor prognosis in many cancers,[73] such as ovarian, breast, renal, and pancreatic cancer.[72] This indicates that Treg cells suppress effector T cells and hinder the body's immune response against the cancer. However, in some types of cancer the opposite is true, and high levels of Treg cells are associated with a positive prognosis. This trend is seen in cancers such as colorectal carcinoma and follicular lymphoma. This could be due to Treg cells' ability to suppress general inflammation which is known to trigger cell proliferation and metastasis .[72] These opposite effects indicate that Treg cells' role in the development of cancer is highly dependent on both type and location of the tumor.

Although it is still not entirely understood how Treg cells are preferentially trafficked to the tumor microenvironment, the chemotaxis is probably driven by the production of chemokines by the tumor. Treg infiltration into the tumor microenvironment is facilitated by the binding of the chemokine receptor CCR4, which is expressed on Treg cells, to its ligand CCL22, which is secreted by many types of tumor cells.[74] Treg cell expansion at the site of the tumor could also explain the increased levels of Treg cells. The cytokine, TGF-β, which is commonly produced by tumor cells, is known to induce the differentiation and expansion of Treg cells.[74]

Forkhead box protein 3 (

TGF-β.[75]

Treg cells present in the tumor microenvironment (TME)  can be either induced Tregs or natural (thymic) Tregs which develop from naive precursors. However, tumor-associated Tregs may also originate from IL-17A+Foxp3+ Tregs which develop from Th17 cells.[76][77]

In general, the immunosuppression of the tumor microenvironment has largely contributed to the unsuccessful outcomes of many cancer immunotherapy treatments. Depletion of Treg cells in animal models has shown an increased efficacy of immunotherapy treatments, and therefore, many immunotherapy treatments are now incorporating Treg depletion.[2]

Cancer therapies targeting regulatory T lymphocytes

Tregs in the TME are abundantly effector Tregs which over-express immunosuppressive molecules such as CTLA-4. Anti-CTLA-4 antibodies cause depletion of Tregs and thus increase CD8+ T cells effective against the tumor. Anti-CTLA-4 antibody ipilimumab was approved for patients with advanced melanoma. Immune-checkpoint molecule PD-1 inhibits activation of both conventional T cells and Tregs and use of anti-PD-1 antibodies may lead to activation and immunosuppressive function of Tregs. Resistance to anti-PD-1-mAb treatment is probably caused by enhanced Treg cell activity. Rapid cancer progression upon PD-1 blockade is called hyperprogressive disease. Therapies targeting Treg suppression include anti-CD25 mAbs and anti-CCR4 mAbs. OX40 agonist and GITR agonists are currently being investigated.[76][78] Therapy targeting TCR signaling is also possible by blocking tyrosine kinases. For example, tyrosine-kinase inhibitor dasatinib is used for treatment of chronic myeloid leukemia and is associated with Treg inhibition.[79]

Molecular characterization

Similar to other T cells, regulatory T cells develop in the

interleukin-2 receptor alpha chain (CD25). In addition to the FOXP3-expressing CD4+ CD25+, there also appears to be a minor population of MHC class I restricted CD8+ FOXP3-expressing regulatory T cells. These FOXP3-expressing CD8+ T cells do not appear to be functional in healthy individuals but are induced in autoimmune disease states by T cell receptor stimulation to suppress IL-17-mediated immune responses.[81]
Unlike conventional T cells, regulatory T cells do not produce IL-2 and are therefore anergic at baseline.

A number of different methods are employed in research to identify and monitor Treg cells. Originally, high expression of CD25 and CD4 surface markers was used (CD4+CD25+ cells). This is problematic as CD25 is also expressed on non-regulatory T cells in the setting of immune activation such as during an immune response to a pathogen. As defined by CD4 and CD25 expression, regulatory T cells comprise about 5–10% of the mature CD4+ T cell subpopulation in mice and humans, while about 1–2% of Treg can be measured in whole blood. The additional measurement of cellular expression of FOXP3 protein allowed a more specific analysis of Treg cells (CD4+CD25+FOXP3+ cells). However, FOXP3 is also transiently expressed in activated human effector T cells, thus complicating a correct Treg analysis using CD4, CD25 and FOXP3 as markers in humans. Therefore, the gold standard surface marker combination to defined Tregs within unactivated CD3+CD4+ T cells is high CD25 expression combined with the absent or low-level expression of the surface protein CD127 (IL-7RA). If viable cells are not required then the addition of FOXP3 to the CD25 and CD127 combination will provide further stringency. Several additional markers have been described, e.g., high levels of CTLA-4 (cytotoxic T-lymphocyte associated molecule-4) and

GITR
(glucocorticoid-induced TNF receptor) are also expressed on regulatory T cells, however the functional significance of this expression remains to be defined. There is a great interest in identifying cell surface markers that are uniquely and specifically expressed on all FOXP3-expressing regulatory T cells. However, to date no such molecule has been identified.

The identification of Tregs following cell activation is challenging as conventional T cells will express CD25, transiently express FOXP3 and lose CD127 expression upon activation. It has been shown that Tregs can be detected using an activation-induced marker assay by expression of CD39

OX40(CD134) which define antigen-specific cells following 24-48h stimulation with antigen.[83][84]

In addition to the search for novel protein markers, a different method to analyze and monitor Treg cells more accurately has been described in the literature. This method is based on DNA methylation analysis. Only in Treg cells, but not in any other cell type, including activated effector T cells, a certain region within the FOXP3 gene (TSDR, Treg-specific-demethylated region) is found demethylated, which allows to monitor Treg cells through a PCR reaction or other DNA-based analysis methods.[85] Interplay between the Th17 cells and regulatory T cells are important in many diseases like respiratory diseases.[86]

Recent evidence suggests that

mast cells may be important mediators of Treg-dependent peripheral tolerance.[87]

Epitopes

Regulatory T cell epitopes ('Tregitopes') were discovered in 2008 and consist of linear sequences of amino acids contained within monoclonal antibodies and immunoglobulin G (IgG). Since their discovery, evidence has indicated Tregitopes may be crucial to the activation of natural regulatory T cells.[88][89][90]

Potential applications of regulatory T cell epitopes have been hypothesised: tolerisation to transplants, protein drugs, blood transfer therapies, and

type I diabetes as well as reduction of immune response for the treatment of allergies.[91][92][93][94][95][96][90]

Genetic deficiency

Genetic mutations in the gene encoding FOXP3 have been identified in both humans and mice based on the heritable disease caused by these mutations. This disease provides the most striking evidence that regulatory T cells play a critical role in maintaining normal immune system function. Humans with mutations in FOXP3 develop a severe and rapidly fatal autoimmune disorder known as Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome.[97][98]

The

diabetes mellitus
. Most individuals have other autoimmune phenomena including Coombs-positive hemolytic anemia, autoimmune thrombocytopenia, autoimmune neutropenia, and tubular nephropathy. The majority of affected males die within the first year of life of either metabolic derangements or sepsis. An analogous disease is also observed in a spontaneous FOXP3-mutant mouse known as "scurfy".

See also

References

  1. S2CID 4391497
    .
  2. ^ .
  3. .
  4. .
  5. .
  6. .
  7. .
  8. .
  9. .
  10. .
  11. .
  12. .
  13. .
  14. ^ .
  15. .
  16. .
  17. ^ .
  18. ^ .
  19. .
  20. .
  21. .
  22. ^ .
  23. .
  24. .
  25. .
  26. .
  27. .
  28. .
  29. .
  30. .
  31. .
  32. .
  33. .
  34. .
  35. .
  36. .
  37. ^ .
  38. .
  39. ^ .
  40. .
  41. .
  42. .
  43. .
  44. .
  45. ^ .
  46. .
  47. .
  48. .
  49. .
  50. ^ .
  51. ^ .
  52. .
  53. .
  54. .
  55. .
  56. .
  57. .
  58. ^ .
  59. .
  60. .
  61. .
  62. .
  63. .
  64. .
  65. .
  66. .
  67. .
  68. .
  69. .
  70. .
  71. .
  72. ^ .
  73. .
  74. ^ .
  75. .
  76. ^ .
  77. .
  78. .
  79. .
  80. .
  81. .
  82. .
  83. .
  84. .
  85. .
  86. .
  87. .
  88. ^ "Tregitope: Immunomodulation Power Tool". EpiVax. 2 August 2016.
  89. PMID 23857231
    .
  90. ^ .
  91. ^ "New $2.25M infusion of NIH funds for EpiVax' Tregitope, proposed "Paradigm-Shifting" Treatment". Fierce Biotech Research.
  92. PMID 23729499
    .
  93. .
  94. .
  95. .
  96. .
  97. ^ Online Mendelian Inheritance in Man IPEX
  98. ^ ipex at NIH/UW GeneTests

External links