Abstract
Recent advances highlight the importance of intrinsic peripheral tolerance in the maintenance of a steady state. Peripheral tolerance is tightly regulated and any alteration in its biological process contributes to the breakdown of immune tolerance and induction of autoimmunity. Recent evidence related to T cell tolerance mechanisms inspired researchers to treat autoimmunity via modulation of tolerant checkpoints that are involved in intrinsic T-cell tolerance such as ignorance, anergy, exhaustion, and senescence. So, understanding the underlying mechanisms might present an opportunity for therapeutic intervention. Here, we primarily highlight the importance of T cell-intrinsic peripheral tolerance mechanisms and their contribution to the development of autoimmune disorders, and then briefly discuss potential strategies to normalize T cell hemostasis in autoimmunity.
Keywords
Autoimmunity, Autoimmune-related disorders, Intrinsic peripheral tolerance, Immune checkpoint, Immunotherapy
Introduction
T cells are effector immune cells that coordinate the immune responses to cognate antigens in the presence of appropriate co-stimulatory molecules and cytokines. T-cell stimulation results in extended proliferation and differentiation of immune cells to eliminate pathogens/antigens (expansion phase). Clearance of antigens is associated with the contraction phase, during this phase, most of the immune cells undergo apoptotic cell death, while a small fraction is differentiated into central or effector memory cells, which accelerates pathogen clearance following the next exposure [1]. Given the nature of the immune system, its functional mechanisms should be under precise control, on one hand avoiding unwanted immune responses (over-activation or autoimmunity) and on the other initiating robust immune responses against infectious agents or tumors [2-4]. Several regulatory mechanisms have been suggested to control T cell hemostasis during differentiation, which mainly refers to T cell tolerance. T-cell tolerance can be divided into central and peripheral tolerance. Central tolerance is composed of several processes that finally induce clonal deletion of high-affinity autoreactive T cells in the thymus [5]. However, it seems to be almost imperfect, partly due to the lack of proper presentation of all peripheral auto-antigens in the thymus and incomplete clonal deletion (60-70% efficacy) [6], so there is an urgent need for peripheral tolerance mechanisms to avoid autoimmunity [7]. Peripheral tolerance involves several arms to eliminate/inactive autoreactive T cells that escape from central tolerance, including clonal deletion or suppression by regulatory T cells (Treg) and intrinsic peripheral tolerance mechanisms that force autoreactive T cells to remain unresponsiveness in a steady state [8,9]. In fact, T cell tolerance is tightly regulated and any alteration in its biological process contributes to the breakdown of immune tolerance and induction of autoimmunity. Recent evidence related to T cell tolerance mechanisms inspired researchers to treat autoimmunity via modulation of tolerance checkpoints that are involved in intrinsic T cell tolerance such as ignorance, anergy, exhaustion, and senescence. These tolerance checkpoints control immune tolerance through different mechanisms (Figure 1). At the naive stage, ignorance and anergy maintain tolerance, while at the effector stage, exhaustion and senescence are the main tolerance checkpoints that prevent the over-activation of the immune system and limit its responses (More details are shown in Table 1). So, understanding the specific role of each tolerance checkpoint in T cells might represent an opportunity for therapeutic intervention. Here, we primarily highlight the importance of T cell-intrinsic peripheral tolerance mechanisms and their contribution to autoimmunity. Then discuss potential strategies to normalize T cell hemostasis in autoimmunity. On this basis, the authors wish to build up a framework to develop more refined therapies for autoimmune-related disorders.
Figure 1. Intrinsic T cell tolerance checkpoints. T cell tolerance checkpoints regulate T cell responses through different mechanisms. At the naive stage, ignorance and anergy enforce T cell tolerance, while exhaustion and senescence are responsible for T cell hypo-responsiveness at the effector stage.
Intrinsic peripheral tolerance checkpoints |
Main features |
T cell status |
Predictive markers |
Reversible/ Irreversible |
Ignorance |
|
|
|
|
Anergy |
|
|
|
|
Exhaustion |
|
|
|
|
Senescence |
|
|
|
|
Tolerance Checkpoints
Ignorance
T-cell ignorance is the maintenance of low affinity autoreactive naïve T cell phenotype in a steady state [7,10]. Simply, autoreactive T cells fail to induce autoimmunity despite the presence of auto-antigens and remain ignorant or unaware. The precise mechanisms that control immune ignorance are not fully understood. However, one probable mechanism that maintains T cells in a naïve state is their lower affinity for auto-antigens [11]. In fact, self-reactive T cells that escape from central tolerance have different affinities for auto-antigens. It’s believed that autoreactive T cells with high affinity can rarely pass the clonal deletion process in the thymus, but if so, they probably become anergic T cells in the absence of appropriate co-stimulatory signals [12]. Thus, primarily low-affinity autoreactive T cells constitute an immunologically ignorant state. Besides, ignorance could be related to low expression of auto-antigens and/or their anatomical location such as immune-privileged tissues (eye, testis, blood-brain barrier) that sequestered antigens from the immune system [13]. Indeed, the concept of ignorance came from observations that indicate physical damage to a specific organ leads to altered antigen expression and the development of autoimmune disease as seen in the case of sympathetic ophthalmia [14]. Besides, transgenic mice have brought substantial information due to the ignorance phenomenon. Various foreign antigens could be determined as auto-antigen when inserted into the genome of mice. Antigens-specific autoreactive T cells probably proceed to be ignorant, anergic, or regulatory. In fact, immunological ignorance was originally described by Ohashi and colleagues using transgenic mice. They observed LCMV-specific T cells of RIP-LCMV mice which crossed with LCMV-specific TCR transgenic P14 mice ignored LCMV antigens [15]. However, further studies demonstrated that immune ignorance could be broken down following LCMV infection (appropriate viral load) which leads to autoimmune diabetes and mainly indicates immune ignorance is reversible [16]. Generally, it has been accepted that inflammation caused by infectious agents is one of the most important mechanisms for activating autoreactive T cells [17]. Of note, ignorant T cells are not unresponsive/dysfunctional and have the capacity to activate in the presence of exogenous stimuli such as infection, inflammatory context, or cytokines [15,18,19]. Therefore, there is a possibility that immunological ignorance overcomes and potentially induces auto-inflammatory disorders. However, the underlying mechanisms or key initiating mediators which contribute to the breakdown of immunological ignorance are less clear and further investigation is essential to determine its role in autoimmunity.
Anergy
T cell anergy is a long-term hyporesponsive state of T cells which is characterized by a defect in growth factors secretion and proliferation following strong engagement of TCR in the absence of co-stimulatory molecules [20,21]. Interestingly, the engagement of co-inhibitory receptors alongside cognate antigens induces/ or maintains an anergy state [22]. Recent studies indicate the upregulation of multiple negative checkpoints such as cytotoxic T lymphocyte associated antigen 4 (CTLA-4), programmed cell death protein-1 (PD-1), lymphocyte activation gene-3 (LAG-3), and E3 ubiquitin ligases by anergic T contribute to the maintenance of anergy state [23,24]. Of note, there are no specific markers to distinguish anergic T cells from other T cell subsets. However, several predictive markers have been suggested to characterize anergic T cells, including expression of high levels of CD44, CD73, FR4, and lack of Foxp3 expression [25]. Correlation of these factors with impairment in cell cycle and cytokines production (main hallmarks of anergy) has been reported [20]. Besides, anergic T cells tend to be hypo-responsive even after exposure to the same antigen stimuli in the presence of optimal co-stimulation [20]. However, in vivo studies showed that the anergy state is reversible and anergic T cells could slowly recover their functional properties in the absence of cognate antigens or Treg cells, which indicates maintenance of a long-lasting anergy state requires persistence of antigen exposure and/or the presence of Treg cells [25-27].
Anergic T cells are commonly classified as clonal anergy (in vitro) and adaptive tolerance (in vivo) [20]. Indeed, clonal anergy could be induced in the presence of a strong first signal (TCR-peptide interaction) without supporting secondary signals (infection/adjuvant) or in the existence of low doses of agonist in optimal co-stimulation [28, 29], whereas in vivo anergic T cells primarily induced in the thymus and in the periphery following exposure to auto-antigens in suboptimal stimulation or inhibitory microenvironment which seen in cancer [28,30]. It is worth noting that, despite several overlapping properties such as an impairment in IL-2 production and T cell proliferation, deep investigation demonstrated substantial differences in the molecular pattern, phenotype, and functional properties of the in vitro and in vivo anergic state which indicates the requirement for different approaches to restore or maintain their properties [30]. For example, different signaling pathways have been found in clonal anergy and adoptive tolerance. Clonal anergy mainly showed deficiency in RAS/MAPK activity and NF-κB mobilization to the nucleus whereas defects in Zap70, calcium mobilization and NF-κB activity have been reported in in vivo anergy [28,30]. In addition, only in vitro anergy showed reversible capacity in the presence of external IL-2 or diacylglycerol kinase-α inhibitor [31,32].
It is believed that anergy has the capacity to limit the responsiveness of potentially autoreactive T cells and induce the progenitors of Treg cells [25,33]. Of note, it has been found that anergy-derived Treg cells could suppress the development of inflammatory bowel disease and arthritis in animal models [33]. Besides, anergy-derived Treg cells could induce anergy in other autoreactive T cells [33]. In turn, Treg cells are substantial for anergy state induction/maintenance [34]. Moreover, inhibition of mTORC1 activity induced anergy state [35]. Similarly, impairment in mTORC1 signaling leads to Foxp3 expression and differentiation of Treg cells [36]. Thus, anergic T cells along with Treg cells dampen the autoimmunity, so any alteration in the anergy state might lead to autoimmune-related disorders. Of note, the association of anergic phenotypes with several autoimmune disorders has been reported [37,38]. It has been reported that most insulin-specific CD4+ T cells in diabetes-susceptible NOD mice had an anergic phenotype in diabetes-susceptible NOD mice which decreased with age. These data suggest that NOD mice maintain tolerance via anergy which is lost by age [39]. In addition, the correlation of the anergy state with a better prognosis of rheumatoid arthritis (RA) has been reported [34]. Besides, Moulton et al. reported distinctive properties of T cells that were isolated from systemic lupus erythematosus (SLE) patients. Somewhat they showed activated/effector function and, on the other hand, anergic state properties which probably indicate impairment in anergy induction is associated with SLE progression [40]. It seems that many molecular mechanisms, especially defects in anergy state, are involved in autoimmunity pathogenesis, thus identification of these mechanisms might eventually lead to a better understanding of the nature of autoimmune disorders and the development of novel approaches to normalized T cell function.
Exhaustion
T-cell exhaustion is a hyporesponsive state of T cells at the effector stage which contributes to immune system dysfunction [41]. It was originally described in CD8 T cells during chronic viral infection in the presence of constant antigen exposure with the support of co-stimulatory signals from the local microenvironments [42,43]. Of interest, a strong T cell exhaustion state is associated with antigen exposure for longtime, poor CD4 T cell help, and over co-expression of inhibitory receptors [44,45]. The exact mechanisms that initiate the transition of effector T-cells into an exhausted state are not fully determined. However, various transcription factors have been reported to be involved in the induction of T cell exhaustion state, such as T-bet, Tox, TCF-1, XBP1, Eomes, and NFAT [46-48]. Exhausted T cells are mainly characterized by substantial changes in gene expression, transcription molecules, epigenetic profile, and cellular metabolic, which leads to over co-expression of inhibitory receptors (PD-1, LAG-3, T-cell immunoglobulin domain and mucin domain-3 (TIM-3), B and T lymphocyte attenuator (BTLA), T-cell immune receptor with Ig and ITIM domains (TIGIT), CTLA-4, and 2B4/CD244) and finally failure of effector functions [48,49].
It has been thought that T cell exhaustion is a compensatory mechanism in the presence of persistent antigen (in the case of chronic infection or cancer) to avoid over-activation of the immune system that may cause tissue injury, immunopathology, or even host death [50-53]. Since exhaustion could act in favor of dampening autoimmunity, it has been proposed that T cell exhaustion might be considered as another regulatory mechanism that suppresses the immune system against self-antigens that escape from central tolerance [54]. There are some studies that indicate an association of T cell exhaustion with better prognosis in several autoimmune-related disorders such as anti-neutrophil cytoplasmic antibody-associated vasculitis (ANCA), idiopathic pulmonary fibrosis, type 1 diabetes (T1D), SLE, and RA [55-57]. Interestingly, the extent of T cell exhaustion is inversely related to the severity of the disorders [57]. There was a positive link between CD8+ T-cell exhaustion accompanied by poor CD4+ help with a better prognosis in SLE, ANCA, and T1D [55,58]. Of interest, the proportion of the polyclonal exhausted population was relatively higher in systemic autoimmune diseases compared to T1D [56-58]. This difference might be related to the disease nature, as SLE and ANCA are systemic autoimmune diseases whereas T1D is organ-specific (autoimmune responses mainly localize in the pancreas) [59]. Besides, unique expression of inhibitory receptors on enriched exhausted CD8+ T-cells in different disorders has been reported [60]. It seems that T cell exhaustion is specifically context-dependent and its contribution to each disease is complex and needs further research to elucidate the precise mechanisms.
It is generally accepted that T cell exhaustion is a reversible process, so blocking the inhibitory receptors might have a beneficial effect against chronic viral infection or cancer, as documented in several studies [61,62]. There are various inhibitory pathways that are deeply studied in T cell exhaustion, especially PD-1/PD-L1-2 and CTLA-4 inhibitory receptor signaling [63-65]. It is widely known that the PD-1 and CTLA-4 receptors have a negative effect on the immune system and the U.S. Food and Drug Administration has approved their inhibitory function. Other immune checkpoints implicated in T cell exhaustion are TIM-3, BTLA, CD160, 2B4, CD39, TIGIT, and LAG-3 [66,67]. It is also worth noting that the clinical use of immune checkpoints has a significant effect on the treatment of chronic infectious diseases and cancers [50,68,69]. Interestingly, the 2018 Nobel Prize in Physiology or Medicine was awarded for cancer immunotherapy using an immune checkpoint to Allison and Honjo. Regarding the promising results obtained from cancer therapy using checkpoint blockers, it is tempting to propose that the induction of checkpoint molecules in the case of autoimmune disease might have a beneficial effect and targeted research in this field might be an important step to developing novel therapeutic approaches.
Senescence
Immunosenescence is a dysregulated function of immune cells which is primarily characterized by a reduction in telomerase activity and/or increment of telomere frailty, cessation in proliferative capacity, reduction in chemotactic and phagocytic activity, and changes in inflammatory mediators profile [70,71]. Indeed, immunosenescence proceeds during the normal aging process, but inflammation due to several circumstances such as chronic infection, cancer, and chronic autoimmune disorders could imitate the aging process and accelerate immunosenescence regardless of physiological age [72-75]. It’s believed that repeated exposure to antigens and stress during the lifespan leads to constant activation of the immune system and induction of low-grade inflammation which is collectively called inflammaging. Inflammaging is an important hallmark of immunosenescence and mainly contributes to the development of several chronic age-related disorders and accelerated immunosenescence in young healthy individuals [76]. Reciprocally, immunosenescence could potentially induce chronic autoimmune disorders, indicating its substantial role in immune system homeostasis [76]. Generally, aging alters all aspects of the immune system, in particular T cell bioactivity [77]. Aging affects the proportion of circulatory naïve T cell decrease which is accompanied by alteration in T cell subtype composition [78]. In fact, the frequency of T helper type 2 cells (Th2), Treg cells, and memory T cells increases. This alteration might contribute to insufficient adaptive immune responses and/or alteration in memory cell responses, which finally leads to T cell dysfunction [79-81].
Interestingly, the association of T cell senescence phenotype with the progressive down-regulation of co-stimulatory molecules (CD28 and CD27), reduction in TCR signaling activity, and up-regulation of other surface receptors such as killer cell lectin-like receptor subfamily G member 1 (KLRG-1), CD57, T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and CD45RA have been reported [72,82-85]. Besides, senescent T cells produce a myriad of bio-active molecules known as senescence-associated secretory phenotypes (SASP) including cytokines, chemokines, proteases, and other pro-inflammatory mediators which probably are involved in the dysregulated properties of the immune system [83,86-88].
Immunosenescence also plays a substantial role in the development of autoimmune-related disorders [89,90]. Broux and colleagues reported the infiltration of CD4+ CD28- T cells in the CNS of MS patients [91]. Along this line, van nierop et al. observed the presence of chronically activated cytotoxic CD8+ T cells in MS patients [92]. Further studies in the auto-inflammatory setting demonstrated significant telomere frailty in CD4+ T cells obtained from inflamed synovium of RA patients [93,94]. Notably, senescent T cell phenotype (CD28− CD57+ KLRG-1+ CD8+ T cells) was associated with SLE pathogenesis [95,96]. Similarly, a higher frequency of cytotoxic CD28− CD8+ T cells has been observed in patients with Ankylosing Spondylitis (AS), Graves’ disease (GD), and Behçet’s disease (BD) [97-99]. Taken together, these results strongly suggest the involvement of T cell senescence in autoimmunity pathogenesis. However, relatively little is known about the underlying mechanisms. It appears that understanding the precise mechanisms might be a means to prevent chronic inflammation right before its contribution to developing autoimmunity.
Therapeutic Potential of Immune Checkpoints
In recent years, autoimmune diseases have increased uncontrollably and despite great endeavor and vast expense, current treatments are not specific and efficient enough to treat autoimmune-related disorders. In addition, long-term use of these non-specific immunomodulatory agents increases the risk of infectious diseases or malignancies [100]. Thus, there is still an urgent necessity to develop novel treatments that specifically focus on mechanisms driving autoimmunity. The breakdown of intrinsic peripheral tolerance is one of the most important mechanisms that contributes to autoimmunity pathogenesis [101-103]. So, restoring function or modulating pathways involved in autoimmunity might be an important step in developing new immunotherapeutic strategies. Recent studies in the understanding of intrinsic peripheral tolerance mechanisms inspired researchers to treat autoimmunity via modulation of tolerance checkpoints that are involved in intrinsic T cell tolerance. These include the use of soluble peptides, nanoparticles, small molecules, or agonists/antagonists that block co-stimulatory molecules or trigger inhibitory receptor expression. The first example is immunotherapy with soluble peptides designed as antigen-processing independent T-cell epitopes (apitopes). Sundstedt et al. reported Ac1–9 (N-terminal peptide of MBP) as an apitope and administration of its analog (4Y) induces an anergy state in the Tg4 mouse model [104]. Further studies by Burton et al. showed that repeated exposure to soluble apitopes (4Y) in Tg4 mice induces upregulation of inhibitory receptors including CTLA-4, PD-1, LAG-3, and TIM-3 and anergy state, leading to lifelong protection of disease [105]. Nanoparticles containing disease-specific antigens might induce anergy, as Jamison et al. demonstrated the efficacy of nanoparticles containing insulin–ChgA hybrid peptide in the induction of anergy state in NOD mice [106]. Besides, antagonists such as teplizumab, an anti-CD3 blocking antibody, could modulate inhibitory receptor expression indirectly and induce an anergic or exhausted state in T cells of T1D patients [55,107]. Of interest, there are several clinical trials that investigated the efficacy of immune checkpoints in autoimmune-related disorders. Among them, abatacept (CTLA-4-Ig), which blocks co-stimulatory signals, showed significant clinical benefits for RA patients and has now been approved by the US Food and Drug Administration (FDA) [108-111]. Abatacept was also studied in clinical trials for lupus nephritis and juvenile RA, the results were encouraging [112-115]. Rosnilimab and Peresolimab are PD-1 agonist antibodies that have been used for RA therapy and showed significant clinical benefits for RA in phases I and II clinical studies [116,117]. In addition, Rosnilimab demonstrated a significant reduction in ulcerative colitis (UC) clinical manifestation and is now in Phase II clinical studies [118]. These clinical trial studies indicate the promising effects of immune checkpoints for the treatment of autoimmune-related disorders.
Taken together, it seems that this therapeutic intervention has a great advantage over current treatment. Most importantly, it is antigen-specific, so it avoids systemic suppression of the immune system and maintains the benefit of the immune response. Despite such valuable benefits and promising results, tolerance checkpoints are still far from being an effective treatment for autoimmune disorders. To begin with, the timing of therapeutic intervention is critical. Given the epitope spread and complex inflammatory milieu after the development of autoimmune disorders, it is reasonable to suggest that such therapeutic strategies may be more effective early in the course of autoimmune disease and be defined as a prophylactic treatment in genetically susceptible individuals. Furthermore, it is often highly challenging to identify appropriate agonists or antagonists to treat autoimmune disorders based on the nature or stage of the disease to achieve therapeutic purposes. Accordingly, there is a lack of appropriate biomarkers to evaluate its clinical benefits. Finally, it should not be forgotten that the concept of the therapeutic potential of immune checkpoints is very abstract, and the current data is limited, so further studies are still needed to evaluate the potential of these approaches and probable side effects.
Conclusions and Final Remarks
The evidence that emerges from recent studies highlights the potential clinical benefits of tolerance checkpoints to treat autoimmunity. However, the precise mechanisms that maintain a tolerant state remain to be elucidated. One major challenge is identifying master genes, transcription/epigenetic factors, or other regulators that are involved in the maintenance or disruption of intrinsic peripheral tolerance. In addition, there is little or no data about the outcome of therapeutic intervention. It is not clear whether targeted tolerance checkpoint therapy has enough efficacy and safety or can even modulate the immune responses without deleterious side effects. This data and a comprehensive conception of the tolerant state will be essential for designing new effective therapeutic strategies to treat autoimmune-related disorders.
Statements and Declarations
Conflict of interest
The authors declare that they have no conflict of interest.
Funding
The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.
Acknowledgments
The authors have not acknowledged to declare.
Authors’ contributions
Naser GHolijani: performed the literature search, Manuscript writing, final approval of the manuscript; Golamreza Daryabor: performed the literature search, Manuscript writing, final approval of the manuscript; Fatemeh Rezaei Kahmini: Conceptualized the study, performed the literature search, manuscript writing, final approval of the manuscript, revising the manuscript and overall supervision.
References
2. Tang R, Rangachari M, Kuchroo VK. Tim-3: A co-receptor with diverse roles in T cell exhaustion and tolerance. Seminars in Immunology. 2019 Apr 1;42:101302.
3. Courtney AH, Lo WL, Weiss A. TCR signaling: mechanisms of initiation and propagation. Trends in Biochemical Sciences. 2018 Feb 1;43(2):108-23.
4. Skapenko A, Leipe J, Lipsky PE, Schulze-Koops H. The role of the T cell in autoimmune inflammation. Arthritis Research & Therapy. 2005 Mar;7:1-11.
5. Tripathi C. Tolerance and Autoimmunity. In: Singh IK, Sharma P, eds. An Interplay of Cellular and Molecular Components of Immunology. CRC Press; 2022 Dec 19. pp. 207-16.
6. Cebula A, Kuczma M, Szurek E, Pietrzak M, Savage N, Elhefnawy WR, et al. Dormant pathogenic CD4+ T cells are prevalent in the peripheral repertoire of healthy mice. Nature Communications. 2019 Oct 25;10(1):4882.
7. Theofilopoulos AN, Kono DH, Baccala R. The multiple pathways to autoimmunity. Nature Immunology. 2017 Jul 1;18(7):716-24.
8. Klein L, Robey EA, Hsieh CS. Central CD4+ T cell tolerance: deletion versus regulatory T cell differentiation. Nature Reviews Immunology. 2019 Jan;19(1):7-18.
9. Kahmini FR, Shahgaldi S. Harnessing the inherent power of chimeric antigen receptor (CAR)-expressing regulatory T cells (CAR-Tregs) to treat autoimmune-related disorders. Molecular Biology Reports. 2022 May;49(5):4069-78.
10. Parish IA, Heath WR. Too dangerous to ignore: self-tolerance and the control of ignorant autoreactive T cells. Immunology and Cell Biology. 2008 Feb;86(2):146-52.
11. Leube J, Mühlbauer A, Andrä I, Biggel M, Busch DH, Kretschmer L, et al. Single‐cell fate mapping reveals widespread clonal ignorance of low‐affinity T cells exposed to systemic infection. European Journal of Immunology. 2023 Mar;53(3):2250009.
12. Kurts C, Sutherland RM, Davey G, Li M, Lew AM, Blanas E, et al. CD8 T cell ignorance or tolerance to islet antigens depends on antigen dose. Proceedings of the National Academy of Sciences. 1999 Oct 26;96(22):12703-7.
13. Grabowski MM, Sankey EW, Ryan KJ, Chongsathidkiet P, Lorrey SJ, Wilkinson DS, et al. Immune suppression in gliomas. Journal of Neuro-Oncology. 2021 Jan;151:3-12.
14. Parchand S, Agrawal D, Ayyadurai N, Agarwal A, Gangwe A, Behera S, et al. Sympathetic ophthalmia: A comprehensive update. Indian Journal of Ophthalmology. 2022 Jun 1;70(6):1931-44.
15. Ohashi PS, Oehen S, Buerki K, Pircher H, Ohashi CT, Odermatt B, et al. Ablation of “tolerance” and induction of diabetes by virus infection in viral antigen transgenic mice. Cell. 1991 Apr 19;65(2):305-17.
16. Fousteri G, Dave Jhatakia A. Viral infections and autoimmune disease: roles of LCMV in delineating mechanisms of immune tolerance. Viruses. 2019 Sep 21;11(10):885.
17. Lang KS, Recher M, Junt T, Navarini AA, Harris NL, Freigang S, et al. Toll-like receptor engagement converts T-cell autoreactivity into overt autoimmune disease. Nature Medicine. 2005 Feb 1;11(2):138-45.
18. Vezys V, Lefrançois L. Cutting edge: inflammatory signals drive organ-specific autoimmunity to normally cross-tolerizing endogenous antigen. The Journal of Immunology. 2002 Dec 15;169(12):6677-80.
19. Ramanathan S, Dubois S, Chen XL, Leblanc C, Ohashi PS, Ilangumaran S. Exposure to IL-15 and IL-21 enables autoreactive CD8 T cells to respond to weak antigens and cause disease in a mouse model of autoimmune diabetes. The Journal of Immunology. 2011 May 1;186(9):5131-41.
20. Schwartz RH. T cell anergy. Annual Review of Immunology. 2003 Apr;21(1):305-34.
21. Tuncel J, Benoist C, Mathis D. T cell anergy in perinatal mice is promoted by T reg cells and prevented by IL-33. Journal of Experimental Medicine. 2019 Jun 3;216(6):1328-44.
22. Greenwald RJ, Boussiotis VA, Lorsbach RB, Abbas AK, Sharpe AH. CTLA-4 regulates induction of anergy in vivo. Immunity. 2001 Feb 1;14(2):145-55.
23. Mueller DL. E3 ubiquitin ligases as T cell anergy factors. Nature Immunology. 2004 Sep 1;5(9):883-90.
24. Williams JB, Horton BL, Zheng Y, Duan Y, Powell JD, Gajewski TF. The EGR2 targets LAG-3 and 4-1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment. Journal of Experimental Medicine. 2017 Feb 1;214(2):381-400.
25. Kalekar LA, Mueller DL. Relationship between CD4 regulatory T cells and anergy in vivo. The Journal of Immunology. 2017 Apr 1;198(7):2527-33.
26. Pape KA, Merica R, Mondino A, Khoruts A, Jenkins MK. Direct evidence that functionally impaired CD4+ T cells persist in vivo following induction of peripheral tolerance. The Journal of Immunology. 1998 May 15;160(10):4719-29.
27. Rocha B, Tanchot C, Von Boehmer H. Clonal anergy blocks in vivo growth of mature T cells and can be reversed in the absence of antigen. The Journal of Experimental Medicine. 1993 May 1;177(5):1517-21.
28. Chiodetti L, Choi S, Barber DL, Schwartz RH. Adaptive tolerance and clonal anergy are distinct biochemical states. The Journal of Immunology. 2006 Feb 15;176(4):2279-91.
29. Mirshahidi S, Huang CT, Sadegh-Nasseri S. Anergy in peripheral memory CD4+ T cells induced by low avidity engagement of T cell receptor. The Journal of Experimental Medicine. 2001 Sep 17;194(6):719-32.
30. Choi S, Schwartz RH. Molecular mechanisms for adaptive tolerance and other T cell anergy models. Seminars in Immunology. 2007 Jun 1;19(3):140-52.
31. Zha Y, Marks R, Ho AW, Peterson AC, Janardhan S, Brown I, et al. T cell anergy is reversed by active Ras and is regulated by diacylglycerol kinase-α. Nature Immunology. 2006 Nov 1;7(11):1166-73.
32. Boussiotis VA, Barber DL, Nakarai T, Freeman GJ, Gribben JG, Bernstein GM, et al. Prevention of T cell anergy by signaling through the γc chain of the IL-2 receptor. Science. 1994 Nov 11;266(5187):1039-42.
33. Kalekar LA, Schmiel SE, Nandiwada SL, Lam WY, Barsness LO, Zhang N, et al. CD4+ T cell anergy prevents autoimmunity and generates regulatory T cell precursors. Nature Immunology. 2016 Mar;17(3):304-14.
34. Martinez RJ, Zhang N, Thomas SR, Nandiwada SL, Jenkins MK, Binstadt BA, et al. Arthritogenic self-reactive CD4+ T cells acquire an FR4hiCD73hi anergic state in the presence of Foxp3+ regulatory T cells. The Journal of Immunology. 2012 Jan 1;188(1):170-81.
35. Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T cells are metabolically anergic. The Journal of Immunology. 2009 Nov 15;183(10):6095-101.
36. Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity. 2009 Jun 19;30(6):832-44.
37. Itoh A, Ortiz L, Kachapati K, Wu Y, Adams D, Bednar K, et al. Soluble CD137 ameliorates acute type 1 diabetes by inducing T cell anergy. Frontiers in Immunology. 2019 Nov 7;10:2566.
38. Vallejo SA, Basallo HS, Narvaes M, Medina YF, Quintana-López G. Frequency of anergy in a group of patients with rheumatoid arthritis on immunosuppressive therapy. Revista Colombiana de Reumatología. 2021 Mar;28(1):16-27.
39. Pauken KE, Linehan JL, Spanier JA, Sahli NL, Kalekar LA, Binstadt BA, et al. Cutting edge: type 1 diabetes occurs despite robust anergy among endogenous insulin-specific CD4 T cells in NOD mice. The Journal of Immunology. 2013 Nov 15;191(10):4913-7.
40. Moulton VR, Tsokos GC. Abnormalities of T cell signaling in systemic lupus erythematosus. Arthritis Research & Therapy. 2011 Apr;13:1-10.
41. Belk JA, Daniel B, Satpathy AT. Epigenetic regulation of T cell exhaustion. Nature Immunology. 2022 Jun;23(6):848-60.
42. Kahan SM, Wherry EJ, Zajac AJ. T cell exhaustion during persistent viral infections. Virology. 2015 May 1;479:180-93.
43. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaustion during chronic viral infection and cancer. Annual Review of Immunology. 2019 Apr 26;37:457-95.
44. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nature Immunology. 2009 Jan;10(1):29-37.
45. Wherry EJ. T cell exhaustion. Nature Immunology. 2011 Jun;12(6):492-9.
46. Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity. 2020 May 19;52(5):825-41.
47. Yao C, Sun HW, Lacey NE, Ji Y, Moseman EA, Shih HY, et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nature Immunology. 2019 Jul;20(7):890-901.
48. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nature Reviews Immunology. 2015 Aug;15(8):486-99.
49. Yi JS, Cox MA, Zajac AJ. T‐cell exhaustion: characteristics, causes and conversion. Immunology. 2010 Apr;129(4):474-81.
50. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends in Immunology. 2015 Apr 1;36(4):265-76.
51. Jin X, Bauer DE, Tuttleton SE, Lewin S, Gettie A, Blanchard J, et al. Dramatic rise in plasma viremia after CD8+ T cell depletion in simian immunodeficiency virus–infected macaques. The Journal of Experimental Medicine. 1999 Mar 15;189(6):991-8.
52. Frebel H, Nindl V, Schuepbach RA, Braunschweiler T, Richter K, Vogel J, et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. Journal of Experimental Medicine. 2012 Dec 17;209(13):2485-99.
53. Zinselmeyer BH, Heydari S, Sacristán C, Nayak D, Cammer M, Herz J, et al. PD-1 promotes immune exhaustion by inducing antiviral T cell motility paralysis. Journal of Experimental Medicine. 2013 Apr 8;210(4):757-74.
54. Carney EF. T-cell exhaustion limits immune reactivity and is associated with good prognosis in autoimmune disease. Nature Reviews Rheumatology. 2015 Sep;11(9):501.
55. Long SA, Thorpe J, DeBerg HA, Gersuk V, Eddy JA, Harris KM, et al. Partial exhaustion of CD8 T cells and clinical response to teplizumab in new-onset type 1 diabetes. Science Immunology. 2016 Nov 18;1(5):eaai7793.
56. McKinney EF, Smith KG. T cell exhaustion and immune-mediated disease—the potential for therapeutic exhaustion. Current Opinion in Immunology. 2016 Dec 1;43:74-80.
57. McKinney EF, Lee JC, Jayne DR, Lyons PA, Smith KG. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature. 2015 Jul 30;523(7562):612-6.
58. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nature Immunology. 2007 Mar;8(3):239-45.
59. Culina S, Lalanne AI, Afonso G, Cerosaletti K, Pinto S, Sebastiani G, et al. Islet-reactive CD8+ T cell frequencies in the pancreas, but not in blood, distinguish type 1 diabetic patients from healthy donors. Science Immunology. 2018 Feb 2;3(20):eaao4013.
60. Gao Z, Feng Y, Xu J, Liang J. T-cell exhaustion in immune-mediated inflammatory diseases: new implications for immunotherapy. Frontiers in Immunology. 2022 Sep 23;13:977394.
61. Dolina JS, Van Braeckel-Budimir N, Thomas GD, Salek-Ardakani S. CD8+ T Cell Exhaustion in Cancer. Front Immunol. 2021; 12: 715234.
62. Utzschneider DT, Gabriel SS, Chisanga D, Gloury R, Gubser PM, Vasanthakumar A, et al. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nature Immunology. 2020 Oct;21(10):1256-66.
63. Jin HT, Anderson AC, Tan WG, West EE, Ha SJ, Araki K, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proceedings of the National Academy of Sciences. 2010 Aug 17;107(33):14733-8.
64. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006 Sep 21;443(7109):350-4.
65. Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, et al. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathogens. 2009 Feb 27;5(2):e1000313.
66. Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS pathogens. 2015 Oct 20;11(10):e1005177.
67. Woroniecka KI, Rhodin KE, Chongsathidkiet P, Keith KA, Fecci PE. T-cell dysfunction in glioblastoma: applying a new framework. Clinical Cancer Research. 2018 Aug 15;24(16):3792-802.
68. Xu F, Jin T, Zhu Y, Dai C. Immune checkpoint therapy in liver cancer. Journal of Experimental & Clinical Cancer Research. 2018 Dec;37:1-2.
69. Velu V, Titanji K, Zhu B, Husain S, Pladevega A, Lai L, et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009 Mar 12;458(7235):206-10.
70. Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. InSeminars in Immunopathology 2020 Oct (Vol. 42, pp. 559-572). Springer Berlin Heidelberg.
71. Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et.al. Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell Metabolism. 2020 Jul 7;32(1):44-55.
72. Weyand CM, Yang Z, Goronzy JJ. T-cell aging in rheumatoid arthritis. Current Opinion in Rheumatology. 2014 Jan 1;26(1):93-100.
73. Fessler J, Fasching P, Raicht A, Hammerl S, Weber J, Lackner A, et al. Lymphopenia in primary Sjögren’s syndrome is associated with premature aging of naïve CD4+ T cells. Rheumatology. 2021 Feb 1;60(2):588-97.
74. Bellon M, Nicot C. Telomere dynamics in immune senescence and exhaustion triggered by chronic viral infection. Viruses. 2017 Oct 5;9(10):289.
75. Zhao Y, Shao Q, Peng G. Exhaustion and senescence: two crucial dysfunctional states of T cells in the tumor microenvironment. Cellular & Molecular Immunology. 2020 Jan;17(1):27-35.
76. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences. 2014 Jun 1;69(Suppl_1):S4-9.
77. Rodriguez IJ, Lalinde Ruiz N, Llano León M, Martínez Enríquez L, Montilla Velásquez MD, Ortiz Aguirre JP, et al. Immunosenescence study of T cells: a systematic review. Frontiers in Immunology. 2021 Jan 15;11:604591.
78. Cossarizza A, Ortolani C, Paganelli R, Barbieri D, Monti D, Sansoni P, et al. CD45 isoforms expression on CD4+ and CD8+ T cells throughout life, from newborns to centenarians: implications for T cell memory. Mechanisms of Ageing and Development. 1996 Mar 29;86(3):173-95.
79. Fulop T, Larbi A, Pawelec G. Human T cell aging and the impact of persistent viral infections. Frontiers in Immunology. 2013 Sep 13;4:52731.
80. Goronzy JJ, Li G, Yu M, Weyand CM. Signaling pathways in aged T cells–a reflection of T cell differentiation, cell senescence and host environment. Seminars in Immunology. 2012 Oct 1;24(5):365-72.
81. Jergović M, Smithey MJ, Nikolich-Žugich J. Intrinsic and extrinsic contributors to defective CD8+ T cell responses with aging. Experimental Gerontology. 2018 May 1;105:140-5.
82. Pereira BI, De Maeyer RP, Covre LP, Nehar-Belaid D, Lanna A, Ward S, et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nature Immunology. 2020 Jun;21(6):684-94.
83. Covre LP, De Maeyer RP, Gomes DC, Akbar AN. The role of senescent T cells in immunopathology. Aging cell. 2020 Dec;19(12):e13272.
84. Li H, Wu K, Tao K, Chen L, Zheng Q, Lu X, et al. Tim‐3/galectin‐9 signaling pathway mediates T‐cell dysfunction and predicts poor prognosis in patients with hepatitis B virus‐associated hepatocellular carcinoma. Hepatology. 2012 Oct;56(4):1342-51.
85. Heffner M, Fearon DT. Loss of T cell receptor-induced Bmi-1 in the KLRG1+ senescent CD8+ T lymphocyte. Proceedings of the National Academy of Sciences. 2007 Aug 14;104(33):13414-9.
86. Goronzy JJ, Weyand CM. Mechanisms underlying T cell ageing. Nature Reviews Immunology. 2019 Sep;19(9):573-83.
87. Akbar AN, Henson SM, Lanna A. Senescence of T lymphocytes: implications for enhancing human immunity. Trends in Immunology. 2016 Dec 1;37(12):866-76.
88. Libri V, Azevedo RI, Jackson SE, Di Mitri D, Lachmann R, Fuhrmann S, et al. Cytomegalovirus infection induces the accumulation of short‐lived, multifunctional CD4+ CD45RA+ CD27− T cells: the potential involvement of interleukin‐7 in this process. Immunology. 2011 Mar;132(3):326-39.
89. Zuo L, Prather ER, Stetskiv M, Garrison DE, Meade JR, Peace TI, et al. Inflammaging and oxidative stress in human diseases: from molecular mechanisms to novel treatments. International Journal of Molecular Sciences. 2019 Sep 10;20(18):4472.
90. Chalan P, van den Berg A, Kroesen BJ, Brouwer L, Boots A. Rheumatoid arthritis, immunosenescence and the hallmarks of aging. Current Aging Science. 2015 Jul 1;8(2):131-46.
91. Broux B, Pannemans K, Zhang X, Markovic-Plese S, Broekmans T, Eijnde BO, et al. CX3CR1 drives cytotoxic CD4+ CD28− T cells into the brain of multiple sclerosis patients. Journal of Autoimmunity. 2012 Feb 1;38(1):10-9.
92. van Nierop GP, van Luijn MM, Michels SS, Melief MJ, Janssen M, Langerak AW, et al. Phenotypic and functional characterization of T cells in white matter lesions of multiple sclerosis patients. Acta neuropathologica. 2017 Sep;134:383-401.
93. Li Y, Shen Y, Hohensinner P, Ju J, Wen Z, Goodman SB, et al. Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis. Immunity. 2016 Oct 18;45(4):903-16.
94. Petersen LE, Grassi-Oliveira R, Siara T, Ribeiro dos Santos SG, Ilha M, De Nardi T, et al. Premature immunosenescence is associated with memory dysfunction in rheumatoid arthritis. Neuroimmunomodulation. 2015 Apr 15;22(3):130-7.
95. Kalim H, Pratama MZ, Mahardini E, Winoto ES, Krisna PA, Handono K. Accelerated immune aging was correlated with lupus‐associated brain fog in reproductive‐age systemic lupus erythematosus patients. International Journal of Rheumatic Diseases. 2020 May;23(5):620-6.
96. Handono K, Wahono CS, Pratama MZ, Kalim H. Association of the premature immunosenescence with the presence and severity of anemia among patients with systemic lupus erythematosus. Lupus. 2021 Oct;30(12):1906-14.
97. Schirmer M, Goldberger C, Würzner R, Duftner C, Pfeiffer KP, Clausen J, et al. Circulating cytotoxic CD8+ CD28-T cells in ankylosing spondylitis. Arthritis Research & Therapy. 2001 Oct;4:1-8.
98. Yang JY, Park MJ, Park S, Lee ES. Increased senescent CD8+ T cells in the peripheral blood mononuclear cells of Behçet’s disease patients. Archives of Dermatological Research. 2018 Mar;310:127-38.
99. Sun Z, Zhong W, Lu X, Shi B, Zhu Y, Chen L, et al. Association of Graves’ Disease and Prevalence of Circulating IFN-γ-producing CD28− T Cells. Journal of Clinical Immunology. 2008 Sep;28:464-72.
100. Kahmini FR, Shahgaldi S. Therapeutic potential of mesenchymal stem cell-derived extracellular vesicles as novel cell-free therapy for treatment of autoimmune disorders. Experimental and Molecular Pathology. 2021 Feb 1;118:104566.
101. ElTanbouly MA, Noelle RJ. Rethinking peripheral T cell tolerance: checkpoints across a T cell’s journey. Nature Reviews Immunology. 2021 Apr;21(4):257-67.
102. Datta SK, Zhang L, Xu L. T-helper cell intrinsic defects in lupus that break peripheral tolerance to nuclear autoantigens. Journal of Molecular Medicine. 2005 Apr;83:267-78.
103. Jeker LT, Bour-Jordan H, Bluestone JA. Breakdown in peripheral tolerance in type 1 diabetes in mice and humans. Cold Spring Harbor perspectives in medicine. 2012 Mar 1;2(3):a007807.
104. Sundstedt A, O’Neill EJ, Nicolson KS, Wraith DC. Role for IL-10 in suppression mediated by peptide-induced regulatory T cells in vivo. The Journal of Immunology. 2003 Feb 1;170(3):1240-8.
105. Burton BR, Britton GJ, Fang H, Verhagen J, Smithers B, Sabatos-Peyton CA, et al. Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nature communications. 2014 Sep 3;5(1):4741.
106. Jamison BL, Neef T, Goodspeed A, Bradley B, Baker RL, Miller SD, et al. Nanoparticles containing an insulin–ChgA hybrid peptide protect from transfer of autoimmune diabetes by shifting the balance between effector T cells and regulatory T cells. The Journal of Immunology. 2019 Jul 1;203(1):48-57.
107. Long SA, Thorpe J, Herold KC, Ehlers M, Sanda S, Lim N, et al. Remodeling T cell compartments during anti-CD3 immunotherapy of type 1 diabetes. Cellular immunology. 2017 Sep 1;319:3-9.
108. Fife BT, Bluestone JA. Control of peripheral T‐cell tolerance and autoimmunity via the CTLA‐4 and PD‐1 pathways. Immunological reviews. 2008 Aug;224(1):166-82.
109. Ahamada MM, Wu X. Analysis of efficacy and safety of abatacept for rheumatoid arthritis: systematic review and meta-analysis. Clinical and Experimental Rheumatology. 2023;41(9):1882-900.
110. Cope AP, Jasenecova M, Vasconcelos JC, Filer A, Raza K, Qureshi S, et al. Abatacept in individuals at high risk of rheumatoid arthritis (APIPPRA): a randomised, double-blind, multicentre, parallel, placebo-controlled, phase 2b clinical trial. The Lancet. 2024 Feb 13;403(10429):838-849.
111. Rech J, Tascilar K, Hagen M, Kleyer A, Manger B, Schoenau V, et al. Abatacept inhibits inflammation and onset of rheumatoid arthritis in individuals at high risk (ARIAA): a randomised, international, multicentre, double-blind, placebo-controlled trial. The Lancet. 2024 Mar 2;403(10429):850-859.
112. Calatayud E, Montomoli M, Ávila A, Sancho Calabuig A, Alegre-Sancho JJ. Experience with abatacept in refractory lupus nephritis. Rheumatology International. 2023 Dec;43(12):2319-26.
113. Chen P, Zhou Y, Wu L, Chen S, Han F. Efficacy and safety of biologic agents for lupus nephritis: a systematic review and meta-analysis. JCR: Journal of Clinical Rheumatology. 2023 Mar 1;29(2):95-100.
114. Brunner HI, Tzaribachev N, Louw I, Calvo Penades I, Avila‐Zapata F, Horneff G, et al. Long‐Term Maintenance of Clinical Responses by Individual Patients With Polyarticular‐Course Juvenile Idiopathic Arthritis Treated With Abatacept. Arthritis Care & Research. 2023 Nov;75(11):2259-66.
115. Lovell DJ, Tzaribachev N, Henrickson M, Simonini G, Griffin TA, Alexeeva E, et al. Safety and effectiveness of abatacept in juvenile idiopathic arthritis: results from the PRINTO/PRCSG registry. Rheumatology. 2024 Jan 18:keae025.
116. Tuttle J, Drescher E, Simón-Campos JA, Emery P, Greenwald M, Kivitz A, et al. A phase 2 trial of peresolimab for adults with rheumatoid arthritis. New England Journal of Medicine. 2023 May 18;388(20):1853-62.
117. Mullard A. PD1 agonist antibody passes first phase II trial for autoimmune disease. Nature reviews. Drug Discovery. 2023 Jul;22(7):526.
118. Luu K, Dahl M, Hare E, Sibley C, Lizzul P, Randazzo B. DOP81 Rosnilimab, a novel PD-1 agonist monoclonal antibody, reduces T cell proliferation, inflammatory cytokine secretion, and PD-1high expressing CD4 and CD8 T cells: Results from a Phase 1 healthy volunteer clinical trial. Journal of Crohn's and Colitis. 2024 Jan 1;18(Supplement_1):i226.