Cytokine xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Cytokine journal homepage: www.journals.elsevier.com/cytokine

Review Article

T cell exhaustion and Interleukin 2 downregulation Mumtaz Y. Balkhi a,⇑, Qiangzhong Ma a, Shazia Ahmad b, Richard P. Junghans a a b

Department of Medicine, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, United States Boston University Medical Center, Boston, MA, United States

a r t i c l e

i n f o

Article history: Received 2 October 2014 Received in revised form 13 November 2014 Accepted 14 November 2014 Available online xxxx Keywords: T cells Exhaustion Interleukin 2 Chromatin PD1

a b s t r a c t T cells reactive to tumor antigens and viral antigens lose their reactivity when exposed to the antigenrich environment of a larger tumor bed or viral load. Such non-responsive T cells are termed exhausted. T cell exhaustion affects both CD8+ and CD4+ T cells. T cell exhaustion is attributed to the functional impairment of T cells to produce cytokines, of which the most important may be Interleukin 2 (IL2). IL2 performs functions critical for the elimination of cancer cells and virus infected cells. In one such function, IL2 promotes CD8+ T cell and natural killer (NK) cell cytolytic activities. Other functions include regulating naïve T cell differentiation into Th1 and Th2 subsets upon exposure to antigens. Thus, the signaling pathways contributing to T cell exhaustion could be linked to the signaling pathways contributing to IL2 loss. This review will discuss the process of T cell exhaustion and the signaling pathways that could be contributing to T cell exhaustion. Ó 2014 Elsevier Ltd. All rights reserved.

1. T cell exhaustion T cells are a central component of an adaptive immune system that is indispensable for immune system maintenance and human health. A principal function of T cells is immune surveillance to eliminate transformed cells and suppress virus infection. There are two processes, correlated, by which cognate T cells may lose their ability to react to antigens, anergy and exhaustion. Anergy is defined in terms of T cells receiving insufficient antigen activation and co-stimulation [1,2]. Exhaustion is a process in which T cells reactive to tumor antigens or viral antigens lose their ability to kill cancer cells or virus infected cells upon chronic antigen exposure. Antigen reactive T cells may be particularly vulnerable to exhaustion during immune surveillance, as reactive T cells may enter antigen rich-environment and be repeatedly stimulated triggering exhaustion. Exhaustion affects both long lived central memory and short lived effector memory T cells with debilitating consequences over immune functions. As, upon recurrent exposure to viruses and tumor antigens, the memory T cell pool loses its ability to reactivate. T cell exhaustion is characterized by a downregulation of effector functions including diminished cytolytic activity and type I cytokine secretion [3–6] and an upregulation in the expression of inhibitory molecules such as programmed death receptor 1 ⇑ Corresponding author at: Tufts Medical Center, 800 Washington, St. #817 Boston, MA 02111, United States. Tel.: +1 617 636 8247; fax: +1 617 636 8516. E-mail address: [email protected] (M.Y. Balkhi).

(PD1), cytotoxic T-lymphocyte antigen 4 (CTLA4), T cell immunoglobulin and mucin domain-containing molecule 3 (TIM3), lymphocyte activation gene 3 (LAG3), CD244 (2B4) and shift in the cytokine expression to IL10 and TGFb production that mark a suppressed state [7–9]. The gene profiling studies have demonstrated that the molecular profile of exhausted T cells in malignancy is similar to the one observed in chronic viral infections [10]. For example, tumor infiltrating CD8+ T cells have been shown to express inhibitory receptors such as PD1, BTLA, Tim3, Lag3, CTLA4 and shift in their cytokines expression, that is similar to molecular profiles in T cells exposed to chronic viral infections [2]. The T cell exhaustion to tumor cells is exacerbated by the presence of inhibitory ligands on tumor cells, regulatory T cells and immunomodulatory enzymes in tumor microenvironment [11]. The molecular details of exhaustion are not fully characterized yet, thus, making its distinction difficult from the other forms of T cell dysfunction such as anergy. However, kinetics of calcium signaling, constitutive MAPK and ERK signaling pathways could be distinctive features of exhaustion. The gene transcription profiles of exhausted T cells are distinct from anergic, memory and terminally differentiated T cells [2,12,13]. A core set of transcription factors such as Blimp1, Eomes, T-bet, IKZF, and Foxo can distinguish these T cell subsets (Table 1). Crawford et al. demonstrated recently that the pattern of expression of these transcription factors along with expression of inhibitory molecules and type I cytokine expression could distinguish exhausted CD4+ T cells from the effector and memory T cells [14].

http://dx.doi.org/10.1016/j.cyto.2014.11.024 1043-4666/Ó 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

2

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

Table 1 Distinct transcription factor profile of T cell subtypes. Characteristics

Terminally differentiated T cells

Exhausted T cells

Anergic T cells

Memory T cells

Transcription factors

EOMES, T-bet, BLIMP1, THPOK, KLRG1

BATF, Eomesodermin, BLIMP1, IKZF2, Eomes

GRAIL, CBL-B, ITCH, NEDD4

Foxo1, BCL-2

Table 2 Unanswered questions about T cell exhaustion. No.

Unanswered questions about T cell exhaustion

1 2 3 4 5 6

Whether exhausted T cells represent a distinct differentiation state or exhaustion represent dysfunctional CD8+ and CD4+ subtypes Whether T cell exhaustion arises to certain types of viruses and tumor-antigens only or a process is more common to viruses and tumor-antigens Signaling pathways contributing to the transcriptional changes associated with T cell exhaustion Apart from T cells of Th1/Th2 lineage, can other cell types such as Treg, Th17, Tfh or antigen presenting cells be exhausted Mechanism leading to loss of the IL2 production and other cytokines in exhausted T cells Whether T cell exhaustion is genetically regulated or only acquired through chronic infections and in malignancy

T cell exhaustion is a reversible state. Several studies have demonstrated that exhausted tumor reactive T cells can be recovered functionally in metastatic melanomas during immunotherapy [15]. Such T cells begin to produce IL2, IFNc and TNFa cytokines and the activation of STAT1, STAT5 and ERK1/2 signaling pathway which are crucial for cell survival and cytokine synthesis [16,17]. An antibody mediated blockade of Tim3 and PD1 partially reverses T cell exhaustion to colon carcinoma tumors in mice [18]. Likewise, an antibody mediated blockade of PD1 receptor or ligand alone or in combination with inhibition of IL10 in early clinical trials has shown promise in reversing T cell exhaustion and promoting anti-viral responses against LCMV infection [19–22]. Despite progress, much remains to be elucidated about the mechanism of T cell exhaustion (Table 2). In the following sections we will discuss various aspects of exhaustion. 1.1. T cell exhaustion in virus infections The phenomenon of T cell exhaustion was first observed in CD8+ T cells of immune-competent mice upon chronic infection with noncytopathic lymphocytic choriomeningitis virus (LCMV) [5,23]. Two separate studies by Richter et al., and Ahmed et al., showed that antigen presenting cells play central role in T cell exhaustion. Antigen presentation and the relative amount of antigen presented through MHC class I play critical role in the process of T cell exhaustion [24,25]. Whether high viral load through continuing replication of virus within the cells is sufficient to induce T cell exhaustion or additional mechanisms such as impairment of CD4 and NK cell activities are needed is still unclear [26]. Apart from LCMV, T cell exhaustion is also observed for many different chronic viruses including hepatitis C virus, hepatitis B Virus, HIV, and HTLV, etc. [27,28]. However, no such T cell exhaustion was observed for gamma herpes viruses such as cytomegalovirus and Epstein-Barr virus which persist in infected host indefinitely [29], confirming that T cell exhaustion does not arise to all viruses. The viral load is consistently high in settings with exhaustion [24,25], and this along with differences in antigen presentation, may distinguish those viral infections that induce exhaustion and those that do not [24,30]. Robust T cell activity can be recovered in exhausted T cells upon depletion of virus or antigens from the body, indicating that viral load play major role in the process of exhaustion [31]. 1.2. T cell exhaustion in parasitic and bacterial infections T cell exhaustion is reported in parasitic infections. Malaria is caused by zoonotic parasite plasmodium that during blood stage infection induces exhaustion of T cells [32,33]. Enhanced expression of PD1 in CD4+ and CD8+ T cells is reported in patients

infected with malaria and in experimental rodent malaria model [33,34]. In murine model of blood stage malaria, combined blockade of PDL-1 and LAG3 has shown enhanced clearance of Plasmodium yoelii and Plasmodium chabaudi [35]. Likewise, exhaustion related phenotype is also reported in murine and canine leishmaniasis. Leishmania infantum is a protozoan parasite that causes visceral leishmaniasis (VL). In canine leishmaniasis and murines infected with Leishmania major strain, CD4+ and CD8+ T cell express PD1 exhaustion markers and show impaired IFNc synthesis [36,37]. Likewise, CD8+ T cells undergo exhaustion in diffuse cutaneous leishmaniasis caused by Leishmania mexicana [38]. Toxoplasmosis is another case in which T cell undergoes exhaustion. It is caused by a protozoan Toxoplasma gondii (T. gondii). Infection of mice with T. gondii has revealed exhaustion of CD8+ T cells [39]. These T cells express PD1 in later phase of chronic toxoplasmosis and blockade of PD1/PDL-1 control Toxoplasma reactivation [40]. Exhaustion in bacterial infections is also reported. For example, higher expression of PD1 and its ligand have been reported in PBMCs of patients infected with active Mycobacterium tuberculosis antigens. While as blockade of PD1 in vitro in T cells derived from M.tb infected patients resist apoptosis and decline in IL2 and IFNc synthesis [41]. However, these results are inconsistent with the findings made in mice model of M.tb infection. A study by Reiley et al., has revealed in mice M.tb infection model that a fraction of antigen specific CD4+ T though express PD1 and KLRG1 markers but does not undergo exhaustion as evidenced by the continued production of IFNc and TNFa [42]. T cell exhaustion is also reported during gut bacterial infections of patients suffering from rare genetic disease, common variable immunodeficiency (CVID). The PD-1-PD-L1/2 blockade in vitro reactivated the T cells of CVID patients [43]. 1.3. T cell exhaustion in malignancy Tumor antigens are broadly divided into tumor associated antigens (TAA) and tumor specific antigens (TSA). TAA are self-antigens expressed on tumor cells and normal cells. Three means have been proposed by which self-antigens can become tumor associated antigen; (1) through overexpression of peptides because of mutations or epigenetic changes in the regulatory elements of the genes, (2) due to mutations or recombination in the genes itself leading to overexpression of proteins, (3) due to posttranslational modification of a peptides such as phosphorylation, glycosylation, and acetylation, leading to abnormal processing and presentation by the MHC class I [44]. Examples include, CEA, Her2/Neu, Mucin, vimentin, alpha-catenin and melanoma antigens, tyrosinaserelated protein-1 and tyrosinase-related protein-2, Survivin, and NY-ESO-1 etc [45–48]. Failure of the immune system to suppress tumors expressing TAA is attributed to the phenomenon of

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

3

Table 3 Distinct features of tolerance and exhaustion. No.

T cell exhaustion

T cell tolerance

1

Arises against chronic tumor and viral antigens

2

Exhaustion does not require any tissue specific antigen presentation. Phenotypically exhausted T cells could be T-bethi PD1int or Emoes hiPD1hi [2] Inhibitory molecules such as PD1, TIM3, LAG3, CTLA4, B- and T-lymphocyte attenuator (BTLA) [134], BLIMP1 are commonly expressed Cytokines such as IL2, TNFa, IFNc are lost in hierarchical order IL10 and TGFb cytokines are synthesized Immunosuppressive microenvironment such as the presence of Treg, MDSCs, galectin 3, and iNOS may play role in inducing T cell exhaustion

Arises against self-antigens and is an important mechanism to prevent autoimmunity (central tolerance). Commonly observed for tumor antigens that are self-antigens Lymph nodes and spleen are the primary sites of tumor antigen presentation for T cell tolerance induction [131,132]. APCs play a major role in T cell tolerance. Such T cells may be CD44hi, CD69int, CD25, CD62Lint, CD49dlo [133] Inhibitory molecules such as PD1 and LAG3 are commonly expressed

3 4 5 6

tolerance. Two principal mechanisms of tolerance operate in T cells exposed to TAAs. In one mechanism, if T cells are strongly activated (i.e., with high avidity TCRs) against tumor associated antigens presented by the MHC, there is a chance that T cells will attack the normal tissue which will be detrimental to the host. These T cells are typically deleted in the thymus (negative selection) [49,50]. In a second mechanism, weakly activated T cells (i.e., with low avidity TCR to tumor associated antigens) will survive in the host if they escape deletion (positive selection). These T cells under persistent tumor antigens will become anergic due to lack of co-stimulation. These anergic T cells express inhibitory molecules such PD1, Lag3 and secretion of IFNc declines contributing to the overall decline in anti-tumor immunity [50]. In malignancy, both tumor cells and immature dendritic cells fail to provide optimal co-stimulations to the T cells [51]. Tumor specific antigens (TSA) are the antigens that are uniquely expressed by tumors. TSA are more likely to be immunogenic generating tumor-antigen specific T cells. Some examples of TSA include viral oncoproteins, HTLV virus Tax oncoprotein, HPV oncogenes E6 and E7, SV40 tumor (T) antigen, mutated p53, oncogenic mutant Ras, etc. [50,52]. Immunogenic fusion oncoproteins, thyroid tumor specific antigen rearranged during transfection/papillary thyroid carcinoma (RET/PTC) fusion protein and Chronic myelogenous leukemia (CML) specific BCR-ABL [53–55]. Cancer germline genes, melanoma antigens (MAGE-1) and somatic tumor mutations also termed as neo-antigens such as BRAFV600E, and CDKR24C [56,57], are some other examples of tumor specific antigens [58]. Exhaustion is likely to arise in T cells reactive to immunogenic tumor specific antigens under the settings of antigen-rich environment [56]. For example, exhaustion of the leukemia antigen specific cytotoxic T lymphocytes was observed in the murine models of retroviral induced chronic phase CML and blast crisis

Cytokines such as IL2, TNFa, IFNc are lost. IFNc loss is critical for tolerance induction Expresses IL15 receptor alpha chain [135] Immunosuppressive tumor microenvironment such as the presence of CD11b+Gr-1+ MDSc play prominent role in T cell tolerance [136]

CML [59]. Exhaustion is also reported to occur to metastases melanoma antigen, Melan-A/MART-1 [16], suggesting that the process of exhaustion could be more related to presentation and antigenicity of tumor antigens [57,60]. Several algorithms are now available that can be used to predict the antigenicity of tumor antigens [61–63]. This information can be used to examine the possible exhaustion of T cells in patients presented with immunogenic tumor antigens. Important distinctions between tolerance and exhaustion are provided (Table 3).

2. Interleukin 2 downregulation during T cell exhaustion Changes in the cytokines synthesis constitute a major component of the exhaustion phenotype. During T cell exhaustion, loss of secretion of IL2, IFNc and TNFa occurs in hierarchical manner [9]. One of the most important cytokines whose production is lost during T cell exhaustion is IL2. IL2 is a pivotal cytokine for T cell survival and for activating a robust cellular immune response against infections and transformation. The role of IL2 in activating T cell response against tumor cells is depicted schematically (Fig. 1A). The schematics describe if IL2 synthesis is inhibited under antigen persistence, the overall antitumor response declines, resulting in tumor progression (Fig. 1B). It is reported extensively that exogenous IL2 infusion can enhance destructive immune responses against tumors, both in adoptively transferred tumor reactive T cell and gene modified T cells [15,64–66]. However, addition of exogenous IL2 does not by itself support long term antigenic response and survival [67,68]. In adoptive T cell transfers, T cells capable of sustaining IL2 production after coming in contact with reactive-antigens may produce stronger antigenic responses, thus, helping to break T cell exhaustion.

Fig. 1. (A) Schematics illustrate the crucial role of IL2 in antitumor response. (B) Illustrates negative impact of IL2 shutdown on overall antitumor response.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

4

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

The signaling pathways, both stimulatory and inhibitory, to the IL2 transcription have been extensively studied and new knowledge is constantly being added. The knowledge about these signaling pathways could provide vital clues about dysregulation of the IL2 synthesis during exhaustion. T cell receptor (TCR) mediated signaling pathway and CD28 costimulatory signaling are important for the IL2 transcription. These two signaling pathways are briefly described here. TCR signaling pathways: Upon ligation of MHC molecules loaded with antigenic peptides with TCR-CD3 complex, the first event to occur within T cells is the phosphorylation of tyrosine residues within cytoplasmic side of immunoreceptor tyrosine-based transactivation motifs (ITAMs) of TCR associated CD3 molecules by Src family tyrosine-protein kinases, lymphocyte protein tyrosine kinase (LCK) and Fyn [69]. Both LCK and Fyn undergo phosphorylation in the process, the mechanism of which is not completely understood. However, LCK is also activated by the interaction of CD4 and CD8 molecules with MHC class II [70,71]. Phosphorylated tyrosine residues of ITAM of CD3 zeta chains serve as a docking site for the zeta-chain associated protein tyrosine kinase (ZAP70). Phosphorylation in the tyrosine residues Tyr319/Tyr352 of ZAP70 amplifies the signal by promoting the recruitment of membrane proximal SH2 domain-containing leukocyte protein-76 (SLP76) scaffold protein complex, membrane integral protein linker for activation of T cells (LAT) and IL2 inducible T-cell kinase (ITK) [72]. Phosphorylation of LAT at tyrosine residues Tyr171 and Tyr191 by ZAP70 enables the binding and recruitment of SLP76, PLCc1 and GRB2 adaptor protein complex that also includes Gads/SOS/VAV. Phosphorylation of Vav1 stimulates guanine nucleotide exchange factors (GEFs) for CDC42-Rac molecules activating MEKK [73–75]. MEKK phosphorylates MKK3/6 and MKK4/7. Phoshpho-MKK3(Ser189)/MKK6(Ser207) and phospho-MKK4(Ser257/Thr261) activates p38MAPK and cJunN-terminal kinase (JNK) pathways, respectively [76,77]. Phosphop38MAPK(Thr180/Tyr182) in turn activates cJun transcription factor. Phospho-JNK (Thr183/Tyr185) activates transcription factor Fos and ATF2 [78,79]. The PLCc1 hydrolysis Phosphotidylinositol-4, 5 Biphosphate (PIP2) into inositol-1, 4, 5 triphosphatase (IP3) and diacyl glycerol (DAG). Second messenger DAG activates and recruits RAS-GRP and PKCh to the plasma membrane. Ras-GRP activates RAS, RAF, MEK1/2 and ERK kinase cascade activating transcription factor Fos. Fos heterodimerizes with cJun to form AP1 transcription factor that activates IL2 transcription by binding to the AP1 consensus binding site, ATGAGTCAT, of IL2 gene [3,80–83]. The IP3 binds to the IP3 receptor on the endoplasmic reticulum triggering calcium release from the ER. The two protein families, the stromal interacting molecules-(STIM) 1 and 2, functions as sensors of calcium ion levels in the ER. The depletion of calcium stores in the ER allow STIM proteins to migrate to the plasma membrane where they interact with Orai family proteins. This interaction is essential for the store-operated calcium entry through activating CRAC channels [84]. Influx of extracellular calcium ions into the T cells is sensed by calmodulin (CaM). CaM activates calcineurin phosphatase (CaN) by binding to its regulatory domain (RD) and displacing an autoinhibitory domain (AID) from the active site [85,86]. Activated calcineurin subsequently activates NF-AT transcription factor by dephosphorylating serine residues, allowing NF-AT to translocate to nucleus to initiate the IL2 gene transcription. NF-AT cooperates with AP1 to form dimers to bind to the NF-AT consensus sites GGAGGAAAAA or on several composite NF-AT-AP1 sites on IL2 promoter [87,88]. CD28 costimulatory signaling pathway: CD28 generated signaling pathway engages molecules that are partially distinct from TCR-CD3 receptor complex. NF-jB is the main transcription factor that is activated upon CD28 costimulation through the signaling

pathway that is yet to be completely defined. NF-jB binds to a sequence on the IL2 promoter, the CD28 response element, to activate IL2 transcription. Evidences also exist that support the integration of CD28 and TCR-CD3 signaling pathway by PDK1 molecule and Grb2 [89,90]. Ligation of CD28 extracellular domain motif comprised of a conserved sequence, MYPPPY, with B7 costimulatory molecules of the antigen presenting cells initiates CD28 costimulatory signaling cascade [91]. Phosphorylation of tyrosine residues within the short cytoplasmic domain of CD28 by the Src family kinases LCK and Fyn act as a docking site for p85 SH2 domain of phosphatidylinositol 3-kinase (PI3K) and Grb2 [92]. CD28 binds with PI3K through YMNM motif. As reported in mice the phosphorylation of PI3K on Tyr173 during CD28/B7.1 ligation is crucial for IL2 synthesis [93]. T cell expresses four different isoforms of PI3K, each capable of catalyzing PIP2 to PIP3 (PtdIns (3,4,5) P3) [94]. The requirement of second messenger PIP3 for further downstream signaling for the IL2 transcription is still unclear. Grb2-CD28 binding is required for the phosphorylation and activation of Vav1 for relaying and integrating signaling to the Rac1-CDC42 [95]. Another molecule that has gained much attention recently for providing distinct signals to the NF-jB during CD28 costimulation is Akt. Several studies have reported the activation of Akt during CD28 costimulation and its requirement for the IL2 synthesis [96,97]. CD28 family signaling pathways inhibitory to IL2 transcription: The signaling pathways generated by CD28 family molecules also block T cell activation. There are several CD28 family members chief among them CTLA4 and PD1 that have proven inhibitory to the T cell activation. The mechanism by which these molecules deliver coinhibitory signals to limit T cell response is not completely understood. However, the activation of phosphatases such as SHP2 and PP2A have been reported and those are believed to generate inhibitory signals [98]. CTLA4 uses YVKM motif to bind with PI3K, SHP2 and PP2A [90,91]. Likewise, PD1 ligation is reported to block PI3K activity and Akt response [99]. How exactly these varied signals integrate and influence IL2 synthesis and contribute to the exhaustion is yet to be discovered. 2.1. Epigenetic and posttranscriptional regulation of IL2 during exhaustion One of the reasons for gene repression or cellular inactivation is the epigenetic changes associated with the chromatin such as phosphorylation, methylation and acetylation. Several studies have shown that specific epigenetic modifications of chromatin represent distinct differentiation states of T cell subtypes such as naïve, effector and memory T cells [100–102]. However, it remains to be investigated whether exhausted T cells are presented with specific epigenetic signature and whether such epigenetic marks are conserved in T cells encountering malignancy and chronic viral infections. Along with the epigenetic regulation, posttranscriptional gene regulation could represent another mechanism of regulating IL2 mRNA metabolism during T cell exhaustion (Fig. 2). Mature microRNAs are class of small naturally occurring non-coding RNA molecules that extensively regulate protein coding genes by targeting mRNA 30 UTRs. More than 1000 microRNAs have been discovered. A single microRNA can regulate multiple genes due to their ability to pair with mRNAs with partial complimentary in Watson–Crick base pairing mechanism. MicroRNA targeting of mRNA can result in either mRNA decay, deadenylation or translational repression in RNA-induced silencing complex (RISC) [103]. The role of microRNAs in T cell activity and lineage differentiation has been extensively reported [104,105]. In a bioinformatics screen, Targetscan and miRANDA databases predicted a perfect seed match for human IL2 30 UTR with miR-181a (Fig. 3). The data suggest that miR 181a could be regulating IL2 mRNA stability

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

5

Fig. 2. Schematics illustrate the possible involvement of chromatin remodeling complex in IL2 gene transcription (Upper panel). Lower panel demonstrates the posttranscriptional regulation of IL2 mRNA through microRNAs and RNA binding proteins during T cell exhaustion.

Fig. 3. Bioinformatics Targetscan analysis predicted miR-181a as a potential microRNA targeting IL2 30 UTR.

during T cell activation. This mechanism is supported by findings that show miR 181a expression in mature T cells is required for antigen sensitivity that regulates the positive and negative selection process for T cell tolerance to self antigens [104]. RNA binding proteins represent a class of proteins that specifically bind to mRNAs and play a prominent role in the posttranscriptional gene regulation. In a bioinformatics screen, RNA binding protein PUM2 turns out to be a predicted target for IL2 mRNA (Table 4). Human Pumilio Proteins have been reported to recruit multiple deadenylases to efficiently repress messenger RNAs [106]. PUM1 and PUM2 also known as PUMILIO proteins have been shown to cause repression of p27 mRNA through microRNA dependent gene silencing in rapidly proliferating cells [107]. Several other genes including hunchback (mat) mRNA have also been shown to be repressed by binding of PUM2. The signaling pathways regulating IL2 shutdown during T cell exhaustion are unknown. One interesting link could be the association of PD1/PDL-1 signaling pathway with IL2 transcription. The rationale for this model is derived from the studies that have demonstrated elevated expression of PD1 in tumor infiltrating T lymphocytes and the higher levels of PD1 ligand (PDL-1) on tumors. The PD1/PDL-1 interaction is associated with exhaustion phenotype and impaired cytokine production including that of IL2 [108,109]. Experimental evidence shows that PD1 activation through its ligand PDL-1/2 inhibits PI3K activation by activating src-homology phosphatases (SHP2) [110]. A relevant question to ask is whether PI3K inhibition could block IL2 transcription. It is possible, due to the PI3K inhibition; T cells do not receive sufficient CD28 costimulatory signals resulting in decline of the IL2 production. Lack of IL2 production in turn could render PI3K inactivation persistent because PI3Ks are strongly activated not only through IL2 receptor signaling but also by cytokines such as IL15, IL3, IL6, and interferons (IFNs), creating a cumulative effect contributing to T cell exhaustion [111,112]. A model is proposed that describes the relation between PD1 signaling and IL2 transcription (Fig. 4). Table 4 Pum2 scored 100% coverage for IL2 30 UTR binding. Score

Relative score

RBP name

Start

End

Matching sequence

7.2294196 7.2294196 7.2294196 7.2294196

100% 100% 100% 100%

Pum2 Pum2 Pum2 Pum2

132 113 188 272

135 116 191 275

UGUA UGUA UGUA UGUA

Fig. 4. Model depicting the signaling pathway involving PD1 mediated regulation of IL2 transcription.

The observation of development of spontaneous autoimmune diseases in PD1, PDL-1 and IL2 knockout mice provides a rationale that PD1 signaling could be regulating the IL2 transcription [113–116]. Thus, PD1 signaling may be contributing significantly to decline of IL2 production and to the process of T cell exhaustion [113]. Further research into this direction will reveal in-depth of understanding of the mechanism of T cell exhaustion. 3. Prevention of T cell exhaustion and its therapeutic applications As T cells play critical role in virus clearance and tumor rejection, there has been considerable interest in preventing or reversing T cell exhaustion during chronic viral infection and malignancy. For example, PD1/PDL-1 blockade reverses CD8+ T cell exhaustion to chronic virus infections [19]. An antibody mediated blockade of PD1 or PDL-1 molecules restored T cell responses against tumor-antigens and demonstrated promising clinical benefits for melanomas [117,118]. Doedens et al., showed recently

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

6

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

that genetically inactivating the tumor suppressor VHL (von Hippel-Lindau), which is a negative regulator of hypoxia-inducible factor (HIF) signaling pathway, is able to prevent exhaustion. Moreover, CD8+ T cells were able to maintain potent effector functions while encountering persistent melanoma tumor antigens and chronic LCMV Armstrong virus [119]. Our lab is currently investigating whether rescuing IL2 production or opposing IL2 decline in T cells exposed to chronic tumor antigens could reverse the T cell exhaustion (Balkhi et al.; unpublished data). T cell adoptive immunotherapy is a promising tool for cancer cure. It has been reported that T cell exhaustion not only affects T cells encountering persistent tumor antigens in a body but also ex vivo expanded therapeutic T cells such as tumor infiltrating T cells (TILs). TILs are tumor infiltrated T cells that are isolated from tumors. TILs are extensively expanded ex vivo and infused back into cancer patients to generate anti-tumor reactivity. However, exhaustion limits the potential of TILs to suppress tumors. Exhaustion in TILs has been demonstrated in human prostate cancer patients and during metastases and in mice bearing solid tumors [16,18,109,120]. Exhaustion is also reported in chimeric antigen receptor modified T cells (CAR-T) [121,122]. CAR-T cells are generated by genetically modifying T cell receptor with immunoglobulin fusion products of an antibody binding domain (immunoglobulin, Ig) with the zeta signaling chain of the TCR and CD28 costimulatory molecule sequences, to form Immunoglobulin chimeric antigen receptor (IgCAR), which redirects T cells for cancer or viral antigen specific cytotoxicity [123–125]. The single chain variable fragment (scFV) antibody binding domain of CAR is specific of tumor associated antigens. On tumor contact T cell chimeric antigen receptor ‘‘IgCAR with the fusion of CD28’’ yields signal 1 + 2. Signal 1 + 2 is important for the full activation of T cells and IL2 synthesis. The design of CAR-T cells is represented sche-

matically in Fig. 5. The design of CAR-T cells, apart from CD28 molecule, has also included several other costimulatory molecules to improve antitumor activity. For example, CAR-T cells modified with B cell specific CD19 antigen and costimulatory molecule CD137 (4-1BB) have shown success in clearing chronic lymphocytic leukemia [64]. However, one factor that could limit designer T cell efficacy is their failure to sustain IL2 production in vivo and in patients as reported by Emtage et al. [121,122]. IL2 shutdown is consistent feature in antigen-specific T cells and gene modified T cells. In-depth understanding of the mechanism of IL2 shutdown in T cells exposed to chronic antigens will significantly help improving the design of CAR-T cells in a way that T cells maintain long term antigenic responses and promote powerful antitumor or antiviral T cell responses. 4. Future directions A global approach to identify epigenetic modifications associated with the chromatin in T cells at various stages of differentiation including exhaustion could be very informative. Such an approach has already been tested successfully in activating and resting T cells [126,127]. A global approach to identify repressive histone marks on IL2 region using ChIP-Seq technique could be useful in understanding the mechanism leading to T cell exhaustion to persistent infections or in malignancy. This technique could be particularly useful in determining whether histones around IL2 genomic region ‘‘4q26-q27 Chromosome: 4; NC_000004.11 (123372625 to 123377650’’ undergoes chromatin modifications [128]. The binding of high mobility group proteins (HMGA1) to the CD28 response element has been reported to promote interaction of NF-jB, NF-AT, and AP1 at this region [129]. The HMGA1 putative binding site on CD28 response element lies in close proximity to TATA binding protein, TFIID. Proximity to these sites makes all these transcription factors suitable candidates to influence gene expression [130]. It could be potentially interesting to investigate how these factors influence IL2 shutdown in T cells exposed to persistent antigens. The role of microRNAs and RNA binding proteins in the regulation of IL2 and T cell exhaustion in malignancy and in chronic virus infections is not commonly described. It could be interesting to determine the expression levels miR-181 in T cells infiltrating tumor antigens. This will allow exploring in vitro whether modulating miR-181 expression produces effect on the IL2 production. Likewise, investigating the expression levels of ‘‘PUMA’’ RNA binding protein in T cells infiltrating tumors or exploring its significance for T cell exhaustion is potentially interesting. Likewise, investigating whether PUMA and miR-181a cooperate to influence the stability of IL2 mRNA could be vital for understanding the mechanism of IL2 downregualtion in exhaustion. 5. Conclusions The in-depth understanding of the mechanism of IL2 decline in T cells exposed to chronic antigens will help genetically modify T cells, so that when they are exposed back to an antigen-rich environment could resist IL2 decline. Maintaining constant production of the IL2 in antigen-rich environment will allow T cell to harness powerful antigenic responses. Thus, the identification of the signaling pathways involved in the IL2 downregulation and posttranscriptional regulation of the IL2 mRNA could prove vital in making breakthrough improvements in T cell immunotherapy.

Fig. 5. Structure of T cell chimeric antigen receptor (IgCAR). Upper panel: Tumor antigen specific Ab binding domain is cloned together with TCR zeta chain and co-stimulatory CD28 molecule and packaged in retroviral vector and subsequently expressed in T cells. Lower: Designer T cells expressing IgCAR.

Acknowledgement This work is supported by Breast Cancer Research Program, Department of Defense.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

References [1] Schwartz RH. T cell anergy. Annu Rev Immunol 2003;21:305–34. [2] Schietinger A, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol 2014;35:51–60. [3] Liao W, Lin JX, Leonard WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 2013;38:13–25. [4] Yi JS, Cox MA, Zajac AJ. T-cell exhaustion: characteristics, causes and conversion. Immunology 2010;129:474–81. [5] Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med 1998;188:2205–13. [6] Fuller MJ, Khanolkar A, Tebo AE, Zajac AJ. Maintenance, loss, and resurgence of T cell responses during acute, protracted, and chronic viral infections. J Immunol 2004;172:4204–14. [7] Grosso JF, Goldberg MV, Getnet D, Bruno TC, Yen HR, Pyle KJ, et al. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J Immunol 2009;182:6659–69. [8] Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A 2010;107:7875–80. [9] Wherry EJ. T cell exhaustion. Nat Immunol 2011;12:492–9. [10] Speiser DE. A molecular profile of T-cell exhaustion in cancer. Oncoimmunology 2012;1:369–71. [11] Atkins MB, Elder DE, Essner R, Flaherty KT, Gajewski TF, Haluska FG, et al. Innovations and challenges in melanoma: summary statement from the first Cambridge conference. Clin Cancer Res 2006;12:2291s–6s. [12] Kim MV, Ouyang W, Liao W, Zhang MQ, Li MO. The transcription factor Foxo1 controls central-memory CD8+ T cell responses to infection. Immunity 2013;39:286–97. [13] Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol 2012;12:749–61. [14] Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, et al. Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 2014;40:289–302. [15] Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011;17:4550–7. [16] Baitsch L, Baumgaertner P, Devevre E, Raghav SK, Legat A, Barba L, et al. Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. J Clin Invest 2011;121:2350–60. [17] Baumgaertner P, Jandus C, Rivals JP, Derre L, Lovgren T, Baitsch L, et al. Vaccination-induced functional competence of circulating human tumorspecific CD8 T-cells. Int J Cancer 2012;130:2607–17. [18] Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010;207:2187–94. [19] Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439:682–7. [20] Brooks DG, Ha SJ, Elsaesser H, Sharpe AH, Freeman GJ, Oldstone MB. IL-10 and PD-L1 operate through distinct pathways to suppress T-cell activity during persistent viral infection. Proc Natl Acad Sci U S A 2008;105:20428–33. [21] Ha SJ, Mueller SN, Wherry EJ, Barber DL, Aubert RD, Sharpe AH, et al. Enhancing therapeutic vaccination by blocking PD-1-mediated inhibitory signals during chronic infection. J Exp Med 2008;205:543–55. [22] Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, Crotty S, et al. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp Med 2006;203:2461–72. [23] Moskophidis D, Lechner F, Pircher H, Zinkernagel RM. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 1993;362:758–61. [24] Richter K, Brocker T, Oxenius A. Antigen amount dictates CD8+ T-cell exhaustion during chronic viral infection irrespective of the type of antigen presenting cell. Eur J Immunol 2012;42:2290–304. [25] Mueller SN, Ahmed R. High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proc Natl Acad Sci U S A 2009;106:8623–8. [26] Day CL, Kiepiela P, Leslie AJ, van der Stok M, Nair K, Ismail N, et al. Proliferative capacity of epitope-specific CD8 T-cell responses is inversely related to viral load in chronic human immunodeficiency virus type 1 infection. J Virol 2007;81:434–8. [27] Klenerman P, Hill A. T cells and viral persistence: lessons from diverse infections. Nat Immunol 2005;6:873–9. [28] Goepfert PA, Bansal A, Edwards BH, Ritter Jr GD, Tellez I, McPherson SA, et al. A significant number of human immunodeficiency virus epitope-specific cytotoxic T lymphocytes detected by tetramer binding do not produce gamma interferon. J Virol 2000;74:10249–55. [29] Obar JJ, Fuse S, Leung EK, Bellfy SC, Usherwood EJ. Gammaherpesvirus persistence alters key CD8 T-cell memory characteristics and enhances antiviral protection. J Virol 2006;80:8303–15. [30] Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution

[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]

7

and results in distinct stages of functional impairment. J Virol 2003;77: 4911–27. Han S, Asoyan A, Rabenstein H, Nakano N, Obst R. Role of antigen persistence and dose for CD4+ T-cell exhaustion and recovery. Proc Natl Acad Sci U S A 2010;107:20453–8. Chandele A, Mukerjee P, Das G, Ahmed R, Chauhan VS. Phenotypic and functional profiling of malaria-induced CD8 and CD4 T cells during bloodstage infection with Plasmodium yoelii. Immunology 2011;132:273–86. Wykes MN, Horne-Debets JM, Leow CY, Karunarathne DS. Malaria drives T cells to exhaustion. Front Microbiol 2014;5:249. Horne-Debets JM, Faleiro R, Karunarathne DS, Liu XQ, Lineburg KE, Poh CM, et al. PD-1 dependent exhaustion of CD8+ T cells drives chronic malaria. Cell Rep 2013;5:1204–13. Butler NS, Moebius J, Pewe LL, Traore B, Doumbo OK, Tygrett LT, et al. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established bloodstage Plasmodium infection. Nat Immunol 2012;13:188–95. Esch KJ, Juelsgaard R, Martinez PA, Jones DE, Petersen CA. Programmed death 1-mediated T cell exhaustion during visceral leishmaniasis impairs phagocyte function. J Immunol 2013;191:5542–50. Mou Z, Muleme HM, Liu D, Jia P, Okwor IB, Kuriakose SM, et al. Parasitederived arginase influences secondary anti-Leishmania immunity by regulating programmed cell death-1-mediated CD4+ T cell exhaustion. J Immunol 2013;190:3380–9. Hernandez-Ruiz J, Salaiza-Suazo N, Carrada G, Escoto S, Ruiz-Remigio A, Rosenstein Y, et al. CD8 cells of patients with diffuse cutaneous leishmaniasis display functional exhaustion: the latter is reversed, in vitro, by TLR2 agonists. PLoS Negl Trop Dis 2010;4:e871. Bhadra R, Khan IA. Redefining chronic toxoplasmosis – a T cell exhaustion perspective. PLoS Pathog 2012;8:e1002903. Bhadra R, Gigley JP, Weiss LM, Khan IA. Control of Toxoplasma reactivation by rescue of dysfunctional CD8+ T-cell response via PD-1-PDL-1 blockade. Proc Natl Acad Sci U S A 2011;108:9196–201. Singh A, Mohan A, Dey AB, Mitra DK. Inhibiting the programmed death 1 pathway rescues Mycobacterium tuberculosis-specific interferon gammaproducing T cells from apoptosis in patients with pulmonary tuberculosis. J Infect Dis 2013;208:603–15. Reiley WW, Shafiani S, Wittmer ST, Tucker-Heard G, Moon JJ, Jenkins MK, et al. Distinct functions of antigen-specific CD4 T cells during murine Mycobacterium tuberculosis infection. Proc Natl Acad Sci U S A 2010;107: 19408–13. Perreau M, Vigano S, Bellanger F, Pellaton C, Buss G, Comte D, et al. Exhaustion of bacteria-specific CD4 T cells and microbial translocation in common variable immunodeficiency disorders. J Exp Med 2014;211: 2033–45. Finn OJ. Cancer immunology. N Engl J Med 2008;358:2704–15. Chin L, Hahn WC, Getz G, Meyerson M. Making sense of cancer genomic data. Genes Dev 2011;25:534–55. Preiss S, Kammertoens T, Lampert C, Willimsky G, Blankenstein T. Tumorinduced antibodies resemble the response to tissue damage. Int J Cancer 2005;115:456–62. Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour immunogenicity. Nat Rev Cancer 2012;12:307–13. Andersen RS, Thrue CA, Junker N, Lyngaa R, Donia M, Ellebaek E, et al. Dissection of T-cell antigen specificity in human melanoma. Cancer Res 2012;72:1642–50. Yu Z, Theoret MR, Touloukian CE, Surman DR, Garman SC, Feigenbaum L, et al. Poor immunogenicity of a self/tumor antigen derives from peptide-MHC-I instability and is independent of tolerance. J Clin Invest 2004;114:551–9. Mapara MY, Sykes M. Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. J Clin Oncol 2004;22:1136–51. Steinman RM, Nussenzweig MC. Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci U S A 2002;99:351–8. Helt AM, Galloway DA. Mechanisms by which DNA tumor virus oncoproteins target the Rb family of pocket proteins. Carcinogenesis 2003;24:159–69. Powell Jr DJ, Eisenlohr LC, Rothstein JL. A thyroid tumor-specific antigen formed by the fusion of two self proteins. J Immunol 2003;170:861–9. Cathcart K, Pinilla-Ibarz J, Korontsvit T, Schwartz J, Zakhaleva V, Papadopoulos EB, et al. A multivalent bcr-abl fusion peptide vaccination trial in patients with chronic myeloid leukemia. Blood 2004;103:1037–42. Cai A, Keskin DB, DeLuca DS, Alonso A, Zhang W, Zhang GL, et al. Mutated BCR-ABL generates immunogenic T-cell epitopes in CML patients. Clin Cancer Res 2012;18:5761–72. Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, et al. Neoantigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res 2014;24:743–50. Heemskerk B, Kvistborg P, Schumacher TN. The cancer antigenome. EMBO J 2013;32:194–203. van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991;254:1643–7. Mumprecht S, Schurch C, Schwaller J, Solenthaler M, Ochsenbein AF. Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression. Blood 2009;114: 1528–36.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

8

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx

[60] Lennerz V, Fatho M, Gentilini C, Frye RA, Lifke A, Ferel D, et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005;102:16013–8. [61] Bui HH, Sidney J, Peters B, Sathiamurthy M, Sinichi A, Purton KA, et al. Automated generation and evaluation of specific MHC binding predictive tools: ARB matrix applications. Immunogenetics 2005;57:304–14. [62] Peters B, Sidney J, Bourne P, Bui HH, Buus S, Doh G, et al. The immune epitope database and analysis resource: from vision to blueprint. PLoS Biol 2005;3:e91. [63] Vita R, Zarebski L, Greenbaum JA, Emami H, Hoof I, Salimi N, et al. The immune epitope database 2.0. Nucl Acid Res 2010;38:D854–62. [64] Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptormodified T cells in chronic lymphoid leukemia. N Engl J Med 2011;365: 725–33. [65] Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, et al. B-cell depletion and remissions of malignancy along with cytokineassociated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptortransduced T cells. Blood 2012;119:2709–20. [66] Lo AS, Ma Q, Liu DL, Junghans RP. Anti-GD3 chimeric sFv-CD28/T-cell receptor zeta designer T cells for treatment of metastatic melanoma and other neuroectodermal tumors. Clin Cancer Res 2010;16:2769–80. [67] Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, Downey S, et al. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2. Hum Gene Ther 2008;19:496–510. [68] Lotze MT, Matory YL, Ettinghausen SE, Rayner AA, Sharrow SO, Seipp CA, et al. In vivo administration of purified human interleukin 2. II. Half life, immunologic effects, and expansion of peripheral lymphoid cells in vivo with recombinant IL 2. J Immunol 1985;135:2865–75. [69] Davis MM. A new trigger for T cells. Cell 2002;110:285–7. [70] Molina TJ, Kishihara K, Siderovski DP, van Ewijk W, Narendran A, Timms E, et al. Profound block in thymocyte development in mice lacking p56lck. Nature 1992;357:161–4. [71] Van Laethem F, Tikhonova AN, Pobezinsky LA, Tai X, Kimura MY, Le Saout C, et al. Lck availability during thymic selection determines the recognition specificity of the T cell repertoire. Cell 2013;154:1326–41. [72] Hussain A, Mohammad DK, Gustafsson MO, Uslu M, Hamasy A, Nore BF, et al. Signaling of the ITK (interleukin 2-inducible T cell kinase)-SYK (spleen tyrosine kinase) fusion kinase is dependent on adapter SLP-76 and on the adapter function of the kinases SYK and ZAP70. J Biol Chem 2013;288: 7338–50. [73] Gong Q, Cheng AM, Akk AM, Alberola-Ila J, Gong G, Pawson T, et al. Disruption of T cell signaling networks and development by Grb2 haploid insufficiency. Nat Immunol 2001;2:29–36. [74] Margarit SM, Sondermann H, Hall BE, Nagar B, Hoelz A, Pirruccello M, et al. Structural evidence for feedback activation by Ras.GTP of the Ras-specific nucleotide exchange factor SOS. Cell 2003;112:685–95. [75] Kortum RL, Balagopalan L, Alexander CP, Garcia J, Pinski JM, Merrill RK, et al. The ability of Sos1 to oligomerize the adaptor protein LAT is separable from its guanine nucleotide exchange activity in vivo. Sci Signal 2013;6:ra99. [76] Raingeaud J, Gupta S, Rogers JS, Dickens M, Han J, Ulevitch RJ, et al. Proinflammatory cytokines and environmental stress cause p38 mitogenactivated protein kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem 1995;270:7420–6. [77] Kang YJ, Seit-Nebi A, Davis RJ, Han J. Multiple activation mechanisms of p38alpha mitogen-activated protein kinase. J Biol Chem 2006;281:26225–34. [78] Ichijo H. From receptors to stress-activated MAP kinases. Oncogene 1999;18:6087–93. [79] Freshney NW, Rawlinson L, Guesdon F, Jones E, Cowley S, Hsuan J, et al. Interleukin-1 activates a novel protein kinase cascade that results in the phosphorylation of Hsp27. Cell 1994;78:1039–49. [80] Alberola-Ila J, Forbush KA, Seger R, Krebs EG, Perlmutter RM. Selective requirement for MAP kinase activation in thymocyte differentiation. Nature 1995;373:620–3. [81] Cantrell DA. T cell antigen receptor signal transduction pathways. Cancer Surv 1996;27:165–75. [82] Chang F, Steelman LS, Lee JT, Shelton JG, Navolanic PM, Blalock WL, et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia 2003;17:1263–93. [83] Kim HP, Imbert J, Leonard WJ. Both integrated and differential regulation of components of the IL-2/IL-2 receptor system. Cytokine Growth Factor Rev 2006;17:349–66. [84] Putney JW. Pharmacology of store-operated calcium channels. Mol Interv 2010;10:209–18. [85] Ye Q, Feng Y, Yin Y, Faucher F, Currie MA, Rahman MN, et al. Structural basis of calcineurin activation by calmodulin. Cell Signal 2013;25:2661–7. [86] Rumi-Masante J, Rusinga FI, Lester TE, Dunlap TB, Williams TD, Dunker AK, et al. Structural basis for activation of calcineurin by calmodulin. J Mol Biol 2012;415:307–17. [87] Muller MR, Rao A. NFAT, immunity and cancer: a transcription factor comes of age. Nat Rev Immunol 2010;10:645–56. [88] Hogan PG, Chen L, Nardone J, Rao A. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev 2003;17:2205–32. [89] Park SG, Schulze-Luehrman J, Hayden MS, Hashimoto N, Ogawa W, Kasuga M, et al. The kinase PDK1 integrates T cell antigen receptor and CD28 coreceptor

[90] [91] [92]

[93]

[94] [95] [96]

[97] [98] [99]

[100] [101]

[102]

[103] [104] [105] [106]

[107] [108] [109]

[110]

[111] [112]

[113] [114]

[115]

[116]

[117]

[118]

[119]

[120]

signaling to induce NF-kappaB and activate T cells. Nat Immunol 2009;10:158–66. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev 2009;229:12–26. Rudd CE, Schneider H. Unifying concepts in CD28, ICOS and CTLA4 coreceptor signalling. Nat Rev Immunol 2003;3:544–56. Raab M, Cai YC, Bunnell SC, Heyeck SD, Berg LJ, Rudd CE. P56Lck and p59Fyn regulate CD28 binding to phosphatidylinositol 3-kinase, growth factor receptor-bound protein GRB-2, and T cell-specific protein-tyrosine kinase ITK: implications for T-cell costimulation. Proc Natl Acad Sci U S A 1995;92:8891–5. Bocko D, Kosmaczewska A, Ciszak L, Teodorowska R, Frydecka I. CD28 costimulatory molecule–expression, structure and function. Arch Immunol Ther Exp (Warsz) 2002;50:169–77. Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nat Rev Immunol 2003;3:317–30. Bustelo XR. Regulatory and signaling properties of the Vav family. Mol Cell Biol 2000;20:1461–77. Kane LP, Andres PG, Howland KC, Abbas AK, Weiss A. Akt provides the CD28 costimulatory signal for up-regulation of IL-2 and IFN-gamma but not TH2 cytokines. Nat Immunol 2001;2:37–44. Kane LP, Shapiro VS, Stokoe D, Weiss A. Induction of NF-kappaB by the Akt/ PKB kinase. Curr Biol 1999;9:601–4. Riley JL. PD-1 signaling in primary T cells. Immunol Rev 2009;229:114–25. Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 2005;25:9543–53. Cuddapah S, Barski A, Zhao K. Epigenomics of T cell activation, differentiation, and memory. Curr Opin Immunol 2010;22:341–7. Youngblood B, Noto A, Porichis F, Akondy RS, Ndhlovu ZM, Austin JW, et al. Cutting edge: Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. J Immunol 2013;191:540–4. Lee GR, Kim ST, Spilianakis CG, Fields PE, Flavell RA. T helper cell differentiation: regulation by cis elements and epigenetics. Immunity 2006;24:369–79. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97. Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, et al. MiR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 2007;129:147–61. Chen G, Cao P, Goeddel DV. TNF-induced recruitment and activation of the IKK complex require Cdc37 and Hsp90. Mol Cell 2002;9:401–10. Van Etten J, Schagat TL, Hrit J, Weidmann CA, Brumbaugh J, Coon JJ, et al. Human pumilio proteins recruit multiple deadenylases to efficiently repress messenger RNAs. J Biol Chem 2012;287:36370–83. Triboulet R, Gregory RI. Pumilio turns on microRNA function. Nat Cell Biol 2010;12:928–9. Wei S, Curiel T, Coukos G, Liu R, Zou W. Inhibitory B7 family members in human ovarian carcinoma. Adv Exp Med Biol 2008;622:261–71. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114: 1537–44. Kaufmann DE, Walker BD. PD-1 and CTLA-4 inhibitory cosignaling pathways in HIV infection and the potential for therapeutic intervention. J Immunol 2009;182:5891–7. Ward SG, Cantrell DA. Phosphoinositide 3-kinases in T lymphocyte activation. Curr Opin Immunol 2001;13:332–8. Jiang K, Zhong B, Gilvary DL, Corliss BC, Hong-Geller E, Wei S, et al. Pivotal role of phosphoinositide-3 kinase in regulation of cytotoxicity in natural killer cells. Nat Immunol 2000;1:419–25. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008;26:677–704. Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M, et al. PD-L1deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci U S A 2004;101: 10691–6. Sadlack B, Lohler J, Schorle H, Klebb G, Haber H, Sickel E, et al. Generalized autoimmune disease in interleukin-2-deficient mice is triggered by an uncontrolled activation and proliferation of CD4+ T cells. Eur J Immunol 1995;25:3053–9. Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ. Interleukin 2 signaling is required for CD4(+) regulatory T cell function. J Exp Med 2002;196:851–7. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 2012;24: 207–12. Doedens AL, Phan AT, Stradner MH, Fujimoto JK, Nguyen JV, Yang E, et al. Hypoxia-inducible factors enhance the effector responses of CD8(+) T cells to persistent antigen. Nat Immunol 2013;14:1173–82. Olurinde MO, Shen CH, Drake A, Bai A, Chen J. Persistence of tumorinfiltrating CD8 T cells is tumor-dependent but antigen-independent. Cell Mol Immunol 2011;8:415–23.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

M.Y. Balkhi et al. / Cytokine xxx (2014) xxx–xxx [121] Rossig C, Brenner MK. Genetic modification of T lymphocytes for adoptive immunotherapy. Mol Ther 2004;10:5–18. [122] Walker RE, Bechtel CM, Natarajan V, Baseler M, Hege KM, Metcalf JA, et al. Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection. Blood 2000;96:467–74. [123] Yun CO, Nolan KF, Beecham EJ, Reisfeld RA, Junghans RP. Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin T-cell receptors. Neoplasia 2000;2:449–59. [124] Beecham EJ, Ortiz-Pujols S, Junghans RP. Dynamics of tumor cell killing by human T lymphocytes armed with an anti-carcinoembryonic antigen chimeric immunoglobulin T-cell receptor. J Immunother 2000;23:332–43. [125] Nolan KF, Yun CO, Akamatsu Y, Murphy JC, Leung SO, Beecham EJ, et al. Bypassing immunization: optimized design of ‘‘designer T cells’’ against carcinoembryonic antigen (CEA)-expressing tumors, and lack of suppression by soluble CEA. Clin Cancer Res 1999;5:3928–41. [126] Boyle AP, Davis S, Shulha HP, Meltzer P, Margulies EH, Weng Z, et al. Highresolution mapping and characterization of open chromatin across the genome. Cell 2008;132:311–22. [127] Araki Y, Wang Z, Zang C, Wood 3rd WH, Schones D, Cui K, et al. Genome-wide analysis of histone methylation reveals chromatin state-based regulation of gene transcription and function of memory CD8+ T cells. Immunity 2009;30:912–25. [128] Hassig CA, Tong JK, Fleischer TC, Owa T, Grable PG, Ayer DE, et al. A role for histone deacetylase activity in HDAC1-mediated transcriptional repression. Proc Natl Acad Sci U S A 1998;95:3519–24.

9

[129] Murayama A, Sakura K, Nakama M, Yasuzawa-Tanaka K, Fujita E, Tateishi Y, et al. A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory. EMBO J 2006;25:1081–92. [130] Riquet FB, Tan L, Choy BK, Osaki M, Karsenty G, Osborne TF, et al. YY1 is a positive regulator of transcription of the Col1a1 gene. J Biol Chem 2001;276:38665–72. [131] Ugel S, Peranzoni E, Desantis G, Chioda M, Walter S, Weinschenk T, et al. Immune tolerance to tumor antigens occurs in a specialized environment of the spleen. Cell Rep 2012;2:628–39. [132] Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med 2007;13:828–35. [133] Hernandez J, Aung S, Redmond WL, Sherman LA. Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen. J Exp Med 2001;194:707–17. [134] Youngblood B, Wherry EJ, Ahmed R. Acquired transcriptional programming in functional and exhausted virus-specific CD8 T cells. Curr Opin HIV AIDS 2012;7:50–7. [135] Teague RM, Sather BD, Sacks JA, Huang MZ, Dossett ML, Morimoto J, et al. Interleukin-15 rescues tolerant CD8+ T cells for use in adoptive immunotherapy of established tumors. Nat Med 2006;12:335–41. [136] Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012;12:253–68.

Please cite this article in press as: Balkhi MY et al. T cell exhaustion and Interleukin 2 downregulation. Cytokine (2014), http://dx.doi.org/10.1016/ j.cyto.2014.11.024

T cell exhaustion and Interleukin 2 downregulation.

T cells reactive to tumor antigens and viral antigens lose their reactivity when exposed to the antigen-rich environment of a larger tumor bed or vira...
1MB Sizes 3 Downloads 9 Views