Next Article in Journal
NOTCH1 Intracellular Domain and the Tumor Microenvironment as Prognostic Markers in HNSCC
Next Article in Special Issue
Systematic Review of Available CAR-T Cell Trials around the World
Previous Article in Journal
External Validation of a Radiomics Model for the Prediction of Complete Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer
Previous Article in Special Issue
Strategies for Improving the Efficacy of CAR T Cells in Solid Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Knowns and Unknowns about CAR-T Cell Dysfunction

1
Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
2
Laboratory of Transplantation Immunology, National Research Centre for Hematology, 125167 Moscow, Russia
3
Institute of Hematology, Almazov National Medical Research Center, 197341 Saint Petersburg, Russia
4
Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(4), 1078; https://doi.org/10.3390/cancers14041078
Submission received: 6 December 2021 / Revised: 29 January 2022 / Accepted: 11 February 2022 / Published: 21 February 2022
(This article belongs to the Special Issue Current Advances in Chimeric Antigen Receptor Technology)

Abstract

:

Simple Summary

The primary issue of adoptive cell therapy is the poor in vivo persistence. In this context, it is necessary to clarify the fundamental mechanisms of T cell dysfunction. Here we review common dysfunctional states, including exhaustion and senescence, and discuss the challenges associated with phenotypical characterization of these T cell subsets. We overview the heterogeneity among exhausted T cells as well as mechanisms by which T cells get reinvigorated by checkpoint inhibitors. We emphasize that some cancers not responding to such treatment may activate distinct T cell dysfunction programs. Finally, we describe the dysfunction-promoting mechanisms specific for CAR-T cells and the ways to mitigate them.

Abstract

Immunotherapy using chimeric antigen receptor (CAR) T cells is a promising option for cancer treatment. However, T cells and CAR-T cells frequently become dysfunctional in cancer, where numerous evasion mechanisms impair antitumor immunity. Cancer frequently exploits intrinsic T cell dysfunction mechanisms that evolved for the purpose of defending against autoimmunity. T cell exhaustion is the most studied type of T cell dysfunction. It is characterized by impaired proliferation and cytokine secretion and is often misdefined solely by the expression of the inhibitory receptors. Another type of dysfunction is T cell senescence, which occurs when T cells permanently arrest their cell cycle and proliferation while retaining cytotoxic capability. The first section of this review provides a broad overview of T cell dysfunctional states, including exhaustion and senescence; the second section is focused on the impact of T cell dysfunction on the CAR-T therapeutic potential. Finally, we discuss the recent efforts to mitigate CAR-T cell exhaustion, with an emphasis on epigenetic and transcriptional modulation.

1. Introduction

Cellular immunity dysfunction is a hallmark of cancer—a result of the disease’s natural evolution toward counteracting the immune pressure. In contrast to dysfunctional T cells, functional T cells perform a range of activities upon stimulation with the cognate antigen. They can (1) expand; (2) secrete effector cytokines (e.g., IL-2, IFNγ, TNFα, perforins, and granzymes) and lyse target cells; (3) survive after removal of antigen stimulation; and (4) do all of the above upon secondary antigen challenge. In this review, a T cell is considered dysfunctional if at least one of these criteria is not met. The most studied dysfunctional state of T cells is exhaustion, which is characterized by the loss of all aforementioned properties of the functional T cells. Nonetheless, T and NK cells exhibit at least two additional well-defined dysfunctional states (senescence and anergy) [1]. Senescent T cells are phenotypically similar to terminal effector T cells in that they do not proliferate in response to antigen stimulation but retain the ability to secrete effector cytokines and kill target cells, in contrast to exhausted T cells (Tex).
T cell failure in cancer is mediated by multiple mechanisms, with the tumor microenvironment playing a central role in this process. Since the microenvironment varies significantly among different types of cancer, tumor-infiltrating lymphocytes will adopt tumor-specific dysfunctional states that, while similar to T cell exhaustion, possess distinct characteristics. These dysfunctional states are of great interest to the scientific community and require further classification. Additionally, these processes may affect not only naturally developed T cells but also adoptively transferred cell products expanded ex vivo. Although chimeric antigen receptor (CAR) T cells have revolutionized the treatment of hematological malignancies, further advancements are necessary to achieve long-lasting clinical outcomes. Indeed, many relapses of hematological tumors are associated with poor CAR-T cell persistence and are not due to target antigen loss. The low efficacy of CAR-T cells in solid tumors is also a result of intrinsic or tumor-associated T cell dysfunction and the subsequent loss of persistence. Moreover, T cells may be pre-enriched with dysfunctional populations during the manufacturing process. Additional variables affecting cell functionality include genetic modification, such as CAR transduction.
In this review, we examine mechanisms underlying T cell dysfunction beyond classical CD8+ exhaustion during chronic infection. We overview a variety of dysfunctional T cell states, discuss how they may develop in CAR-T cells, and look at the role of CAR-T cell-specific pathways in promoting this dysfunction. We elaborate on the recently discovered tonic CAR signaling and several unanswered questions in this field, such as the optimal composition of the CAR-T cell product and role of the chemotherapeutic pre-treatment in the “fitness” of CAR-T cells. Finally, we also mention the strategies for ameliorating CAR-T cell dysfunction via targeted “point” modification of the distinct inhibitory pathway (i.e., PD-1), as well as broad modulation of the CAR-T cell transcriptional and epigenetic state.

2. Dysfunctional States of T Cells

2.1. CD8+ T Cell Exhaustion

Initially, T cell exhaustion was regarded as a sequential process, with T cells sequentially transitioning first to effector state and ultimately toward dysfunction in response to persistent stimulation with their cognate antigen [2,3]. However, a novel model was proposed that describes exhaustion as an alternative pathway for chronically stimulated T cell differentiation (as opposed to the “conventional” effector/memory differentiation) [4,5,6,7]. According to the model, only T cells with differentiation plasticity (e.g., naïve or memory precursor T cells) can become exhausted (in contrast to terminally differentiated effector cells) [8,9,10].
T cell exhaustion was extensively investigated using a murine chronic lymphocytic choriomeningitis (LCMV) model. Apart from this chronic infection, exhaustion is thought to occur in various settings where antigen exposure persists: in mouse cancer models [4,11,12,13] as well as in human chronic infections (HIV, HCV, and HBV), cancer, and autoimmune diseases [14,15,16,17,18,19,20,21,22,23,24,25,26]. It is widely accepted that Tex exhibit upregulation of inhibitory receptors (IRs), decreased survival (due to lower Bcl-2 and higher Bim expression), poor cytotoxicity, and impaired secretion of cytokines such as IL-2, TNFα, and IFNγ [3,27,28,29,30,31]. Moreover, Tex adopt a distinct epigenetic landscape that is unique enough to consider them a separate T cell lineage [32,33]. There is evidence suggesting that exhaustion is already epigenetically imprinted in T cells on day 5 of chronic LCMV, distinguishing them from memory cells formed on day 5 of acute LCMV [8]. In contrast to memory precursor cells from acute LCMV, these cells expressed increased levels of PD-1 and TOX and expanded inefficiently upon secondary rechallenge. After removal of the chronic antigen stimulation (by transferring Tex into noninfected or acutely infected hosts), some aspects of T cell function could still be restored [11,34,35,36,37]. Despite this, the epigenetic landscape and impaired functionality of terminally Tex remained stable even after checkpoint blockade or antigen removal [6,18,38].
Tex cells are highly heterogeneous [4,5,31,39,40]. CD8+ Tex cells are broadly represented by TCF1+ Tex progenitors capable of proliferation/self-renewal and TCF1 terminally exhausted cells [39,41]. TCF1 is a transcription factor essential for the development of Tex progenitors and therapeutic response to checkpoint inhibition. More specifically, CD8+ Tex were recently found to linearly progress along the following trajectory: Ly108+ CD69+ (TCF1+ PD-1int) -> Ly108+ CD69 (TCF1+ PD-1int) -> Ly108 CD69 (TCF1 PD-1int) -> Ly108 CD69+ (TCF1 PD-1hi) [4]. Although described in the chronic LCMV model, these phenotypes also apply to Tex cells from mouse and human melanoma [4,42]. In another study, the authors found a correlation between the epigenetic landscape of Tex and their surface phenotype [37]. TCF1-expressing CD38low CD101low Tex cells possessed a plastic epigenetic state and were able to restore IFNγ and TNFα production, in contrast to CD38hi CD101hi Tex cells that were resistant to reprogramming.
A specific population of terminally differentiated CD8+ Tex superior in viral or tumor control was reported [43,44,45]. It resembles short-lived effector cells (SLECs) from acute infection, forms in the presence of IL-21, is distinct from dysfunctional PD-1hi terminally CD8+ Tex, has higher cytolytic activity and cytokine secretion, and is characterized by increased expression of CX3CR1, KLRG1, T-bet, and Zeb-2. To support this, the evidence suggests that exhaustion is caused by suboptimal priming of T cells and not persistent antigen per se. For the proper T cell priming, several conditions are required such as strong T cell receptor (TCR) signal, engagement of costimulatory molecules, cytokine-driven inflammation (presence of IL-2 and IL-12, or IFNα/b), and the lack of inhibitory signals [46,47,48,49,50]. If these conditions are met, T cells differentiate into a large pool of cytotoxic short-term effector cells and a small pool of memory cells. When multiple signals are suboptimal, activated T cell differentiation is skewed towards exhaustion. In agreement with this, when CD8+ T cells are primed with nonhematopoietic cells or DCs from chronically infected mice, they exhibit greater exhaustion than when primed with appropriate APCs [51,52,53,54].
The clinical relevance of the Tex heterogeneity is supported by the fact that checkpoint inhibition has a differential effect on various exhausted subpopulations. In humans and mice, the effect of at least PD-1/PD-L1 blockade is mediated by TCF1+ Tex progenitors capable of self-renewal and replenishment of the TCF1 Tex cell pool [4,6,39,55,56]. Indeed, following PD-1 blockade, CD8+ tumor-specific cells do not escape from exhaustion but are largely replaced by other tumor-specific CD8+ T cells with a distinct TCR repertoire [57]. For the development of a TCF1+ Tex progenitor subset and its continuous recruitment to the tumor site, tumor-draining lymph nodes play the major role [42,58]. In line with this, DCs in those lymph nodes, mediating priming of new T cells are crucial for success of the anti-PD-1 therapy [59,60,61]. Nonetheless, some studies indicate that T cell dysfunction in cancer may differ from the classical exhaustion landscape [11,14]. For example, in sharp contrast to the findings above, a subset of TCF1+ dysfunctional lung adenocarcinoma-infiltrating T cells was found to be irresponsive to checkpoint inhibition [14]. Representation of cancer-associated T cell dysfunction as a distinct entity is further supported by comparing its transcriptomic profile with classical exhaustion cases. Although significantly overlapped, the profile included 567 genes with altered expression that was specific for T-cells exhausted in the cancer environment [11,62]. Moreover, a recent study revealed that the dysfunction program might depend on TCR affinity with high-affinity clones adopting a classical exhaustion state, and low-affinity cells developing distinct dysfunction mechanism of “functional inertness” [63]. This observation further supports the broader landscape of cancer-associated dysfunction.

2.2. CD4+ T Cell Exhaustion

It is well established that CD4+ help is necessary for CD8+ T cells to function properly [10]. Similar to CD8+ T cells, CD4+ T cells are prone to exhaustion during chronic infection, with diminished effector functionality [36,62,64,65], impaired TNFα and IFNγ production, and increased PD-1 expression [62,64]. Chronic stimulation can result in a rapid loss of antigen-specific CD4+ T cells, which is relevant for human viral infections, such as HIV or HCV [66,67]. The acute self-resolving course of HCV infection was associated with a preserved virus-specific CD4+ response [66]. Similar to CD8+ T cells, CD4+ Tex form a T cell lineage distinct from the well-characterized CD4+ T cell phenotypes (Th1, Th2, and Th17) [64]. These cells may promote CD8+ T cell exhaustion by producing IL-10 [64,68], but they can also exhibit an upregulated IFN type I response [64,69] and increased production of IL-21 [64]. Interestingly, IL-21 is known to ameliorate CD8+ T cell exhaustion. Recently, Chiara et al. demonstrated that melanoma-associated dysfunction in CD4+ T cells, characterized by the expression of the known IRs (TIGIT, PD-1, and LAG-3), is IL-27 dependent [62]. This IL-27-driven program was dependent on Blimp-1 and c-MAF and was associated with the expression of several novel dysfunction drivers (PROCR and PDPN). Knocking -out of these genes resulted in improved tumor control. Although both CD4+ and CD8+ T cells share a common core transcriptional signature of T cell exhaustion, certain IRs (including CTLA4, CD200, and BTLA) and costimulatory molecules (OX40 and ICOS) are biased toward CD4+, demonstrating their differential regulation. Finally, similar to CD8+ T cell exhaustion, substantial heterogeneity is observed within CD4+ Tex cells [64].
These studies demonstrate that while CD4+ T cell exhaustion largely resembles that of CD8+ T cells, it also possesses distinct regulatory mechanisms that require further elucidation. Though largely ignored by the scientific community, CD4+ T cell exhaustion is equally essential and should be considered when developing novel immunotherapies.

2.3. How to Combat Exhaustion?

The most well-known and clinically approved approach is the checkpoint blockade (i.e., CTLA inhibition with ipilimumab or PD-L1 inhibition with nivolumab, pembrolizumab, etc.). Importantly, as previously stated, the checkpoint blockade does not reverse exhaustion but rather boosts proliferation of the less differentiated T cell populations, thereby generating an extensive pool of terminally exhausted T cells. While there is currently no way to completely reverse T cell exhaustion (i.e., dedifferentiate Tex into nonexhausted T cell lineages such as memory T cells), the research community has developed numerous strategies to mitigate it and thus improve T cell function. For instance, the engagement of costimulatory molecules is beneficial for T cell antitumor and antiviral activity. T cell functionality is enhanced by the activation of OX40 [70], 4-1BB [71,72], CD27 [73,74], and CD2 costimulatory receptors [15]. Moreover, there is a synergy between the checkpoint blockade and activation of costimulatory molecules that allowed various therapy combinations to achieve significant results. Examples include PD-1 blockade and 4–1BB stimulation in chronic LCMV [75]; PD-1 blockade and OX-40 stimulation in ovarian cancer [76]; CTLA-4 blockade and ICOS or 4–1BB stimulation in melanoma [44,77]; and PD-1 blockade and CD28 stimulation in prostate cancer [78].
Similarly, treatment with inflammatory cytokines, such as IL-2 +/- IL7 [53,79,80], IL-12 [81,82], and IL-21 [45,51,83,84], also improved T cell functionality. Interestingly, these cytokines not only improved T cell functionality but also reinvigorated Tex cells [45,53,81,85]. The efficacy of this strategy was validated in a clinical setting for tumor-infiltrated lymphocyte (TIL)-based therapy [86,87]. Moreover, the PD-1 blockade was synergized with IL-2 in chronic LCMV infection or tumor models [88] and with IL-12 in HBV [85].
On the other hand, several soluble factors, including IL-10 [68,89,90,91], IL-35 [92], TGF-β [93], glucocorticoids [13], PGE2 [94], and VEGF-A [25] were shown to promote exhaustion. Along those lines, the PD-1/PD-L1 blockade was synergized with IL-10 or PGE2 inhibition in chronic LCMV [89,90,94,95] and with TGF-β inhibition in melanoma [96].
Although multiple strategies were suggested for improving the functionality of Tex, one should keep in mind that exhaustion is epigenetically imprinted and requires complex combinatorial approaches to be truly reversed.

2.4. T cell Senescence

Cellular senescence is a stereotypical multicausal process that occurs in a variety of cell types and is characterized by cell cycle arrest. Replicative senescence is telomere-dependent and occurs after a critical number of cell divisions. Telomere-independent senescence, which can be induced by chemotherapy, primarily prevents tumorigenesis upon oncogene activation or DNA damage [97]. Senescent cells exhibit a specific senescence-associated secretory phenotype (SASP) that exerts a paracrine influence on other cells. T cell senescence and T cell exhaustion are distinct processes [2]. They do, however, share some phenotypic features, resulting in terminological confusion and misusage [98,99,100].
Senescent T cells tend to have a CD45RA+CD27CD28KLRG1+CD57+ phenotype, and they lose their proliferative capacity and ability to secrete IL-2, but express cytolytic molecules, IFNγ, and TNFα and are immediately cytotoxic ex vivo [101,102].
T cell senescence is generally associated with the activation of DDR (DNA damage response), which is triggered by telomere erosion, DNA damage caused by reactive oxygen species (ROS), glucose or growth factor deprivation, and cAMP pathway activation [98,103,104]. T cells, in contrast to other cell types, may express telomerase upon activation, conferring on them a certain degree of resistance to replicative senescence [105]. This is demonstrated by the fact that some T cell clones undergo up to 170 population doublings during in vitro culturing [105]. There is mounting evidence that T cell senescence is, in the majority of cases, an active process driven by the telomere-independent p38 pathway [106], and that senescent T cells do not have critically short telomeres [107,108]. The p38-mediated signaling inhibits autophagy, which is necessary for the recycling of damaged mitochondria [108]. Such mitochondria produce excessive ROS leading to DDR, which itself activates p38 in a positive feedback loop [109]. Additionally, p38 inhibits telomerase activity and promotes p16 and p53 activation, which irreversibly arrests the T cell cycle [110,111]. In senescent or glucose-deprived CD4+ T cells, sestrin proteins (SESN1/2/3) inhibit mTOR and activate AMPK that upregulates p38, Erk, and JNK via the sMAC complex, and thus contributes to senescence [112,113]. In senescent CD8+ T cells, sestrin 2 promotes the expression of the NKG2D/DAPC2 complex via JNK activation, leading to TCR-independent cytotoxicity [114].
Interestingly, IFNα treatment (which drives T cells towards terminal effector differentiation) results in p38 activation, and this potentially accounts for the poor proliferative capacity of the terminal effector population, as well as phenotypic and functional similarities to senescent T cells [110]. Senescent T cells possess their distinct p38-dependent SASP, which differs from the SASP of other cell types and includes TNFα, IL-18, IL-29, CCL5, CCL16, and ADAM28 [115]. It is still not clearly known how this SASP affects other immune cells.
T cells become telomere-independently senescent within the immunosuppressive tumor microenvironment as a result of inhibition by tumor-derived Tregs, γδ T cells, and tumor cells themselves [104,116]. Repeated activation during chronic infection or autoimmune diseases is also associated with T cell senescence [100]. Moreover, tumor-induced senescent T cells have been shown to suppress the activity of other T cells within the tumor microenvironment [104,117]. Although there is increasing evidence that tumors induce T cell senescence as an evasion mechanism, additional studies are required to properly investigate this type of T cell dysfunction and identify options for therapeutic modulation.
In summary, senescent T cells exhibit increased p38 activation and ROS accumulation, impaired mitochondria activity, downregulated mTOR signaling, and shorter telomeres [108,113,118,119,120]. Despite undergoing cell cycle arrest, these cells retain cytotoxicity and resemble terminal effector T cells. The T cell dysfunction landscape is schematically depicted in Figure 1.

2.5. How to Identify Exhausted and Senescent T Cells

Classical exhaustion models are designed in a way that allows exhausted T cells to be readily identifiable (e.g., with MHC-tetramer staining). Nevertheless, there is no established strategy for identification and isolation of exhausted T cells while working with human clinical specimens. Ideally, in order to diagnose T cell exhaustion, one should assess T cell functionality, IR expression, as well as the epigenetic and transcriptional state of a given T cell population. The assessment of T cell functionality (proliferation, cytokine secretion, and cytotoxicity) is of great importance because the surface phenotype (expression of IRs) can be misleading. Indeed, although IR expression often correlates with exhaustion, transient expression of almost all IRs is a hallmark of recently activated effector cells that have no relation to exhaustion [69]. PD-1, for instance, was shown to mark T cells with potent antitumor activity rather than exhausted cells [121]. In light of this uncertainty, large multicolor panels for flow or mass cytometry may be beneficial. One such approach is not only based on the surface and characteristic intracellular Tex markers, such as IRs, TOX, CXCR5, and Eomes, but also includes naïve/memory markers CCR7, CD73, CD127, and TCF-1, which are downregulated in many Tex [122]. However, this strategy is likely to identify only terminally exhausted T cells and not precursor subpopulations. To summarize, despite substantial progress in the development of cytometry exhaustion panels, functional assays remain the most robust and validated method to confirm exhaustion.
Some of the existing approaches for identification of senescent T cells (e.g., based on CD57+CD27low and CD28 SA-β-Gal+) are yet to be validated [98,100,123]. Particular caution should be exercised when using the CD45RA+ marker for terminally differentiated effector cells (TEMRA), since the existence of CD45RA+ nonsenescent proliferating memory cells has also been reported [124]. To avoid improper gating, a broad panel of surface markers should be considered that rely on the CD45RA+ CD27 CD28 KLRG1+ CD57+ phenotype of the senescent cells.

3. Dysfunctional States of CAR-T Cells

T cells that have been transduced with CAR exhibit similar mechanisms of dysfunction as unmodified T cells. Indeed, CD8+ CAR-T cells and TILs isolated from the same tumor-bearing mouse have similar transcriptional and epigenetic profiles [125]. Targeting pathways known to contribute to T cell dysfunction may improve CAR-T cell functionality. This impacts PD-1 [126,127,128,129,130] (including three case reports when CAR-T cells were reinvigorated with anti-PD-L1 treatment [131,132,133]), TIM-3 [129,134,135], CTLA-4 [134], TGF receptor [136,137], and adenosine receptor A2 [138,139]. Other approaches target intracellular signaling mediators [129,140,141,142,143,144,145,146], increase telomerase activity [147], or reverse metabolic inhibition of T cell activity [129,148,149,150]. These are described in more detail in Table 1.
Although CAR-T cell-specific exhaustion mechanisms have been recently described, e.g., prolonged tonic CAR signaling, many aspects of such exhaustion remain largely unknown and require further clarification. Does it occur in vivo after adoptive transfer or is it turned on ex vivo during cell expansion? Can it be attributed to the higher affinity of scFv compared to TCR? Is it sustained with the persistence of tumor-associated antigens expressed by healthy tissues (for instance, CD19 on survived tumor B-cells or constantly developing B-cell progenitors)? In addition, the apheresis product composition also plays a critical role, as it may be enriched in dysfunctional/senescent cells or skewed toward a specific phenotype (e.g., exclusively CD8+ T cells). This section focuses on CAR-T-specific dysfunction and modulation of their epigenetic and transcriptional profiles, whereas Table 1 summarizes the targeting of common T cell pathways in CAR-T cells. The factors affecting CAR-T cell function in vivo are depicted in Figure 2.

3.1. CAR Signaling as a Driver of Dysfunction and the Road to Its Prevention

A variety of CAR domains and their combinations may differently impact the CAR-T cells dysfunction. Long et al. revealed that GD2-28z CAR-T cells were more prone to exhaustion than GD2-BBz CAR-T cells. This tonic GD2-28z signaling resulted from scFv-mediated CAR clustering that occurred in the absence of an antigen and thus was specific for GD2 CAR-T cells, as opposed to CD19-28z CAR [152]. However, 4-1BB costimulation is not always beneficial. In comparison to 28z CAR, the overexpression of both GD2-BBz or CD19-BBz led to a lower expansion in vitro, higher target cell viability, CAR-T apoptosis, and decreased survival of tumor-bearing mice [153]. The authors found that tonic signaling of overexpressed BBz CAR induces a positive feedback loop, further increasing CAR expression and FasL-dependent apoptosis. This effect was observed both upon stimulation with the CD19+ NALM6 leukemia cell line in vivo and without antigen stimulation. Downregulation of BBz CAR expression rescued CAR-T cell function. Notably, an increase in the number of CD4+ cells within the total CAR+ T cell population upon upregulation of tonic 4-1BB signaling suggests that CD4+ CAR-T cells may demonstrate superior resistance to tonic signaling.
Recognizing the crucial role of tonic CAR signaling in CAR-T cell exhaustion prompted an effort to resolve it. Weber et al. recently developed a CAR that requires a stabilizing compound for the assembly and signaling. They showed that transient “resting” of CAR-T cells improved functionality and may help mitigate exhaustion both functionally and epigenetically, even in “aged” CAR-T cells with constant tonic signaling lasting up to 45 days [154]. They demonstrated that dasatinib, an FDA-approved multikinase inhibitor [155], has a similar effect on CAR-T cells underlining its potential clinical application. However, the authors emphasize that their findings contradict those of mouse models in which a late (>14 days) “rescue” of T cells from their cognate antigen did not result in their reinvigoration [5,11].
An alteration of costimulation domains may also contribute to CAR-T cell persistence. Apart from 4-1BB and CD28, another promising costimulator is ICOS. Although CD28 and ICOS are structurally similar and share downstream signaling partners, they have distinct extracellular ligands [156]. In contrast to CD28-based CARs, the ICOS-based ones have been shown to result in dramatically higher persistence of CAR-T cells [157]. Guedan et al. established that this effect depends on the ICOS YMFM-motif [158]. By replacing a single amino acid in CD28 (YMNM->YMFM), they gained persistence levels comparable to ICOS-costimulated CAR-T cells. Notably, the differences were observed 14 days postinfusion, highlighting the long-term effect of YMFM-CD28 or ICOS costimulation on CAR-T cell functionality. ICOS and YMFM-CD28 costimulation drove memory-enriched gene signature and induced Th17-like differentiation of CAR-T cells, which was beneficial in clinical settings (CD19 CAR-T cells) and was associated with durable responses and overall therapeutic efficacy [159,160].
A CD2 cell adhesion molecule was previously shown to be an essential costimulator of T cells, preventing exhaustion in an autoimmunity setting, where CD2-dependent costimulation was significantly correlated with a poor prognosis in patients with autoimmune conditions [15]. Accordingly, Majzner et al. observed that the downregulation of CD58 (CD2 ligand) on tumor cells is responsible for significant impairment of CAR-T cell function and leads to markedly reduced progression-free survival in patients with B-cell malignancies [161]. Manipulation of the CD58-CD2 axis may be an attractive option for the generation of improved CAR-T cells, as illustrated by the inclusion of CD2 signaling into the CAR costimulatory module [162].
In summary, T cells with novel CAR designs require extensive investigation to determine their tonic signaling capacity and ability to result in long-lasting cytotoxicity. Multiple costimulation modules (e.g., CD2, ICOS, and YMFM-CD28) may benefit CAR-T cell functionality and help counteract the acquired mechanism of tumor resistance and tumor-dependent CAR-T cell suppression.

3.2. Adjusting Regulatory Networks to Counteract T Cell Dysfunction

The manipulation of a single gene with a defined connection to T cell dysfunction seems an appealing strategy for enhancing the efficacy of CAR-T cells (Table 1). However, inhibitory receptors, for example PD-1, are merely markers of T cell exhaustion and not the cause; thus regulating them is unlikely to reverse this dysfunctional state. Indeed, upon persisting antigen stimulation, PD-1 knockout T cells expanded and functioned well initially; however, later they were severely exhausted, in a much more pronounced manner than wild-type T cells [163]. Moreover, although repeatedly stimulated CD8+ T cells were PD-1+, they could not be rescued by the blockade of PD-1. Such treatment did not significantly recover antigen-specific T cells compared to the acute infection model [164]. These findings demonstrate that disrupting PD-1 signaling is not always effective and may even be harmful.
Instead of editing the IR gene itself, modifying its enhancer might be more advantageous. Gennert et al. disrupted the exhaustion-associated enhancer of the PD-1 gene in GD2-28z CAR-T cells and discovered a marked reduction in PD-1high CAR-T cells following ex vivo expansion (10 days after CAR transduction) [165]. Preservation of the PD-1 gene enables its expression in nonexhaustion conditions (i.e., following acute stimulation).
Manipulation of transcriptional networks involved in T cell exhaustion appears to be a more viable strategy than downregulation of specific IRs. For example, in Tex, there is a disbalance between NFAT and AP-1, leading to predominant activation of NFAT-driven exhaustion-associated genes, and not the genes activated by NFAT:AP-1 complexes [166]. Indeed, CAR-T cells overexpressing c-Jun (subunit of AP-1) are more resistant to exhaustion [167]. Overexpression of c-Jun leads to multiple outcomes: increased production of IL-2 and IFNγ in CD19-28z CAR-T cells; IL-2-dependent increase in proliferation of both CD19-28z and CD19-BBz CAR-T cells; reduction in exhaustion markers; and increased efficacy of CAR-T cells in leukemia and osteosarcoma mouse models. Interestingly the authors also observed that c-Jun disrupted AP-1-IRF complexes, allowing the rescue of exhausted CAR-T cells.
Basic leucine zipper transcriptional factor ATF-like (BATF) is another molecule shown to ameliorate T cell exhaustion [168]. Recent research indicated that overexpression of BATF in CAR-T cells also counteracts exhaustion, and this effect depends on BATF interaction with Interferon Regulatory Factor 4 (IRF4) [169]. CD19-28z CD8+ CAR-T cells overexpressing transcription factor BATF exhibited increased cytotoxicity, survival, and production of IFNγ, as well as decreased TOX and IR expression. Such cells displayed superior antitumor activity and improved persistence in colon cancer and melanoma mouse models after primary tumor clearance, enabling them to differentiate into memory T cells and protect against a secondary tumor challenge. Interestingly, unlike Lynn et al. [167], this group observed only a limited survival benefit of c-Jun overexpressing CAR-T cells in their melanoma model.
CD8+ Tex cells express a high level of the exhaustion-driving transcription factor NR4A and are enriched in NR4A binding DNA motifs [7,37]. The exhaustion-promoting role of NR4A has also been demonstrated in the CAR-T setting [125]. The authors revealed that CD19-28z CD8+ CAR-T cells with NR4A triple KO were capable of secreting IFNγ and TNFα upon restimulation and significantly prolonged the cells’ survival in melanoma and colon cancer mouse models expressing CD19. Unlike control cells, NR4A triple KO T cells exhibited the enrichment of NF-kB and AP-1 binding motifs, which may account for improved CAR-T cell functionality. This group also observed a similar improvement in the functionality of CD19-28z CD8+ CAR-T cells deficient in both TOX and TOX2 [170]. Indeed, TOX defines the epigenetic landscape of Tex [171,172,173]. The authors proposed a positive feedback loop between NR4A and TOX, potentially explaining the enrichment of NF-kB and AP-1 binding motifs in both NR4A triple KO and double TOX-deficient CAR-T cells. Unlike single KO of TOX2, a single KO of TOX also improved the tumor control.
Tex activate complex genetic programs orchestrated by a variety of mutually intertwined transcription factors in a specific epigenetic context. Deciphering these programs will enable a combinatorial targeting of transcription factors and, finally, a modifying of epigenetic traits responsible for T cell exhaustion.

3.3. The Role of CD4+ CAR-T Cells in Counteracting Exhaustion and Overall Therapeutic Efficacy

CD8+ CAR-T cells alone were shown to be efficient in treating B-cell malignancies [174,175]. Nevertheless, distinct subsets of CD4+ T cells bear helper functions essential for antiviral immunity (Th1 for CD8+ T cells and Tfh for B-cell immunity) or autoimmunity (Th17 cells) [15]. Among various predictors of clinical response, the Th17 cytokine secretion profile was identified as important for the clinical efficacy of CAR-T cells [176]. Moreover, in the context of Tex, the loss of CD4+ help results in severe exhaustion [2], whereas the transfer of CD4+ T cells diminishes exhaustion [10]. Indeed, the design and persistence of CD4+ CAR-T cells were critical for the long-lasting function of their CD8+ counterparts. Guedan et al. demonstrated that ICOSz CD4+ CAR-T cells significantly enhanced the persistence of CD8+ CAR-T cells in mice when compared to 28z or BBz CD4+ CAR-T cells [157].
Further supporting their importance, CD4+ CAR-T cells demonstrated similar killing activity to CD8+ in vitro and in vivo, in addition to their helper functions [177,178,179,180]. Although CD4+ CAR-T cells exhibited lower initial granzyme B secretion and tumor killing capabilities, they appear to be less susceptible to exhaustion and apoptosis, in comparison to CD8+ subpopulation [177,180,181], especially upon dual stimulation through CAR and cognate TCR [180]. These observations provide a rationale for using CD4+ T cells for CAR-T cell product manufacturing. Juno Therapeutics pioneered this approach in the clinic with their CAR-T cell therapy consisting of a 1:1 ratio of CD4+:CD8+ CAR-T cells. One of them (Breyanzi/lisocabtagene maraleucel) was approved by the FDA in March 2021. Although the direct comparisons of different CAR-T cell products are not available, one can speculate that Juno Therapeutic concept demonstrates equivalent or superior antitumor activity with a lower rate of serious adverse events [182,183,184].

3.4. Senescence in CAR-T Cells

Senescent T cells may be enriched in the apheresis product obtained from elderly patients or those pretreated with chemotherapy/total body irradiation. Indeed, Guha et al. recently addressed the differential proliferation ability of CAR-T cells derived from young versus elderly donors. They observed that elderly donors showed (i) decreased viral transduction efficiency, (ii) lower levels of proliferation markers, such as p-Akt, p-Erk, and p-STAT5 in CAR-transduced or untransduced IL-2/IL-15 stimulated T cells, and (iii) lower number of either CD4+ and CD8+ effector and effector memory cells [185]. The authors observed that the majority of CAR-T cells derived from elderly donors are represented by the CD45RA+CD45RO+ phenotype that is presumably in the process of transition from naïve to memory cells. This effect is likely explained by the increased proportion of senescent and highly differentiated effector T cells in elderly donors, since their naïve T cells remained functional and responded appropriately to the stimulation [186]. Although such senescent cells possess limited proliferative potential and contribute little to the final therapeutic cell product, they still may affect the quality of other T cells during the manufacturing phase via their SASP and contact-dependent interactions [187].
Therapy-induced senescence of CAR-T cells can occur when they are manufactured from the apheresis product obtained after chemotherapeutic treatment [188,189]. Accordingly, pretreatment with cyclophosphamide/doxorubicin-containing regimens appears to be associated with poorly performing CAR-T cells [190,191], which may indicate cellular senescence. Cytotoxic drugs have an impact on T cell proliferation, blasting, and survival. Cytarabine appeared to be the most toxic chemotherapeutic, whereas cyclophosphamide unexpectedly affected not memory, but naïve T cells that are considered quiescent [191]. In line with that, T cells of postchemotherapy patients are represented mainly by effector and memory populations in adults [192] and to a lesser extent in children [193]..
In summary, we may have observed a substantial increase in highly differentiated and senescent T cells in chemotherapy-treated or elderly patients, resulting in lower ex vivo CAR-T cell transduction and expansion. Yet, it remains unclear whether the naïve T cells depletion is due to the cytotoxic effect suspected by Das et al. [191] or caused by their acquisition of senescent phenotype. Nevertheless, we now see the importance of proper timing for effective adoptive immunotherapy, i.e., we should harvest immune cells earlier in the course of patient treatment before they are significantly affected by chemotherapy.

3.5. Clinical Correlations of CAR-T Cell Activity/Dysfunction

Biomarkers predicting the efficacy of CAR-T cell therapy are a hot topic of research [194]. Some prediction strategies take into account tumor burden and patient premorbidity (LDH level or estimation of measurable disease), while others reflect CAR-T cell expansion levels (IL-7 and peak CAR transcript). Many researchers stress the importance of less differentiated T/CAR-T cells with higher proliferative capacity (memory T cells [195], including CD27+CD45ROCD8+ phenotype [159,196]) or IL17A-producing polyfunctional CD4+ T cells [176]. Finally, recent clinical trials indicate that a 1:1 CD4+:CD8+ ratio of CAR-T cells is clinically beneficial [182,196,197]. Unfortunately, most biomarkers associated with therapy efficacy, such as IL-15, low LDH, or peak CAR-T expansion, also correlate with significant and even fatal toxicities [194,198].

4. Concluding Remarks

Nowadays, immunotherapy and adoptive cell transfer have become the standard of care for patients with certain B-cell hematological malignancies and are included in the international treatment guidelines. Nonetheless, the long-term clinical benefits can be observed in half of the patients. Improving the treatment outcomes is the primary goal of clinical immunology worldwide. In this context, the underlying causes and mechanisms of T cell dysfunction require further investigation and clarification. Certain similarities are evident between T cell states observed in a variety of chronic disease settings. Indeed, although not indisputable, PD-1/PD-L1 inhibition is now acknowledged as a reason for TCF1+ Tex progenitor proliferation and differentiation into TCF1 terminally exhausted T cells, rather than as a cause of terminal exhaustion reversal.
At the same time, despite their functional and transcriptional similarities, tumor-associated dysfunctional T cells may be distinct from their chronic infection counterparts or even from T cells detected in other cancer types. There is a wide range of closely related dysfunctional T cell subtypes associated with autoimmune disorders, infectious diseases, and various types of cancer, each of which requires a unique therapeutic approach.
Although antigen persistence and abundance have long been considered a critical factor for the generation of Tex, evidence is accumulating that optimal priming with optimal cytokine and costimulatory milieu is essential. In agreement with this, Tex were shown to revive, proliferate, and at least partially restore their functionality during cytokine-induced expansion.
CAR-T cells represent a unique phenomenon in the field of synthetic biology. They are regulated by the same exhaustion pathways as conventional T cells, and their modification may significantly boost the efficacy. We believe that CAR signaling itself is an essential exhaustion driver and requires additional in-depth investigations. The “building blocks” of a CAR molecule, including scFv and costimulatory domains, must be carefully examined in terms of compatibility and differential effect on distinct T cell subsets. For instance, ICOS costimulation substantially enhances functions and persistence of CD4+ CAR-T cells, the importance of which was unequivocally demonstrated both in animal models and clinically. Finally, modulation of epigenetic and transcriptional regulators in complex with controllable CAR signaling may reduce exhaustion by providing resistance to epigenetic exhaustion programs, and through a transient interruption of antigen stimulation or tonic CAR signaling. Despite the significant scientific progress in this field, many unknowns, such as modulating the epigenetic landscape of CAR-T cells or their senescence, require further research.
Overall, we believe that future strategies for counteracting T cell dysfunction may help to reduce the relapse rates and complement the range of currently available immunotherapies.

Author Contributions

A.T. and Y.K. conceived the idea and wrote the manuscript; E.Z. prepared the figures; I.G. prepared the tables; A.P., A.V., A.R. (Aygul Rakhmatullina) and R.M. contributed to the part on T cell dysfunction; A.R. (Albert Rizvanov) and E.B. coordinated the process and contributed to the part on dysfunctional states in CAR-T cells. All authors have read and agreed to the published version of the manuscript.

Funding

Work was funded by RSF grant #19-74-20026. This paper has been supported by the Kazan Federal University Strategic Academic Leadership Program (PRIORITY-2030).

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

A2aRAdenosine Receptor A2
ADAR1 Adenosine Deaminase RNA Specific
AKTiAKT inhibitor VIII
ASSArgininosuccinate synthase
BATFBasic leucine zipper transcriptional factor ATF-like
CAIXCarbonic anhydrase IX
CARChimeric Antigen Receptor
CAR-T cellT cell with Chimeric Antigen Receptor
COXCyclooxygenase
DDRDNA Damage Response
HBVHepatitis B Virus
HCVHepatitis C Virus
HIVHuman Immunodeficiency Virus
IDO-1Indoleamine 2,3-dioxygenase
IFNInterferon
iILInducible interleukin
ILInterleukin
IRInhibitory Receptors
IRF4Interferon Regulatory Factor 4
KOKnockout
LCMVMurine Lymphocytic Choriomeningitis
LDHLactate Dehydrogenase
MHCMain Histocompatibility Complex
OTCOrnithine transcarbamylase
PI3KδPhosphatidylinositol-3-kinase p110δ
PKAProtein Kinase A
PP2AProtein Phosphatase 2A
PTPN-2Protein tyrosine phosphatase non-receptor type 2
ROSReactive Oxygen Species
SASPSenescence-Associated Secretory Phenotype
shRNASmall hairpin RNA
SLECShort-Lived Effector Cells
TCRT Cell Receptor
TexExhausted T Cell
TIGITT cell immunoreceptor with Ig and ITIM domain
TILTumor Infiltrating Lymphocyte

References

  1. Judge, S.J.; Murphy, W.J.; Canter, R.J. Characterizing the Dysfunctional NK Cell: Assessing the Clinical Relevance of Exhaustion, Anergy, and Senescence. Front. Cell. Infect. Microbiol. 2020, 10, 49. [Google Scholar] [CrossRef] [Green Version]
  2. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  3. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  4. Beltra, J.-C.; Manne, S.; Abdel-Hakeem, M.S.; Kurachi, M.; Giles, J.R.; Chen, Z.; Casella, V.; Ngiow, S.F.; Khan, O.; Huang, Y.J.; et al. Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. Immunity 2020, 52, 825–841.e8. [Google Scholar] [CrossRef] [PubMed]
  5. Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef] [PubMed]
  6. Pauken, K.E.; Sammons, M.A.; Odorizzi, P.M.; Manne, S.; Godec, J.; Khan, O.; Drake, A.M.; Chen, Z.; Sen, D.R.; Kurachi, M.; et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016, 354, 1160–1165. [Google Scholar] [CrossRef] [Green Version]
  7. Sen, D.R.; Kaminski, J.; Barnitz, R.A.; Kurachi, M.; Gerdemann, U.; Yates, K.B.; Tsao, H.-W.; Godec, J.; LaFleur, M.W.; Brown, F.D.; et al. The epigenetic landscape of T cell exhaustion. Science 2016, 354, 1165–1169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Utzschneider, D.T.; Gabriel, S.S.; Chisanga, D.; Gloury, R.; Gubser, P.M.; Vasanthakumar, A.; Shi, W.; Kallies, A. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol. 2020, 21, 1256–1266. [Google Scholar] [CrossRef]
  9. Angelosanto, J.M.; Blackburn, S.D.; Crawford, A.; Wherry, E.J. Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 2012, 86, 8161–8170. [Google Scholar] [CrossRef] [Green Version]
  10. West, E.E.; Youngblood, B.; Tan, W.G.; Jin, H.T.; Araki, K.; Alexe, G.; Konieczny, B.T.; Calpe, S.; Freeman, G.J.; Terhorst, C.; et al. Tight Regulation of Memory CD8+ T Cells Limits Their Effectiveness during Sustained High Viral Load. Immunity 2011, 35, 285–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Schietinger, A.; Philip, M.; Krisnawan, V.E.; Chiu, E.Y.; Delrow, J.J.; Basom, R.S.; Lauer, P.; Brockstedt, D.G.; Knoblaugh, S.E.; Hämmerling, G.J.; et al. Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. Immunity 2016, 45, 389–401. [Google Scholar] [CrossRef] [Green Version]
  12. Mumprecht, S.; Schürch, C.; Schwaller, J.; Solenthaler, M.; Ochsenbein, A.F. Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression. Blood 2009, 114, 1528–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Acharya, N.; Madi, A.; Zhang, H.; Klapholz, M.; Escobar, G.; Dulberg, S.; Christian, E.; Ferreira, M.; Dixon, K.O.; Fell, G.; et al. Endogenous Glucocorticoid Signaling Regulates CD8+ T Cell Differentiation and Development of Dysfunction in the Tumor Microenvironment. Immunity 2020, 53, 658–671.e6. [Google Scholar] [CrossRef] [PubMed]
  14. Horton, B.L.; Morgan, D.M.; Momin, N.; Zagorulya, M.; Torres-Mejia, E.; Bhandarkar, V.; Wittrup, K.D.; Love, J.C.; Spranger, S. Lack of CD8+ T cell effector differentiation during priming mediates checkpoint blockade resistance in non-small cell lung cancer. Sci. Immunol. 2021, 6, eabi8800. [Google Scholar] [CrossRef]
  15. McKinney, E.F.; Lee, J.C.; Jayne, D.R.W.; Lyons, P.A.; Smith, K.G.C. T cell exhaustion, costimulation and clinical outcome in autoimmunity and infection. Nature 2015, 523, 612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ye, B.; Liu, X.; Li, X.; Kong, H.; Tian, L.; Chen, Y. T-cell exhaustion in chronic hepatitis B infection: Current knowledge and clinical significance. Cell Death Dis. 2015, 6, e1694. [Google Scholar] [CrossRef] [Green Version]
  17. Gruener, N.H.; Lechner, F.; Jung, M.C.; Diepolder, H.; Gerlach, T.; Lauer, G.; Walker, B.; Sullivan, J.; Phillips, R.; Pape, G.R.; et al. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J. Virol. 2001, 75, 5550–5558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Day, C.L.; Kaufmann, D.E.; Kiepiela, P.; Brown, J.A.; Moodley, E.S.; Reddy, S.; Mackey, E.W.; Miller, J.D.; Leslie, A.J.; DePierres, C.; et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006, 443, 350–354. [Google Scholar] [CrossRef] [PubMed]
  19. Kostense, S.; Ogg, G.S.; Manting, E.H.; Gillespie, G.; Joling, J.; Vandenberghe, K.; Veenhof, E.Z.; van Baarle, D.; Jurriaans, S.; Klein, M.R.; et al. High viral burden in the presence of major HIV-specific CD8+ T cell expansions: Evidence for impaired CTL effector function. Eur. J. Immunol. 2001, 31, 677–686. [Google Scholar] [CrossRef]
  20. Shankar, P.; Russo, M.; Harnisch, B.; Patterson, M.; Skolnik, P.; Lieberman, J. Impaired function of circulating HIV-specific CD8+ T cells in chronic human immunodeficiency virus infection. Blood 2000, 96, 3094–3101. [Google Scholar] [CrossRef]
  21. Goepfert, P.A.; Bansal, A.; Edwards, B.H.; Ritter, G.D.J.; Tellez, I.; McPherson, S.A.; Sabbaj, S.; Mulligan, M.J. 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, 10249–10255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kim, C.G.; Jang, M.; Kim, Y.; Leem, G.; Kim, K.H.; Lee, H.; Kim, T.-S.; Choi, S.J.; Kim, H.-D.; Han, J.W.; et al. VEGF-A drives TOX-dependent T cell exhaustion in anti-PD-1-resistant microsatellite stable colorectal cancers. Sci. Immunol. 2019, 4, eaay0555. [Google Scholar] [CrossRef] [PubMed]
  23. Huang, A.C.; Postow, M.A.; Orlowski, R.J.; Mick, R.; Bengsch, B.; Manne, S.; Xu, W.; Harmon, S.; Giles, J.R.; Wenz, B.; et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017, 545, 60–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Fourcade, J.; Sun, Z.; Benallaoua, M.; Guillaume, P.; Luescher, I.F.; Sander, C.; Kirkwood, J.M.; Kuchroo, V.; Zarour, H.M. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010, 207, 2175–2186. [Google Scholar] [CrossRef]
  25. Zhang, Y.; Huang, S.; Gong, D.; Qin, Y.; Shen, Q. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer. Cell. Mol. Immunol. 2010, 7, 389–395. [Google Scholar] [CrossRef]
  26. Bengsch, B.; Ohtani, T.; Khan, O.; Setty, M.; Manne, S.; O’Brien, S.; Gherardini, P.F.; Herati, R.S.; Huang, A.C.; Chang, K.-M.; et al. Epigenomic-Guided Mass Cytometry Profiling Reveals Disease-Specific Features of Exhausted CD8 T Cells. Immunity 2018, 48, 1029–1045.e5. [Google Scholar] [CrossRef] [Green Version]
  27. Baitsch, L.; Baumgaertner, P.; Devêvre, E.; Raghav, S.K.; Legat, A.; Barba, L.; Wieckowski, S.; Bouzourene, H.; Deplancke, B.; Romero, P.; et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Investig. 2011, 121, 2350–2360. [Google Scholar] [CrossRef] [Green Version]
  28. Barber, D.L.; Wherry, E.J.; Masopust, D.; Zhu, B.; Allison, J.P.; Sharpe, A.H.; Freeman, G.J.; Ahmed, R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2005, 439, 682–687. [Google Scholar] [CrossRef]
  29. Petrovas, C.; Chaon, B.; Ambrozak, D.R.; Price, D.A.; Melenhorst, J.J.; Hill, B.J.; Geldmacher, C.; Casazza, J.P.; Chattopadhyay, P.K.; Roederer, M.; et al. Differential association of programmed death-1 and CD57 with ex vivo survival of CD8+ T cells in HIV infection. J. Immunol. 2009, 183, 1120–1132. [Google Scholar] [CrossRef] [Green Version]
  30. Penna, A.; Pilli, M.; Zerbini, A.; Orlandini, A.; Mezzadri, S.; Sacchelli, L.; Missale, G.; Ferrari, C. Dysfunction and functional restoration of HCV-specific CD8 responses in chronic hepatitis C virus infection. Hepatology 2007, 45, 588–601. [Google Scholar] [CrossRef]
  31. He, R.; Hou, S.; Liu, C.; Zhang, A.; Bai, Q.; Han, M.; Yang, Y.; Wei, G.; Shen, T.; Yang, X.; et al. Follicular CXCR5- expressing CD8+ T cells curtail chronic viral infection. Nature 2016, 537, 412–428. [Google Scholar] [CrossRef] [PubMed]
  32. Yates, K.B.; Tonnerre, P.; Martin, G.E.; Gerdemann, U.; Al Abosy, R.; Comstock, D.E.; Weiss, S.A.; Wolski, D.; Tully, D.C.; Chung, R.T.; et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol. 2021, 22, 1020–1029. [Google Scholar] [CrossRef] [PubMed]
  33. Abdel-Hakeem, M.S.; Manne, S.; Beltra, J.C.; Stelekati, E.; Chen, Z.; Nzingha, K.; Ali, M.A.; Johnson, J.L.; Giles, J.R.; Mathew, D.; et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat. Immunol. 2021, 22, 1008–1019. [Google Scholar] [CrossRef]
  34. Chevrier, S.; Levine, J.H.; Zanotelli, V.R.T.; Silina, K.; Schulz, D.; Bacac, M.; Ries, C.H.; Ailles, L.; Jewett, M.A.S.; Moch, H.; et al. An Immune Atlas of Clear Cell Renal Cell Carcinoma. Cell 2017, 169, 736–749.e18. [Google Scholar] [CrossRef] [Green Version]
  35. Correa-Rocha, R.; Lopez-Abente, J.; Gutierrez, C.; Pérez-Fernández, V.A.; Prieto-Sánchez, A.; Moreno-Guillen, S.; Muñoz-Fernández, M.-Á.; Pion, M. CD72/CD100 and PD-1/PD-L1 markers are increased on T and B cells in HIV-1+ viremic individuals, and CD72/CD100 axis is correlated with T-cell exhaustion. PLoS ONE 2018, 13, e0203419. [Google Scholar] [CrossRef]
  36. Brooks, D.G.; McGavern, D.B.; Oldstone, M.B.A. Reprogramming of antiviral T cells prevents inactivation and restores T cell activity during persistent viral infection. J. Clin. Investig. 2006, 116, 1675–1685. [Google Scholar] [CrossRef] [Green Version]
  37. Philip, M.; Fairchild, L.; Sun, L.; Horste, E.L.; Camara, S.; Shakiba, M.; Scott, A.C.; Viale, A.; Lauer, P.; Merghoub, T.; et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 2017, 545, 452–456. [Google Scholar] [CrossRef]
  38. Penaloza-MacMaster, P.; Provine, N.M.; Blass, E.; Barouch, D.H. CD4 T Cell Depletion Substantially Augments the Rescue Potential of PD-L1 Blockade for Deeply Exhausted CD8 T Cells. J. Immunol. 2015, 195, 1054–1063. [Google Scholar] [CrossRef] [Green Version]
  39. Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef]
  40. Kurtulus, S.; Madi, A.; Escobar, G.; Klapholz, M.; Nyman, J.; Christian, E.; Pawlak, M.; Dionne, D.; Xia, J.; Rozenblatt-Rosen, O.; et al. Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1CD8+ Tumor-Infiltrating T Cells. Immunity 2019, 50, 181–194.e6. [Google Scholar] [CrossRef] [Green Version]
  41. Utzschneider, D.T.; Charmoy, M.; Chennupati, V.; Pousse, L.; Ferreira, D.P.; Calderon-Copete, S.; Danilo, M.; Alfei, F.; Hofmann, M.; Wieland, D.; et al. T Cell Factor 1-Expressing Memory-like CD8+ T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 2016, 45, 415–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Schenkel, J.M.; Herbst, R.H.; Canner, D.; Li, A.; Hillman, M.; Shanahan, S.-L.; Gibbons, G.; Smith, O.C.; Kim, J.Y.; Westcott, P.; et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 2021, 54, 2338–2353.e6. [Google Scholar] [CrossRef] [PubMed]
  43. Rome, K.S.; Stein, S.J.; Kurachi, M.; Petrovic, J.; Schwartz, G.W.; Mack, E.A.; Uljon, S.; Wu, W.W.; DeHart, A.G.; McClory, S.E.; et al. Trib1 regulates T cell differentiation during chronic infection by restraining the effector program. J. Exp. Med. 2020, 217, e20190888. [Google Scholar] [CrossRef] [PubMed]
  44. Curran, M.A.; Kim, M.; Montalvo, W.; Al-Shamkhani, A.; Allison, J.P. Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS ONE 2011, 6, e19499. [Google Scholar] [CrossRef]
  45. Zander, R.; Schauder, D.; Xin, G.; Nguyen, C.; Wu, X.; Zajac, A.; Cui, W. CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer. Immunity 2019, 51, 1028–1042.e4. [Google Scholar] [CrossRef] [PubMed]
  46. Joshi, N.S.; Cui, W.; Chandele, A.; Lee, H.K.; Urso, D.R.; Hagman, J.; Gapin, L.; Kaech, S.M. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 2007, 27, 281–295. [Google Scholar] [CrossRef] [Green Version]
  47. Backer, R.A.; Hombrink, P.; Helbig, C.; Amsen, D. The Fate Choice Between Effector and Memory T Cell Lineages: Asymmetry, Signal Integration, and Feedback to Create Bistability. Adv. Immunol. 2018, 137, 43–82. [Google Scholar] [CrossRef]
  48. Pipkin, M.E.; Sacks, J.A.; Cruz-Guilloty, F.; Lichtenheld, M.G.; Bevan, M.J.; Rao, A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 2010, 32, 79–90. [Google Scholar] [CrossRef] [Green Version]
  49. Xin, A.; Masson, F.; Liao, Y.; Preston, S.; Guan, T.; Gloury, R.; Olshansky, M.; Lin, J.-X.; Li, P.; Speed, T.P.; et al. A molecular threshold for effector CD8+ T cell differentiation controlled by transcription factors Blimp-1 and T-bet. Nat. Immunol. 2016, 17, 422–432. [Google Scholar] [CrossRef]
  50. Malek, T.R. The biology of interleukin-2. Annu. Rev. Immunol. 2008, 26, 453–479. [Google Scholar] [CrossRef]
  51. Snell, L.M.; MacLeod, B.L.; Law, J.C.; Osokine, I.; Elsaesser, H.J.; Hezaveh, K.; Dickson, R.J.; Gavin, M.A.; Guidos, C.J.; McGaha, T.L.; et al. CD8+ T Cell Priming in Established Chronic Viral Infection Preferentially Directs Differentiation of Memory-like Cells for Sustained Immunity. Immunity 2018, 49, 678–694.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. 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, 2290–2304. [Google Scholar] [CrossRef] [PubMed]
  53. Bénéchet, A.P.; De Simone, G.; Di Lucia, P.; Cilenti, F.; Barbiera, G.; Le Bert, N.; Fumagalli, V.; Lusito, E.; Moalli, F.; Bianchessi, V.; et al. Dynamics and genomic landscape of CD8+ T cells undergoing hepatic priming. Nature 2019, 574, 200–205. [Google Scholar] [CrossRef]
  54. Mueller, S.N.; Ahmed, R. High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proc. Natl. Acad. Sci. USA 2009, 106, 8623–8628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Siddiqui, I.; Schaeuble, K.; Chennupati, V.; Fuertes Marraco, S.A.; Calderon-Copete, S.; Pais Ferreira, D.; Carmona, S.J.; Scarpellino, L.; Gfeller, D.; Pradervand, S.; et al. Intratumoral Tcf1+PD-1+CD8+ T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity 2019, 50, 195–211.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Miller, B.C.; Sen, D.R.; Al Abosy, R.; Bi, K.; Virkud, Y.V.; LaFleur, M.W.; Yates, K.B.; Lako, A.; Felt, K.; Naik, G.S.; et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 2019, 20, 326–336. [Google Scholar] [CrossRef]
  57. Yost, K.E.; Satpathy, A.T.; Wells, D.K.; Qi, Y.; Wang, C.; Kageyama, R.; McNamara, K.L.; Granja, J.M.; Sarin, K.Y.; Brown, R.A.; et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 2019, 25, 1251–1259. [Google Scholar] [CrossRef]
  58. Connolly, K.A.; Kuchroo, M.; Venkat, A.; Khatun, A.; Wang, J.; William, I.; Hornick, N.I.; Fitzgerald, B.L.; Damo, M.; Kasmani, M.Y.; et al. A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci. Immunol. 2021, 6, eabg7836. [Google Scholar] [CrossRef]
  59. Fransen, M.F.; van Hall, T.; Ossendorp, F. Immune Checkpoint Therapy: Tumor Draining Lymph Nodes in the Spotlights. Int. J. Mol. Sci. 2021, 22, 9401. [Google Scholar] [CrossRef]
  60. Fransen, M.F.; Schoonderwoerd, M.; Knopf, P.; Camps, M.G.; Hawinkels, L.J.; Kneilling, M.; van Hall, T.; Ossendorp, F. Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight 2018, 3, e124507. [Google Scholar] [CrossRef] [Green Version]
  61. Dammeijer, F.; van Gulijk, M.; Mulder, E.E.; Lukkes, M.; Klaase, L.; van den Bosch, T.; van Nimwegen, M.; Lau, S.P.; Latupeirissa, K.; Schetters, S.; et al. The PD-1/PD-L1-Checkpoint Restrains T cell Immunity in Tumor-Draining Lymph Nodes. Cancer Cell 2020, 38, 685–700.e8. [Google Scholar] [CrossRef] [PubMed]
  62. Chihara, N.; Madi, A.; Kondo, T.; Zhang, H.; Acharya, N.; Singer, M.; Nyman, J.; Marjanovic, N.D.; Kowalczyk, M.S.; Wang, C.; et al. Induction and transcriptional regulation of the co-inhibitory gene module in T cells. Nature 2018, 558, 454–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Shakiba, M.; Zumbo, P.; Espinosa-Carrasco, G.; Menocal, L.; Dündar, F.; Carson, S.E.; Bruno, E.M.; Sanchez-Rivera, F.J.; Lowe, S.W.; Camara, S.; et al. TCR signal strength defines distinct mechanisms of T cell dysfunction and cancer evasion. J. Exp. Med. 2021, 219, e20201966. [Google Scholar] [CrossRef] [PubMed]
  64. Crawford, A.; Angelosanto, J.M.; Kao, C.; Doering, T.A.; Odorizzi, P.M.; Barnett, B.E.; Wherry, E.J. Molecular and transcriptional basis of CD4+ T cell dysfunction during chronic infection. Immunity 2014, 40, 289–302. [Google Scholar] [CrossRef] [Green Version]
  65. Brooks, D.G.; Teyton, L.; Oldstone, M.B.A.; McGavern, D.B. Intrinsic functional dysregulation of CD4 T cells occurs rapidly following persistent viral infection. J. Virol. 2005, 79, 10514–10527. [Google Scholar] [CrossRef] [Green Version]
  66. Schulze Zur Wiesch, J.; Ciuffreda, D.; Lewis-Ximenez, L.; Kasprowicz, V.; Nolan, B.E.; Streeck, H.; Aneja, J.; Reyor, L.L.; Allen, T.M.; Lohse, A.W.; et al. Broadly directed virus-specific CD4+ T cell responses are primed during acute hepatitis C infection, but rapidly disappear from human blood with viral persistence. J. Exp. Med. 2012, 209, 61–75. [Google Scholar] [CrossRef]
  67. Morou, A.; Palmer, B.E.; Kaufmann, D.E. Distinctive features of CD4+ T cell dysfunction in chronic viral infections. Curr. Opin. HIV AIDS 2014, 9, 446–451. [Google Scholar] [CrossRef] [Green Version]
  68. Brooks, D.G.; Trifilo, M.J.; Edelmann, K.H.; Teyton, L.; McGavern, D.B.; Oldstone, M.B.A. Interleukin-10 determines viral clearance or persistence in vivo. Nat. Med. 2006, 12, 1301–1309. [Google Scholar] [CrossRef]
  69. McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef] [Green Version]
  70. Boettler, T.; Moeckel, F.; Cheng, Y.; Heeg, M.; Salek-Ardakani, S.; Crotty, S.; Croft, M.; von Herrath, M.G. OX40 facilitates control of a persistent virus infection. PLoS Pathog. 2012, 8, e1002913. [Google Scholar] [CrossRef]
  71. Taraban, V.Y.; Rowley, T.F.; O’Brien, L.; Chan, H.T.C.; Haswell, L.E.; Green, M.H.A.; Tutt, A.L.; Glennie, M.J.; Al-Shamkhani, A. Expression and costimulatory effects of the TNF receptor superfamily members CD134 (OX40) and CD137 (4-1BB), and their role in the generation of anti-tumor immune responses. Eur. J. Immunol. 2002, 32, 3617–3627. [Google Scholar] [CrossRef]
  72. Melero, I.; Shuford, W.W.; Newby, S.A.; Aruffo, A.; Ledbetter, J.A.; Hellström, K.E.; Mittler, R.S.; Chen, L. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat. Med. 1997, 3, 682–685. [Google Scholar] [CrossRef] [PubMed]
  73. Cormary, C.; Hiver, E.; Mariamé, B.; Favre, G.; Tilkin-Mariamé, A.-F. Coexpression of CD40L and CD70 by semiallogenic tumor cells induces anti-tumor immunity. Cancer Gene Ther. 2005, 12, 963–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Keller, A.M.; Xiao, Y.; Peperzak, V.; Naik, S.H.; Borst, J. Costimulatory ligand CD70 allows induction of CD8+ T-cell immunity by immature dendritic cells in a vaccination setting. Blood 2009, 113, 5167–5175. [Google Scholar] [CrossRef]
  75. Vezys, V.; Penaloza-MacMaster, P.; Barber, D.L.; Ha, S.-J.; Konieczny, B.; Freeman, G.J.; Mittler, R.S.; Ahmed, R. 4-1BB signaling synergizes with programmed death ligand 1 blockade to augment CD8 T cell responses during chronic viral infection. J. Immunol. 2011, 187, 1634–1642. [Google Scholar] [CrossRef]
  76. Guo, Z.; Wang, X.; Cheng, D.; Xia, Z.; Luan, M.; Zhang, S. PD-1 blockade and OX40 triggering synergistically protects against tumor growth in a murine model of ovarian cancer. PLoS ONE 2014, 9, e89350. [Google Scholar] [CrossRef]
  77. Fan, X.; Quezada, S.A.; Sepulveda, M.A.; Sharma, P.; Allison, J.P. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J. Exp. Med. 2014, 211, 715–725. [Google Scholar] [CrossRef] [Green Version]
  78. Waite., J.; Bei, W.; Lauric, H.; Aynur, H.; Erica, U.; Xuan, Y.; Drew, D.; Rabih, S.; Ajithdoss, D.K.; Godin, S.J.; et al. Tumor-targeted CD28 bispecific antibodies enhance the antitumor efficacy of PD-1 immunotherapy. Sci. Transl. Med. 2020, 12, eaba2325. [Google Scholar] [CrossRef]
  79. Blattman, J.N.; Grayson, J.M.; Wherry, E.J.; Kaech, S.M.; Smith, K.A.; Ahmed, R. Therapeutic use of IL-2 to enhance antiviral T-cell responses in vivo. Nat. Med. 2003, 9, 540–547. [Google Scholar] [CrossRef]
  80. Zippelius, A.; Batard, P.; Rubio-Godoy, V.; Bioley, G.; Liénard, D.; Lejeune, F.; Rimoldi, D.; Guillaume, P.; Meidenbauer, N.; Mackensen, A.; et al. Effector Function of Human Tumor-Specific CD8 T Cells in Melanoma Lesions: A State of Local Functional Tolerance. Cancer Res. 2004, 64, 2865–2873. [Google Scholar] [CrossRef] [Green Version]
  81. Tucker, C.G.; Mitchell, J.S.; Martinov, T.; Burbach, B.J.; Beura, L.K.; Wilson, J.C.; Dwyer, A.J.; Singh, L.M.; Mescher, M.F.; Fife, B.T. Adoptive T Cell Therapy with IL-12-Preconditioned Low-Avidity T Cells Prevents Exhaustion and Results in Enhanced T Cell Activation, Enhanced Tumor Clearance, and Decreased Risk for Autoimmunity. J. Immunol. 2020, 205, 1449–1460. [Google Scholar] [CrossRef] [PubMed]
  82. Rubinstein, M.P.; Cloud, C.A.; Garrett, T.E.; Moore, C.J.; Schwartz, K.M.; Johnson, C.B.; Craig, D.H.; Salem, M.L.; Paulos, C.M.; Cole, D.J. Ex vivo interleukin-12-priming during CD8+ T cell activation dramatically improves adoptive T cell transfer antitumor efficacy in a lymphodepleted host. J. Am. Coll. Surg. 2012, 214, 700–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yi, J.S.; Du, M.; Zajac, A.J. A vital role for interleukin-21 in the control of a chronic viral infection. Science 2009, 324, 1572–1576. [Google Scholar] [CrossRef] [PubMed]
  84. Micci, L.; Ryan, E.S.; Fromentin, R.; Bosinger, S.E.; Harper, J.L.; He, T.; Paganini, S.; Easley, K.A.; Chahroudi, A.; Benne, C.; et al. Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques. J. Clin. Investig. 2015, 125, 4497–4513. [Google Scholar] [CrossRef] [Green Version]
  85. Schurich, A.; Pallett, L.J.; Lubowiecki, M.; Singh, H.D.; Gill, U.S.; Kennedy, P.T.; Nastouli, E.; Tanwar, S.; Rosenberg, W.; Maini, M.K. The third signal cytokine IL-12 rescues the anti-viral function of exhausted HBV-specific CD8 T cells. PLoS Pathog. 2013, 9, e1003208. [Google Scholar] [CrossRef]
  86. Huang, B.; Luo, L.; Wang, J.; He, B.; Feng, R.; Xian, N.; Zhang, Q.; Chen, L.; Huang, G. B7-H3 specific T cells with chimeric antigen receptor and decoy PD-1 receptors eradicate established solid human tumors in mouse models. Oncoimmunology 2019, 9, 1684127. [Google Scholar] [CrossRef] [Green Version]
  87. Titov, A.; Zmievskaya, E.; Ganeeva, I.; Valiullina, A.; Petukhov, A.; Rakhmatullina, A.; Miftakhova, R.; Fainshtein, M.; Rizvanov, A.; Bulatov, E. Adoptive immunotherapy beyond CAR T-cells. Cancers 2021, 13, 743. [Google Scholar] [CrossRef]
  88. West, E.E.; Jin, H.-T.; Rasheed, A.-U.; Penaloza-Macmaster, P.; Ha, S.-J.; Tan, W.G.; Youngblood, B.; Freeman, G.J.; Smith, K.A.; Ahmed, R. PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. J. Clin. Investig. 2013, 123, 2604–2615. [Google Scholar] [CrossRef]
  89. Said, E.A.; Dupuy, F.P.; Trautmann, L.; Zhang, Y.; Shi, Y.; El-Far, M.; Hill, B.J.; Noto, A.; Ancuta, P.; Peretz, Y.; et al. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection. Nat. Med. 2010, 16, 452–459. [Google Scholar] [CrossRef] [Green Version]
  90. Brooks, D.G.; Ha, S.-J.; Elsaesser, H.; Sharpe, A.H.; Freeman, G.J.; Oldstone, M.B.A. IL-10 and PD-L1 operate through distinct pathways to suppress T-cell activity during persistent viral infection. Proc. Natl. Acad. Sci. USA 2008, 105, 20428–20433. [Google Scholar] [CrossRef] [Green Version]
  91. Zarour, H.M. Reversing T-cell dysfunction and exhaustion in cancer. Clin. Cancer Res. 2016, 22, 1856–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Damo, M.; Joshi, N.S. T(reg) cell IL-10 and IL-35 exhaust CD8+ T cells in tumors. Nat. Immunol. 2019, 20, 674–675. [Google Scholar] [CrossRef] [PubMed]
  93. Tinoco, R.; Alcalde, V.; Yang, Y.; Sauer, K.; Zuniga, E.I. Cell-intrinsic transforming growth factor-beta signaling mediates virus-specific CD8+ T cell deletion and viral persistence in vivo. Immunity 2009, 31, 145–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Chen, J.H.; Perry, C.J.; Tsui, Y.-C.; Staron, M.M.; Parish, I.A.; Dominguez, C.X.; Rosenberg, D.W.; Kaech, S.M. Prostaglandin E2 and programmed cell death 1 signaling coordinately impair CTL function and survival during chronic viral infection. Nat. Med. 2015, 21, 327–334. [Google Scholar] [CrossRef]
  95. Ejrnaes, M.; Filippi, C.M.; Martinic, M.M.; Ling, E.M.; Togher, L.M.; Crotty, S.; von Herrath, M.G. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J. Exp. Med. 2006, 203, 2461–2472. [Google Scholar] [CrossRef]
  96. Budhu, S.; Schaer, D.A.; Li, Y.; Toledo-Crow, R.; Panageas, K.; Yang, X.; Zhong, H.; Houghton, A.N.; Silverstein, S.C.; Merghoub, T.; et al. Blockade of surface-bound TGF-β on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment. Sci. Signal. 2017, 10, eaak9702. [Google Scholar] [CrossRef] [Green Version]
  97. Pérez-Mancera, P.A.; Young, A.R.J.; Narita, M. Inside and out: The activities of senescence in cancer. Nat. Rev. Cancer 2014, 14, 547–558. [Google Scholar] [CrossRef] [Green Version]
  98. Akbar, A.N.; Henson, S.M. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat. Rev. Immunol. 2011, 11, 289–295. [Google Scholar] [CrossRef]
  99. Crespo, J.; Sun, H.; Welling, T.H.; Tian, Z.; Zou, W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr. Opin. Immunol. 2013, 25, 214–221. [Google Scholar] [CrossRef] [Green Version]
  100. Liu, X.; Peng, G. T Cell Senescence and Tumor Immunotherapy. In Handbook of Immunosenescence; Springer International Publishing: Berlin/Heidelberg, Germany, 2018; pp. 1–24. [Google Scholar]
  101. Dunne, P.J.; Belaramani, L.; Fletcher, J.M.; de Mattos, S.F.; Lawrenz, M.; Soares, M.V.D.; Rustin, M.H.A.; Lam, E.W.-F.; Salmon, M.; Akbar, A.N. Quiescence and functional reprogramming of Epstein-Barr virus (EBV)—Specific CD8+ T cells during persistent infection. Blood 2005, 106, 558–565. [Google Scholar] [CrossRef]
  102. Dunne, P.J.; Faint, J.M.; Gudgeon, N.H.; Fletcher, J.M.; Plunkett, F.J.; Soares, M.V.D.; Hislop, A.D.; Annels, N.E.; Rickinson, A.B.; Salmon, M.; et al. Epstein-Barr virus—Specific CD8+ T cells that re-express CD45RA are apoptosis-resistant memory cells that retain replicative potential. Blood 2002, 100, 933–940. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, X.; Mo, W.; Ye, J.; Li, L.; Zhang, Y.; Hsueh, E.C.; Hoft, D.F.; Peng, G. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat. Commun. 2018, 9, 249. [Google Scholar] [CrossRef] [PubMed]
  104. Ye, J.; Ma, C.; Hsueh, E.C.; Dou, J.; Mo, W.; Liu, S.; Han, B.; Huang, Y.; Zhang, Y.; Varvares, M.A.; et al. TLR 8 signaling enhances tumor immunity by preventing tumor-induced T-cell senescence. EMBO Mol. Med. 2014, 6, 1294–1311. [Google Scholar] [CrossRef] [PubMed]
  105. Pawelec, G.; Wagner, W.; Adibzadeh, M.; Engel, A. T cell immunosenescence in vitro and in vivo. Exp. Gerontol. 1999, 34, 419–429. [Google Scholar] [CrossRef]
  106. Akbar, A.N.; Henson, S.M.; Lanna, A. Senescence of T Lymphocytes: Implications for Enhancing Human Immunity. Trends Immunol. 2016, 37, 866–876. [Google Scholar] [CrossRef]
  107. Henson, S.M.; Macaulay, R.; Riddell, N.E.; Nunn, C.J.; Akbar, A.N. Blockade of PD-1 or p38 MAP kinase signaling enhances senescent human CD8+ T-cell proliferation by distinct pathways. Eur. J. Immunol. 2015, 45, 1441–1451. [Google Scholar] [CrossRef] [Green Version]
  108. Henson, S.M.; Lanna, A.; Riddel, N.E.; Franzese, O.; Macaulay, R.; Griffiths, S.J.; Puleston, D.J.; Watson, A.S.; Simon, A.K.; Tooze, S.A.; et al. P38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J. Clin. Investig. 2014, 124, 4004–4016. [Google Scholar] [CrossRef]
  109. Zarubin, T.; Han, J. Activation and signaling of the p38 MAP kinase pathway. Cell Res. 2005, 15, 11–18. [Google Scholar] [CrossRef] [Green Version]
  110. Lanna, A.; Coutavas, E.; Levati, L.; Seidel, J.; Rustin, M.H.A.; Henson, S.M.; Akbar, A.N.; Franzese, O. IFN-α Inhibits Telomerase in Human CD8+ T Cells by Both hTERT Downregulation and Induction of p38 MAPK Signaling. J. Immunol. 2013, 191, 3744–3752. [Google Scholar] [CrossRef] [Green Version]
  111. Freund, A.; Patil, C.K.; Campisi, J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011, 30, 1536–1548. [Google Scholar] [CrossRef] [Green Version]
  112. Lanna, A.; Gomes, D.C.O.; Muller-Durovic, B.; McDonnell, T.; Escors, D.; Gilroy, D.W.; Lee, J.H.; Karin, M.; Akbar, A.N. A sestrin-dependent Erk-Jnk-p38 MAPK activation complex inhibits immunity during aging. Nat. Immunol. 2017, 18, 354–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Lanna, A.; Henson, S.M.; Escors, D.; Akbar, A.N. The kinase p38 activated by the metabolic regulator AMPK and scaffold TAB1 drives the senescence of human T cells. Nat. Immunol. 2014, 15, 965–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Pereira, B.I.; De Maeyer, R.P.H.; Covre, L.P.; Nehar-Belaid, D.; Lanna, A.; Ward, S.; Marches, R.; Chambers, E.S.; Gomes, D.C.O.; Riddell, N.E.; et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat. Immunol. 2020, 21, 684–694. [Google Scholar] [CrossRef] [PubMed]
  115. Callender, L.A.; Carroll, E.C.; Beal, R.W.J.; Chambers, E.S.; Nourshargh, S.; Akbar, A.N.; Henson, S.M. Human CD8+ EMRA T cells display a senescence-associated secretory phenotype regulated by p38 MAPK. Aging Cell 2018, 17, e12675. [Google Scholar] [CrossRef]
  116. Ye, J.; Ma, C.; Hsueh, E.C.; Eickhoff, C.S.; Zhang, Y.; Varvares, M.A.; Hoft, D.F.; Peng, G. Tumor-Derived γδ Regulatory T Cells Suppress Innate and Adaptive Immunity through the Induction of Immunosenescence. J. Immunol. 2013, 190, 2403–2414. [Google Scholar] [CrossRef]
  117. Montes, C.L.; Chapoval, A.I.; Nelson, J.; Orhue, V.; Zhang, X.; Schulze, D.H.; Strome, S.E.; Gastman, B.R. Tumor-induced senescent T cells with suppressor function: A potential form of tumor immune evasion. Cancer Res. 2008, 68, 870–879. [Google Scholar] [CrossRef] [Green Version]
  118. Appay, V.; van Lier, R.A.W.; Sallusto, F.; Roederer, M. Phenotype and function of human T lymphocyte subsets: Consensus and issues. Cytometry. A 2008, 73, 975–983. [Google Scholar] [CrossRef]
  119. Koch, S.; Larbi, A.; Derhovanessian, E.; Ozcelik, D.; Naumova, E.; Pawelec, G. Multiparameter flow cytometric analysis of CD4 and CD8 T cell subsets in young and old people. Immun. Ageing 2008, 5, 6. [Google Scholar] [CrossRef] [Green Version]
  120. Callender, L.A.; Carroll, E.C.; Bober, E.A.; Akbar, A.N.; Solito, E.; Henson, S.M. Mitochondrial mass governs the extent of human T cell senescence. Aging Cell 2020, 19, e13067. [Google Scholar] [CrossRef] [Green Version]
  121. Simon, S.; Labarriere, N. PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy? Oncoimmunology 2018, 7. [Google Scholar] [CrossRef]
  122. Winkler, F.; Bengsch, B. Use of Mass Cytometry to Profile Human T Cell Exhaustion. Front. Immunol. 2020, 10, 3039. [Google Scholar] [CrossRef] [PubMed]
  123. Suen, H.; Brown, R.; Yang, S.; Weatherburn, C.; Ho, P.J.; Woodland, N.; Nassif, N.; Barbaro, P.; Bryant, C.; Hart, D.; et al. Multiple myeloma causes clonal T-cell immunosenescence: Identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade. Leukemia 2016, 30, 1716–1724. [Google Scholar] [CrossRef] [PubMed]
  124. Akondy, R.S.; Monson, N.D.; Miller, J.D.; Edupuganti, S.; Teuwen, D.; Wu, H.; Quyyumi, F.; Garg, S.; Altman, J.D.; Del Rio, C.; et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J. Immunol. 2009, 183, 7919–7930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Chen, J.; López-Moyado, I.F.; Seo, H.; Lio, C.-W.J.; Hempleman, L.J.; Sekiya, T.; Yoshimura, A.; Scott-Browne, J.P.; Rao, A. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 2019, 567, 530–534. [Google Scholar] [CrossRef] [PubMed]
  126. Cross, S.J.; Linker, K.E.; Leslie, F.M. THEMIS-SHP1 Recruitment by 4–1BB Tunes LCK-Mediated Priming of Chimeric Antigen Receptor-Redirected T Cells. Physiol. Behav. 2020, 37, 216–225. [Google Scholar] [CrossRef]
  127. Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Kim, M.; Porter, D.L.; June, C.H.; Gill, S. Identification of PD1 and TIM3 As Checkpoints That Limit Chimeric Antigen Receptor T Cell Efficacy in Leukemia. Biol. Blood Marrow Transpl. 2016, 22, S19–S21. [Google Scholar] [CrossRef]
  128. Rupp, L.J.; Schumann, K.; Roybal, K.T.; Gate, R.E.; Ye, C.J.; Lim, W.A.; Marson, A. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci. Rep. 2017, 7, 737. [Google Scholar] [CrossRef] [Green Version]
  129. Yazdanifar, M.; Zhou, R.; Grover, P.; Williams, C.; Bose, M.; Moore, L.J.; Wu, S.T.; Maher, J.; Dreau, D.; Mukherjee, A.P. Overcoming Immunological Resistance Enhances the Efficacy of A Novel Anti-tMUC1-CAR T Cell Treatment against Pancreatic Ductal Adenocarcinoma. Cells 2019, 8, 1070. [Google Scholar] [CrossRef] [Green Version]
  130. Ren, J.; Liu, X.; Fang, C.; Jiang, S.; June, C.H.; Zhao, Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 2017, 23, 2255–2266. [Google Scholar] [CrossRef] [Green Version]
  131. Chong, E.A.; Melenhorst, J.J.; Lacey, S.F.; Ambrose, D.E.; Gonzalez, V.; Levine, B.L.; June, C.H.; Schuster, S.J. PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: Refueling the CAR. Blood 2017, 129, 1039–1041. [Google Scholar] [CrossRef] [Green Version]
  132. Wang, J.; Deng, Q.; Jiang, Y.Y.; Zhang, R.; Zhu, H.B.; Meng, J.X.; Li, Y.M. CAR-T 19 combined with reduced-dose PD-1 blockade therapy for treatment of refractory follicular lymphoma: A case report. Oncol. Lett. 2019, 18, 4415–4420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Hill, B.T.; Roberts, Z.J.; Xue, A.; Rossi, J.M.; Smith, M.R. Rapid tumor regression from PD-1 inhibition after anti-CD19 chimeric antigen receptor T-cell therapy in refractory diffuse large B-cell lymphoma. Bone Marrow Transpl. 2020, 55, 1184–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Yin, Y.; Boesteanu, A.C.; Binder, Z.A.; Xu, C.; Reid, R.A.; Rodriguez, J.L.; Cook, D.R.; Thokala, R.; Blouch, K.; McGettigan-Croce, B.; et al. Checkpoint Blockade Reverses Anergy in IL-13Rα2 Humanized scFv-Based CAR T Cells to Treat Murine and Canine Gliomas. Mol. Ther.-Oncolytics 2018, 11, 20–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Hoogi, S.; Eisenberg, V.; Mayer, S.; Shamul, A.; Barliya, T.; Cohen, C.J. A TIGIT-based chimeric co-stimulatory switch receptor improves T-cell anti-tumor function. J. Immunother. Cancer 2019, 7, 243. [Google Scholar] [CrossRef]
  136. Kloss, C.C.; Lee, J.; Zhang, A.; Chen, F.; Melenhorst, J.J.; Lacey, S.F.; Maus, M.V.; Fraietta, J.A.; Zhao, Y.; June, C.H. Dominant-Negative TGF-β Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation And Augments Prostate Cancer Eradication. Mol. Ther. 2018, 26, 1855–1866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Tang, N.; Cheng, C.; Zhang, X.; Qiao, M.; Li, N.; Mu, W.; Wei, X.F.; Han, W.; Wang, H. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020, 5, e133977. [Google Scholar] [CrossRef] [PubMed]
  138. Masoumi, E.; Jafarzadeh, L.; Mirzaei, H.R.; Alishah, K.; Fallah-Mehrjardi, K.; Rostamian, H.; Khakpoor-Koosheh, M.; Meshkani, R.; Noorbakhsh, F.; Hadjati, J. Genetic and pharmacological targeting of A2a receptor improves function of anti-mesothelin CAR T cells. J. Exp. Clin. Cancer Res. 2020, 39, 1–12. [Google Scholar] [CrossRef]
  139. Beavis, P.A.; Henderson, M.A.; Giuffrida, L.; Mills, J.K.; Sek, K.; Cross, R.S.; Davenport, A.J.; John, L.B.; Mardiana, S.; Slaney, C.Y.; et al. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J. Clin. Investig. 2017, 127, 929–941. [Google Scholar] [CrossRef] [Green Version]
  140. Wiede, F.; Lu, K.; Du, X.; Liang, S.; Hochheiser, K.; Dodd, G.T.; Goh, P.K.; Kearney, C.; Meyran, D.; Beavis, P.A.; et al. PTPN 2 phosphatase deletion in T cells promotes anti-tumour immunity and CAR T-cell efficacy in solid tumours. EMBO J. 2020, 39, 1–26. [Google Scholar] [CrossRef]
  141. Cui, J.; Wang, H.; Medina, R.; Zhang, Q.; Xu, C.; Indig, I.H.; Zhou, J.; Song, Q.; Dmitriev, P.; Sun, M.Y.; et al. Inhibition of PP2A with LB-100 enhances efficacy of CAR-T cell therapy against glioblastoma. Cancers 2020, 12, 139. [Google Scholar] [CrossRef] [Green Version]
  142. Newick, K.; O’Brien, S.; Sun, J.; Kapoor, V.; Maceyko, S.; Lo, A.; Pure, E.; Moon, E.; Albelda, S.M. Augmentation of CAR T cell trafficking and antitumor efficacy by blocking protein kinase A (PKA) localization. Physiol. Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef]
  143. Bowers, J.S.; Majchrzak, K.; Nelson, M.H.; Aksoy, B.A.; Wyatt, M.M.; Smith, A.S.; Bailey, S.R.; Neal, L.R.; Hammerbacher, J.E.; Paulos, C.M. PI3Kδ inhibition enhances the antitumor fitness of adoptively transferred CD8+ T cells. Front. Immunol. 2017, 8, 1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-Man Clinical Trial of CAR NK-92 Cells: Safety Test of CD33-CAR NK-92 Cells in Patients with Relapsed and Refractory Acute Myeloid Leukemia; e-Century Publishing Corporation: Madison, WI, USA, 2018; Volume 8. [Google Scholar]
  145. Alizadeh, D.; Wong, R.A.; Yang, X.; Wang, D.; Pecoraro, J.R.; Kuo, C.; Aguilar, B.; Qi, Y.; Ann, D.K.; Starr, R.; et al. IL15 enhances CAR-T-cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol. Res. 2019, 7, 759–772. [Google Scholar] [CrossRef] [PubMed]
  146. Narayan, D. Inhibition of AKT signaling uncouples T cell differentiation from expansion for receptor-engineered adoptive immunotherapy. J. Comb. Math. Comb. Comput. 2017, 2, e95103. [Google Scholar] [CrossRef] [Green Version]
  147. Bai, Y.; Kan, S.; Zhou, S.; Wang, Y.; Xu, J.; Cooke, J.P.; Wen, J.; Deng, H. Enhancement of the in vivo persistence and antitumor efficacy of CD19 chimeric antigen receptor T cells through the delivery of modified TERT mRNA. Cell Discov. 2015, 1, 15040. [Google Scholar] [CrossRef]
  148. Ninomiya, S.; Narala, N.; Huye, L.; Yagyu, S.; Savoldo, B.; Dotti, G.; Heslop, H.E.; Brenner, M.K.; Rooney, C.M.; Ramos, C.A. Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugs. Blood 2015, 125, 3905–3916. [Google Scholar] [CrossRef] [Green Version]
  149. Fultang, L.; Booth, S.; Yogev, O.; da Costa, B.M.; Tubb, V.; Panetti, S.; Stavrou, V.; Scarpa, U.; Jankevics, A.; Lloyd, G.; et al. Metabolic engineering against the arginine microenvironment enhances CAR-T cell proliferation and therapeutic activity. Blood 2020, 136, 1155–1160. [Google Scholar] [CrossRef]
  150. Ligtenberg, M.A.; Mougiakakos, D.; Mukhopadhyay, M.; Witt, K.; Lladser, A.; Chmielewski, M.; Riet, T.; Abken, H.; Kiessling, R. Coexpressed Catalase Protects Chimeric Antigen Receptor–Redirected T Cells as well as Bystander Cells from Oxidative Stress–Induced Loss of Antitumor Activity. J. Immunol. 2016, 196, 759–766. [Google Scholar] [CrossRef] [Green Version]
  151. Chmielewski, M.; Abken, H. CAR T Cells Releasing IL-18 Convert to T-Bethigh FoxO1low Effectors that Exhibit Augmented Activity against Advanced Solid Tumors. Cell Rep. 2017, 21, 3205–3219. [Google Scholar] [CrossRef] [Green Version]
  152. Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.M.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21, 581–590. [Google Scholar] [CrossRef] [Green Version]
  153. Gomes-Silva, D.; Mukherjee, M.; Srinivasan, M.; Krenciute, G.; Dakhova, O.; Zheng, Y.; Cabral, J.M.S.; Rooney, C.M.; Orange, J.S.; Brenner, M.K.; et al. Tonic 4-1BB Costimulation in Chimeric Antigen Receptors Impedes T Cell Survival and Is Vector-Dependent. Cell Rep. 2017, 21, 17–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Weber, E.W.; Parker, K.R.; Sotillo, E.; Lynn, R.C.; Anbunathan, H.; Lattin, J.; Good, Z.; Belk, J.A.; Daniel, B.; Klysz, D.; et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 2021, 372. [Google Scholar] [CrossRef] [PubMed]
  155. Caulier, B.; Enserink, J.M.; Wälchli, S. Pharmacologic control of car t cells. Int. J. Mol. Sci. 2021, 22, 4320. [Google Scholar] [CrossRef] [PubMed]
  156. Wikenheiser, D.J.; Stumhofer, J.S. ICOS Co-Stimulation: Friend or Foe? Front. Immunol. 2016, 7, 304. [Google Scholar] [CrossRef] [Green Version]
  157. Guedan, S.; Posey, A.D.; Shaw, C.; Wing, A.; Da, T.; Patel, P.R.; McGettigan, S.E.; Casado-Medrano, V.; Kawalekar, O.U.; Uribe-Herranz, M.; et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 2018, 3, e96976. [Google Scholar] [CrossRef] [Green Version]
  158. Guedan, S.; Madar, A.; Casado-Medrano, V.; Shaw, C.; Wing, A.; Liu, F.; Young, R.M.; June, C.H.; Posey, A.D. Single residue in CD28-costimulated CAR-T cells limits long-term persistence and antitumor durability. J. Clin. Investig. 2020, 130, 3087–3097. [Google Scholar] [CrossRef] [Green Version]
  159. Fraietta, J.A.; Lacey, S.F.; Orlando, E.J.; Pruteanu-Malinici, I.; Gohil, M.; Lundh, S.; Boesteanu, A.C.; Wang, Y.; O’connor, R.S.; Hwang, W.-T.T.; et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 2018, 24, 563–571. [Google Scholar] [CrossRef]
  160. Sheih, A.; Voillet, V.; Hanafi, L.A.; DeBerg, H.A.; Yajima, M.; Hawkins, R.; Gersuk, V.; Riddell, S.R.; Maloney, D.G.; Wohlfahrt, M.E.; et al. Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19 CAR-T immunotherapy. Nat. Commun. 2020, 11, 219. [Google Scholar] [CrossRef] [Green Version]
  161. Majzner, R.G.; Frank, M.J.; Mount, C.; Tousley, A.; Kurtz, D.M.; Sworder, B.; Murphy, K.A.; Manousopoulou, A.; Kohler, K.; Rotiroti, M.C.; et al. CD58 Aberrations Limit Durable Responses to CD19 CAR in Large B Cell Lymphoma Patients Treated with Axicabtagene Ciloleucel but Can be Overcome through Novel CAR Engineering. Blood 2020, 136, 53–54. [Google Scholar] [CrossRef]
  162. Ferraro, B.; Walters, J.N.; Reuschel, E.L.; Balakrishnan, A.; Morrow, M.P.; Yan, J.; Khan, A.S.; Sardesai, N.Y.; Humeau, L.M.; Weiner, D.B.; et al. 211. CD2, the First Identified T cell Co-Stimulator, Demonstrates More Effective Chimeric Antigen Receptor Activity Over CD28 and 4-1BB. Mol. Ther. 2015, 23, S83. [Google Scholar] [CrossRef] [Green Version]
  163. Odorizzi, P.M.; Pauken, K.E.; Paley, M.A.; Sharpe, A.; John Wherry, E. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 2015, 212, 1125–1137. [Google Scholar] [CrossRef] [PubMed]
  164. Bucks, C.M.; Norton, J.A.; Boesteanu, A.C.; Mueller, Y.M.; Katsikis, P.D. Chronic Antigen Stimulation Alone Is Sufficient to Drive CD8+ T Cell Exhaustion. J. Immunol. 2009, 182, 6697–6708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Gennert, D.G.; Lynn, R.C.; Granja, J.M.; Weber, E.W.; Mumbach, M.R.; Zhao, Y.; Duren, Z.; Sotillo, E.; Greenleaf, W.J.; Wong, W.H.; et al. Dynamic chromatin regulatory landscape of human CAR T cell exhaustion. Proc. Natl. Acad. Sci. USA 2021, 118, e2104758118. [Google Scholar] [CrossRef] [PubMed]
  166. Martinez, G.J.; Pereira, R.M.; Äijö, T.; Kim, E.Y.; Marangoni, F.; Pipkin, M.E.; Togher, S.; Heissmeyer, V.; Zhang, Y.C.; Crotty, S.; et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 2015, 42, 265–278. [Google Scholar] [CrossRef] [Green Version]
  167. Lynn, R.C.; Weber, E.W.; Sotillo, E.; Gennert, D.; Xu, P.; Good, Z.; Anbunathan, H.; Lattin, J.; Jones, R.; Tieu, V.; et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 2019, 576, 293–300. [Google Scholar] [CrossRef]
  168. Xin, G.; Schauder, D.M.; Lainez, B.; Weinstein, J.S.; Dai, Z.; Chen, Y.; Esplugues, E.; Wen, R.; Wang, D.; Parish, I.A.; et al. A Critical Role of IL-21-Induced BATF in Sustaining CD8-T-Cell-Mediated Chronic Viral Control. Cell Rep. 2015, 13, 1118–1124. [Google Scholar] [CrossRef] [Green Version]
  169. Seo, H.; González-Avalos, E.; Zhang, W.; Ramchandani, P.; Yang, C.; Lio, C.-W.J.; Rao, A.; Hogan, P.G. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol. 2021, 22, 983–995. [Google Scholar] [CrossRef]
  170. Seo, H.; Chen, J.; González-Avalos, E.; Samaniego-Castruita, D.; Das, A.; Wang, Y.H.; López-Moyado, I.F.; Georges, R.O.; Zhang, W.; Onodera, A.; et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl. Acad. Sci. USA 2019, 116, 12410–12415. [Google Scholar] [CrossRef] [Green Version]
  171. Yao, C.; Sun, H.-W.; Lacey, N.E.; Ji, Y.; Moseman, E.A.; Shih, H.-Y.; Heuston, E.F.; Kirby, M.; Anderson, S.; Cheng, J.; et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 2019, 20, 890–901. [Google Scholar] [CrossRef]
  172. Khan, O.; Giles, J.R.; McDonald, S.; Manne, S.; Ngiow, S.F.; Patel, K.P.; Werner, M.T.; Huang, A.C.; Alexander, K.A.; Wu, J.E.; et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 2019, 571, 211–218. [Google Scholar] [CrossRef]
  173. Alfei, F.; Kanev, K.; Hofmann, M.; Wu, M.; Ghoneim, H.E.; Roelli, P.; Utzschneider, D.T.; von Hoesslin, M.; Cullen, J.G.; Fan, Y.; et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 2019, 571, 265–269. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, X.; Naranjo, A.; Brown, C.E.; Bautista, C.; Wong, C.W.; Chang, W.C.; Aguilar, B.; Ostberg, J.R.; Riddell, S.R.; Forman, S.J.; et al. Phenotypic and functional attributes of lentivirus-modified CD19-specific Human CD8+ central memory T cells manufactured at clinical scale. J. Immunother. 2012, 35, 689–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Terakura, S.; Yamamoto, T.N.; Gardner, R.A.; Turtle, C.J.; Jensen, M.C.; Riddell, S.R. Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. Blood 2012, 119, 72–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Rossi, J.; Paczkowski, P.; Shen, Y.-W.; Morse, K.; Flynn, B.; Kaiser, A.; Ng, C.; Gallatin, K.; Cain, T.; Fan, R.; et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 2018, 132, 804–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Agarwal, S.; Hanauer, J.D.S.; Frank, A.M.; Riechert, V.; Thalheimer, F.B.; Buchholz, C.J. In Vivo Generation of CAR T Cells Selectively in Human CD4+ Lymphocytes. Mol. Ther. 2020, 28, 1783–1794. [Google Scholar] [CrossRef] [PubMed]
  178. Xhangolli, I.; Dura, B.; Lee, G.H.; Kim, D.; Xiao, Y.; Fan, R. Single-cell Analysis of CAR-T Cell Activation Reveals A Mixed TH1/TH2 Response Independent of Differentiation. Genomics. Proteom. Bioinform. 2019, 17, 129–139. [Google Scholar] [CrossRef]
  179. Wang, D.; Aguilar, B.; Starr, R.; Alizadeh, D.; Brito, A.; Sarkissian, A.; Ostberg, J.R.; Forman, S.J.; Brown, C.E. Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight 2018, 3, e99048. [Google Scholar] [CrossRef] [Green Version]
  180. Yang, Y.; Kohler, M.E.; Chien, C.D.; Sauter, C.T.; Jacoby, E.; Yan, C.; Hu, Y.; Wanhainen, K.; Qin, H.; Fry, T.J. TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci. Transl. Med. 2017, 9, eaag1209. [Google Scholar] [CrossRef] [Green Version]
  181. Liadi, I.; Singh, H.; Romain, G.; Rey-Villamizar, N.; Merouane, A.; Adolacion, J.R.T.; Kebriaei, P.; Huls, H.; Qiu, P.; Roysam, B.; et al. Individual motile CD4+ T cells can participate in efficient multikilling through conjugation to multiple tumor cells. Cancer Immunol. Res. 2015, 3, 473–482. [Google Scholar] [CrossRef] [Green Version]
  182. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Investig. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [Green Version]
  183. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Hudecek, M.; Pender, B.; Robinson, E.; Hawkins, R.; Chaney, C.; Cherian, S.; Chen, X.; et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med. 2016, 8, 355ra116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Sommermeyer, D.; Hudecek, M.; Kosasih, P.L.; Gogishvili, T.; Maloney, D.G.; Turtle, C.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016, 30, 492–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Guha, P.; Cunetta, M.; Somasundar, P.; Espat, N.J.; Junghans, R.P.; Katz, S.C. Frontline Science: Functionally impaired geriatric CAR-T cells rescued by increased α5β1 integrin expression. J. Leukoc. Biol. 2017, 102, 201–208. [Google Scholar] [CrossRef] [PubMed]
  186. Li, G.; Smithey, M.J.; Rudd, B.D.; Nikolich-Žugich, J. Age-associated alterations in CD8α+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium. Aging Cell 2012, 11, 968–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Klebanoff, C.A.; Scott, C.D.; Leonardi, A.J.; Yamamoto, T.N.; Cruz, A.C.; Ouyang, C.; Ramaswamy, M.; Roychoudhuri, R.; Ji, Y.; Eil, R.L.; et al. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. J. Clin. Investig. 2016, 126, 318–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Mikuła-Pietrasik, J.; Witucka, A.; Pakuła, M.; Uruski, P.; Begier-Krasińska, B.; Niklas, A.; Tykarski, A.; Książek, K. Comprehensive review on how platinum- and taxane-based chemotherapy of ovarian cancer affects biology of normal cells. Cell. Mol. Life Sci. 2019, 76, 681–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Perkins, D.W.; Haider, S.; Robertson, D.; Buus, R.; O’leary, L.; Isacke, C.M. Therapy-induced senescence in normal tissue promotes breast cancer metastasis. bioRxiv 2020. [Google Scholar] [CrossRef]
  190. Das, R.K.; Storm, J.; Barrett, D.M. Abstract 1631: T cell dysfunction in pediatric cancer patients at diagnosis and after chemotherapy can limit chimeric antigen receptor potential. In Proceedings of the Cancer Research, Chicago, IL, USA, 14–18 April 2018; American Association for Cancer Research (AACR): Philadelphia, PA, US, 2018; Volume 78, p. 1631. [Google Scholar]
  191. Das, R.K.; O’Connor, R.S.; Grupp, S.A.; Barrett, D.M. Lingering effects of chemotherapy on mature T cells impair proliferation. Blood Adv. 2020, 4, 4653–4664. [Google Scholar] [CrossRef]
  192. Fagnoni, F.F.; Lozza, L.; Zibera, C.; Zambelli, A.; Ponchio, L.; Gibelli, N.; Oliviero, B.; Pavesi, L.; Gennari, R.; Vescovini, R.; et al. T-cell dynamics after high-dose chemotherapy in adults: Elucidation of the elusive CD8+ subset reveals multiple homeostatic T-cell compartments with distinct implications for immune competence. Immunology 2002, 106, 27. [Google Scholar] [CrossRef]
  193. Haining, W.N.; Neuberg, D.S.; Keczkemethy, H.L.; Evans, J.W.; Rivoli, S.; Gelman, R.; Rosenblatt, H.M.; Shearer, W.T.; Guenaga, J.; Douek, D.C.; et al. Antigen-specific T-cell memory is preserved in children treated for acute lymphoblastic leukemia. Blood 2005, 106, 1749. [Google Scholar] [CrossRef]
  194. Du, M.; Hari, P.; Hu, Y.; Mei, H. Biomarkers in individualized management of chimeric antigen receptor T cell therapy. Biomark. Res. 2020, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  195. Louis, C.U.; Savoldo, B.; Dotti, G.; Pule, M.; Yvon, E.; Myers, G.D.; Rossig, C.; Russell, H.V.; Diouf, O.; Liu, E.; et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 2011, 118, 6050–6056. [Google Scholar] [CrossRef] [PubMed]
  196. Garfall, A.L.; Dancy, E.K.; Cohen, A.D.; Hwang, W.T.; Fraietta, J.A.; Davis, M.M.; Levine, B.L.; Siegel, D.L.; Stadtmauer, E.A.; Vogl, D.T.; et al. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma. Blood Adv. 2019, 3, 2812–2815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Abramson, J.S.; Lia, P.M.; Gordon, L.I.; Lunning, M.A.; Arnason, J.E.; Wang, M. High Durable CR Rates in Relapsed/Refractory (R/R) Aggressive B-NHL Treated with the CD19-Directed CAR T Cell Product JCAR017 (TRANSCEND NHL 001): Defined Composition Allows for Dose-Finding and Definition of Pivotal Cohort. Blood 2017, 130, 581. [Google Scholar]
  198. Titov, A.; Petukhov, A.; Staliarova, A.; Motorin, D.; Bulatov, E.; Shuvalov, O.; Soond, S.M.; Piacentini, M.; Melino, G.; Zaritskey, A.; et al. The biological basis and clinical symptoms of CAR-T therapy-associated toxicites. Cell Death Dis. 2018, 9, 897. [Google Scholar] [CrossRef]
Figure 1. The landscape and evolution of T cell dysfunction. Depending on the priming condition (given at the bottom of the figure), the trajectory of the T cell development may be driven from naïve T cells to (1) anergic T cells; (2) memory T cells; (3) terminal effector T cells; and (4) exhausted T cells (Tex). Memory T cells remain susceptible to exhaustion. Tex cell pool includes progenitor Tex that are primarily responsive to PD-1 blockade and sustain proliferative potential. They give rise to nearly completely dysfunctional terminal Tex or (under certain circumstances) differentiate into highly cytotoxic Tex effector-like state. In some cancers, T cells with a typical progenitor Tex phenotype do not respond to checkpoint inhibition. Both terminally exhausted and effector/senescent T cells are characterized by negligible proliferation; however, the latter demonstrate significant cytotoxicity.
Figure 1. The landscape and evolution of T cell dysfunction. Depending on the priming condition (given at the bottom of the figure), the trajectory of the T cell development may be driven from naïve T cells to (1) anergic T cells; (2) memory T cells; (3) terminal effector T cells; and (4) exhausted T cells (Tex). Memory T cells remain susceptible to exhaustion. Tex cell pool includes progenitor Tex that are primarily responsive to PD-1 blockade and sustain proliferative potential. They give rise to nearly completely dysfunctional terminal Tex or (under certain circumstances) differentiate into highly cytotoxic Tex effector-like state. In some cancers, T cells with a typical progenitor Tex phenotype do not respond to checkpoint inhibition. Both terminally exhausted and effector/senescent T cells are characterized by negligible proliferation; however, the latter demonstrate significant cytotoxicity.
Cancers 14 01078 g001
Figure 2. Factors affecting functionality, exhaustion, and senescence of CAR-T cells. PBMCs obtained through apheresis vary substantially in composition and quality. In particular, chemotherapy and older age may result in preemptive T cellular senescence. Apheresis product may be also enriched with Tregs or exhausted cells. These may significantly affect the final CAR-T cell product. At the same time, balanced CD4+/CD8+ composition and enrichment with naïve cells are known to be beneficial for cell functionality in the clinical setting. Finally, carefully validated CAR design and manufacturing process, e.g., IL-15/IL-7-based expansion, are essential in this context. On the contrary, chemotherapeutic treatment may lead to senescence and poor persistence of CAR-T cells. Several factors are in turn responsible for exhaustion/dysfunction of CAR-T cells in vivo.
Figure 2. Factors affecting functionality, exhaustion, and senescence of CAR-T cells. PBMCs obtained through apheresis vary substantially in composition and quality. In particular, chemotherapy and older age may result in preemptive T cellular senescence. Apheresis product may be also enriched with Tregs or exhausted cells. These may significantly affect the final CAR-T cell product. At the same time, balanced CD4+/CD8+ composition and enrichment with naïve cells are known to be beneficial for cell functionality in the clinical setting. Finally, carefully validated CAR design and manufacturing process, e.g., IL-15/IL-7-based expansion, are essential in this context. On the contrary, chemotherapeutic treatment may lead to senescence and poor persistence of CAR-T cells. Several factors are in turn responsible for exhaustion/dysfunction of CAR-T cells in vivo.
Cancers 14 01078 g002
Table 1. Modifications of the known T cell dysfunction pathways in CAR-T cells.
Table 1. Modifications of the known T cell dysfunction pathways in CAR-T cells.
Modified Pathway/MoleculeMolecule TypeUpstream SignalingDownstream Signaling (Activation or Inhibition)ModificationTumor Model (Animal, Cell Line) and Observed EffectCAR Construct, Introduction MethodReference
A2aR (adenosine receptor A2)surface moleculeadenosinecAMPshRNA knockdown
A2aR antagonist (SCH-58261)
in vitro, HeLa
no significant reduction in cytotoxic function in presence of the adenosine agonist NECA (5′-(N-ethylcarboxamide), higher IL-2 expression in comparison to unmodified cells
mesothelin-BBz,
lentiviral
[138]
A2aR (adenosine receptor A2)surface molecule adenosinecAMPshRNA knockdown
anti-PD-1 antibody
in vivo, Ly 5.1 mice + 24JK-HER2+ or E0771-HER2+
increased cytokine production of CD8+ and activation of CD8+ and CD4+ CAR-T, particularly with PD-1 blockade;
significant survival advantage in mice
HER2-28z,
retroviral
[139]
ADAR1 (adenosine deaminase RNA specific) cytoplasmic enzymeIFN Type Ibiogenesis of members of the miR-222 family -> ICAM1 expression -> immune resistance EHNA drug (ADAR1 inhibitor) in vitro, HPAFII, CFPAC, MiaPaCa2
combination with EHNA did not result in reduced survival of the target cells
MUC-1-28z, retroviral [129]
AKTintracellular signal transducerPI3KMAPK, FOXO1AKT inhibitor VIII (AKTi)in vitro, NALM6
AKTi repressed glycolysis;
in vivo, NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice + NALM6
AKTi exposed CAR-T elicit significant increase in animal survival
CD19,
retroviral
[146]
Argininosuccinate synthase, ornithine transcarbamylase cancer-associated enzymes constitutivearginine, ornithineKnockin argininosuccinate synthase (ASS) and/or ornithine transcarbamylase (OTC) enzymesin vivo, arginine-depleted NOG-SCID mice + GD2+ SKNMC, KELLY, and LAN-1;
expression of ASS, OTC, or ASS+OTC enhanced proliferation of CAR-T; derived from multiple human donors, regardless of scFv
GD2-BBz
CD33-BBz
mesothelin-BBz
EGFRvIII-BBz
[149]
Catalaseintracellular enzymeconstitutiveH2O2coexpression of catalase with CARin vitro, SkoV3-Her2+
reduced oxidative state, both basal and upon activation,
enhanced proliferation and preserved target cell lysis in presence of H2O2
CEA-28z,
HER2-29z,
retroviral
[150]
CTLA-4surface moleculeCD80/CD86SHP-2, PP2Acheckpoint blockade by CAR-T-secreted minibodies (reduced checkpoint inhibitors)in vivo, NSG mice + D270,
Hu08-BBz CAR-T cells also showed enhanced tumor reduction when used in combination with anti-CTLA-4, whereas 2173BBz CAR-T cells did not benefit from CTLA-4 checkpoint blockade
see TIM-3 section[134]
Cyclooxygenase (COX-1, COX-2)cytoplasmic enzymesconstitutive/inflammation (NFkB)prostaglandin E2celecoxib (specific COX-2 inhibitor) and indomethacin (COX1 and 2 inhibitor)in vitro,
HPAFII and CFPAC cells showed a reduction in survival,
MiaPaCa2 cells showed no difference in survival; Celecoxib did not change the efficacy of CAR-T cells
MUC-1-28z,
retroviral
[129]
Telomerasecancer/memory T-cell-associated enzymep38; DDRRestores telomere lengthtransient delivery of telomerase mRNAin vitro, Raji
prolonged proliferation and inhibited cell senescence;
in vivo, NPG/Vst mice + Raji,
improved persistence, proliferation, and long-term antitumor effects
CD19-28z,
CD19-BBz,
lentiviral
[147]
IDO-1 (indoleamine 2,3-dioxygenase)enzyme with high activity in cancer cellsconstitutivekynurenineIDO inhibitor (1-methyl-tryptophan)in vivo, SCID-Beige mice + Raji/Raji-IDO
CAR-T inhibited IDO-negative but not IDO-positive tumors growth; IDO inhibitor restored IDO-positive tumor control; tryptophan metabolites inhibited expansion, proliferation, cytotoxicity, cytokine secretion, and increased CAR-T apoptosis; 4-1 BB intracellular domain had no effect on the inhibition
CD19-z/BBz,
retroviral
[148]
IL-12, IL-18cytokines-STAT4inducible single-chain p45-p30 IL-12 (iIL-12); 18-kD IL-18 (iIL-18); constitutive IL-18 and IL-12in vivo, CEA transgenic C57BL/6 mice +
Panc02-CEA+
and Rag2/γc/mice + A549 CEA+
iIL-18 CAR-T exhibited superior activity against large pancreatic and lung tumors refractory to CAR-T cells without cytokines. IL-18 induced overall change in the immune microenvironment
CEA-28z,
retroviral
[151]
mTORC1intracellular signal transducercalcineurin/DAPK1mTORC2/T-betrapamycin (mTOR inhibitor)
expansion with IL-15
in vivo, NSG mice, Raji
IL15-expanded CAR-T mediated superior antitumor activity, longer persistence, and significantly greater survival than IL-2-expanded CAR-T;
IL2/rapamycin-cultured CAR-T shared phenotypic features with IL-15-CAR-T, suggesting that IL15- mediated reduction of mTORC1 activity is responsible for preserving less differentiated phenotype
CD19 second generation,
IL13Rα2 second generation,
lentiviral
[145]
PD-1surface moleculePD-L1/2SHP-1, SHP-2antibodiesin vitro MOLM-14, primary leukemia
improved cytokine production and Ki-67 proliferation marker, especially in combinational treatment with PD-1 and TIM-3 antibodies;
in vivo, NSG mice + MOLM14
increased durable complete response rate
CD33-BBz,
CD123-BBz,
lentiviral
[127]
PD-1surface moleculePD-L1/2SHP-1, SHP-2CRISPR/Cas9 KOin vivo, NSG mice + CD19+PD-1L+ K562,
improved clearance of tumor
CD19-BBz,
lentiviral
[128]
PD-1surface moleculePD-L1/2SHP-1, SHP-2checkpoint blockade by CAR-T-secreted minibodies (reduced checkpoint inhibitors)see TIM-3 section[144]
PD-1surface moleculePD-L1/2SHP-1,
SHP-2
anti-PD-1 Ab in vitro,
no improvement in the killing of the resistant cell lines (HPAFII, CFPAC), while sensitive cell line (MiaPaCa2) killing was enhanced
MUC-1-28z,
retroviral
[129]
PD-1surface moleculePD-L1/2SHP-1,
SHP-2
pembrolisumab (anti-PD-1 Ab)clinical case report, mediastinal B cell lymphoma;
remission continuing 12 months posttherapy
CD19-BBz,
lentiviral
[131]
PD-1surface moleculePD-L1/2SHP-1,
SHP-2
nivolumab (anti-PD-1 Ab)clinical case report, diffuse large B cell lymphoma
short tumor volume reduction, lasting 2 months, followed by progression
CD19-BBz[132]
PD-1surface moleculePD-L1/2SHP-1,
SHP-2
nivolumab (anti-PD-1 Ab)clinical case report, refractory follicular lymphoma
remission lasted for >10 months.
CD19-28z,
Axicabtagene ciloleucel (Kymriah®),
retroviral
[133]
PD-1surface moleculePD-L1/2SHP-1,
SHP-2
CRISPR-Cas9 KOin vitro, PC3;
in vivo, NSG mice + NALM6/NALM6-PD-L1+;
increased cytotoxicity
PSCA-second generation,
lentiviral
[130]
PI3Kδ (phosphatidylinositol-3-kinase p110δ)intracellular signal transducerTCR and costimulatory molecules (CD28, 4-1BB, and ICOS)AKTIdelalisib and AKT inhibitor VIII (AKTi)in vivo, NSG mice + M108;
Idelalisib-treated CAR-T cells exerted longer tumor control compared to the AKTi-treated;
Idelalisib T cells have improved engraftment, persistence, less differentiated phenotype, and transcriptional signature
mesothelin-BBz,
lentiviral
[143]
PI3Kδ (phosphatidylinositol-3-kinase p110δ)intracellular signal transducerTCR and costimulatory molecules (CD28, 4-1BB, and ICOS)AKTPI3K inhibitor LY294002in vitro, MOLM-13
increased effector molecules expression, less differentiated phenotype, higher cell number
CD33-BBz,
MOLM-13 target cells (CD33+),
retroviral
[144]
PKA (protein kinase A)intracellular signal transducercAMPCsktransduction with regulatory subunit I anchoring disruptor (RIAD)in vitro, AE17ova, PDA4662 cells,
increased cytokine release;
in vivo, C57BL/6 (strain CD45.2) + EM-meso+ and NSG mice + AE17-meso+;
adenosine or PGE2 did not affect RIAD CAR-T; higher median reduction in tumor volume
mesothelin-BBz,
lentiviral (human),
retroviral (mice)
[142]
PP2A (protein phosphatase 2A)intracellular signal transducerconstitutiveAKT, mTOR, Erk, CaMKKII/IVprotein phosphatase 2A (PP2A) inhibitor (LB-100)in vitro, U251-Luc
significantly increased cytotoxicity in a dose-dependent manner;
in vivo, NSG mice + U251-Luc,
significant increase in CD3+ cells within tumor tissue and more frequent tumor regression
CAIX (carbonic anhydrase IX)-BBz[141]
PTPN-2 (protein tyrosine phosphatase nonreceptor type 2)constitutivec-SrcPTPN2-inhibitor compound 8siRNA duplexes transient knockdown, CRISPR-Cas9 KOin vivo, female Ly5.1 B6.SJL-Ptprc a Pepc b/BoyJ, human HER-2 transgenic + E0771-HER-2+;
improved immune surveillance on spontaneous tumors and after adoptive transfer
murine CAR-T: human HER-2-28z,
human CAR-T: human LeY-28z,
retroviral
[140]
SHP-1/THEMIS complexintracellular signal transducerPD-1; CD19-BBz CD3ζknockdown THEMIS or SHP1 in CD19-BBz- CAR-T cells, shRNAin vitro, BV173-CD19+;
increased CAR-CD3ζ; basal phosphorylation
CD19-BBz,
CD19-28
[126]
TGF-β receptor IIsurface moleculeTGF-βSMAD2/3/4knockin (together with CAR), dominant-negative TGF-βRII in vivo, NSG mice + PC3;
increased proliferation, cytokine secretion, resistance to exhaustion, long-term in vivo persistence, induction tumor eradication in aggressive human prostate cancer
PSMA-BBz,
lentiviral
[136]
TGF-β receptor IIsurface moleculeTGF-βSMAD2/3/4CRISPR/Cas9 KOin vitro, mesothelin+ CRL5826 and OVCAR-3,
improved killing;
in vivo, NPG mice + CRL5826;
better controlling tumor growth;
TGF-β-RII KO outperform PD-1 KO in efficacy;
PD-1 KO complement; TGF-β-RII KO
Mesothelin-28z,
lentiviral
[137]
TIGITsurface moleculeCD155 and CD112SHP-1; b-arrestin/SHIP1-mediated downstream inhibition of NF-kB, PI3K and MAPK pathwaysTIGIT-28 chimeric switch receptor (TIGIT exo- + CD28 signaling domain)in vitro, Raji, JY, 721.221, Nalm6;
improved killing and cytokine secretion
CD19-BBz,
MSGV1-based,
retroviral
[135]
TIM-3surface moleculegalectin-9CD45; CD148checkpoint blockade by CAR-T-secreted minibodies (reduced checkpoint inhibitors)in vivo, NSG mice + D270;
significant inhibition of tumor growth with either 2173BBz or Hu08BBz CAR-T cells and anti-PD-1/anti-TIM-3
anti-IL-13Rα2-BBz (humanized Hu08BBz, murine 2173BBz),
lentiviral
[134]
TIM-3surface moleculegalectin-9CD45; CD148Gal-9 blocking Abin vitro,
HPAFII and CFPAC cells—improvement;
MiaPaCa2 cells—no improvement
MUC-1-28z, retroviral [129]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Titov, A.; Kaminskiy, Y.; Ganeeva, I.; Zmievskaya, E.; Valiullina, A.; Rakhmatullina, A.; Petukhov, A.; Miftakhova, R.; Rizvanov, A.; Bulatov, E. Knowns and Unknowns about CAR-T Cell Dysfunction. Cancers 2022, 14, 1078. https://doi.org/10.3390/cancers14041078

AMA Style

Titov A, Kaminskiy Y, Ganeeva I, Zmievskaya E, Valiullina A, Rakhmatullina A, Petukhov A, Miftakhova R, Rizvanov A, Bulatov E. Knowns and Unknowns about CAR-T Cell Dysfunction. Cancers. 2022; 14(4):1078. https://doi.org/10.3390/cancers14041078

Chicago/Turabian Style

Titov, Aleksei, Yaroslav Kaminskiy, Irina Ganeeva, Ekaterina Zmievskaya, Aygul Valiullina, Aygul Rakhmatullina, Alexey Petukhov, Regina Miftakhova, Albert Rizvanov, and Emil Bulatov. 2022. "Knowns and Unknowns about CAR-T Cell Dysfunction" Cancers 14, no. 4: 1078. https://doi.org/10.3390/cancers14041078

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop