Abstract
Second generation chimeric antigen receptors (CARs) retarget and reprogram T lymphocytes to augment their anti-tumour efficacy. The combined activating and costimulatory domains incorporated in these CARs critically determine the function, differentiation, metabolism and persistence of engineered T cells. CD19 CARs that incorporate CD28 or 4–1BB signalling domains are the best known to date. Both have shown remarkable complete remission rates in patients with refractory B cell malignancies. Recent data indicate that CD28-based CARs direct a brisk proliferative response and boost effector functions, while 4–1BB-based CARs, direct a gradual T cell accumulation that may eventually overcome lesser functional potency. These distinct kinetic features can be exploited to further develop CAR T cell therapies for a variety of cancers. A new field of immuno-pharmacology is emerging.
Introduction
The goal of T cell engineering (TCE) is to rapidly generate potent and specific immune responses.1, 2 TCE is predicated on methods to safely and effectively genetically modify human T lymphocytes, which became available in the mid 1990’s. TCE allows to instruct T cells to recognize any antigen of interest, for example a tumour antigen. Targeting may be achieved through the transfer of a physiological receptor for antigen, which is known as the T cell receptor (TCR),3, 4 or artificial, T cell-activating receptors, which encompass a diverse set of fusion receptors that we eventually grouped under the general term of chimeric antigen receptor (CAR).5 Emulating the TCR, the first chimeric receptors were designed to mimic its T cell activation, a function that is sufficient to specify cytolysis, but not sufficient to direct a sustained, effective T cell response. Such activating receptors are now referred to as first generation CARs.5 To augment T cell function and extend T cell life span, a different kind of receptor had to be designed. Receptors providing both activating and costimulatory signals were designed to augment T cell function as well as redirect T cell specificity.5 These second generation CARs have recently yielded impressive clinical results in patients with B cell malignancies, especially acute lymphoblastic leukaemia6–11 and B cell lymphomas.12–15
Here we review the nature and biological effects of engineered costimulation in tumour-targeted T cells, focusing on the pharmacology of second generation CARs. The best know to date are those that utilize the signalling domains of the CD28 or 4–1BB costimulatory receptors, fused to the cytoplasmic domain of the T cell activating CD3ζ chain.16, 17 Hereafter we refer to these two categories of receptors as 28ζ and BBζ CARs.
Principles of T cell activation and CAR design
Physiological T cell responses are initiated by the TCR, which imparts antigen specificity and acts as the gatekeeper of T cell activation.18, 19 TCR signalling is amplified and modulated by a series of receptors known as costimulatory receptors, the prototype of which is CD28.20 In the absence of costimulation, primary TCR stimulation may fail to drive T cell proliferation beyond the G0/G1 phase of the cell cycle and render the T cell unresponsive, a state known as anergy.21 Costimulation prevents anergy, as well as some forms of activation-induced cell death (AICD) and T cell exhaustion, although senescence will eventually limit T cell function and/or survival.
The first chimeric T cell activating receptors were generated in the early 90’s when the ζ chain of the CD3 complex was cloned.22 As this T cell-specific chain lacks an extracellular domain, the groups of Weiss, Seed and Klausner fused it to the extracellular domain of CD8, CD4 or CD25 (Fig. 1A) to study its function in leukemic T cells.23–25 The cross-linking of these chimeric receptors induced calcium influx and other hallmarks of early T cell activation, thus establishing the physiological function of the ζ chain and providing a road map for the design of T cell activating receptors. The addition of a single chain variable fragment (scFv) to these receptors, as first reported by Eshhar et al,26 afforded binding to the hapten TNP and antigen-induced T cell activation (Fig 1B). These fusion receptors were shown to redirect specificity and induce proliferation in T cell hybridomas and pre-activated T cells, but not in naïve cells.27, 28 Transgenic mouse models later revealed that T cells expressing ζ chain-based CARs only modestly delayed tumour progression in vivo, as they only produced low amounts of interferon-γ (IFNγ) and rapidly anergised.28, 29 Having established methods for transduction of human T cells (based on γ-retroviral mediated gene transfer30), we were able, for the first time, to test ζ chain-based CARs in primary T lymphocytes. Although these receptors effectively mediated cytotoxicity, we found them to be unable to direct T cell expansion upon repeated exposure to antigen.31
Figure 1: Chimeric Antigen Receptor evolution.
A: First fusion receptors as developed by the Weiss,23 Seed24 and Klausner25 teams, coupling CD3ζ to the CD4, CD8 or CD25 extracellular domains. B: First generation CAR (or T body), allowing for antigen recognition through addition of an scFv.26 C: Chimeric cytokine receptor, coupling an scFv to the intracellular domain of CD28 to induce antigen-specific costimulation.32 D: Second generation CARs, facilitating T cell reprogramming by incorporating a costimulatory-signalling domain in tandem with the CD3ζ chain.16
In earlier studies, we had demonstrated that costimulation can be provided to T cells through antigen-specific receptors termed chimeric costimulatory receptors (CCRs, Fig 1C). A CD28-based CCR was shown to direct Interleukin (IL)-2 synthesis and offset activation-induced apoptosis in human primary T cells.32 Having thus validated the costimulatory signal in a relevant cell type, we then fused our CCR construct with a ζ chain-based CAR (Fig. 1D). For the first time, we were able to engineer human primary T cells such that they can expand upon repeated exposure to antigen.16 Finney et al and Hombach et al reported on similar dual domain receptors, showing increased cytokine secretion induced by CARs containing a costimulatory signalling domain.33, 34 Multiple costimulatory domains have since been introduced into CARs, the best known of which are those incorporating CD28 or 4–1BB signalling elements.16, 17 These have both been utilized in patients with B cell malignancies. Here we focus on the pharmacological properties afforded by these receptors, and also briefly summarize other second generation CAR designs.
CD28 costimulation
The TCR alone is not sufficient to prime naïve T cells to clonally expand and differentiate into effector and memory T cells. For a productive immune response to develop, T cell activation requires a concomitant costimulatory signal provided by CD28.35, 36 CD28 is a member of the immunoglobulin superfamily of costimulatory/inhibitory receptors, which also includes ICOS (inducible costimulator), BTLA (B- and T-lymphocyte attenuator), CTLA-4 (cytotoxic T lymphocyte-associated antigen 4) and PD-1 (programmed death receptor-1).37–40 CD28 is a 44kD type I transmembrane protein, expressed as a glycosylated, disulfide-linked homodimer.41, 42 It is expressed in 95% of human CD4+ T cells and 50% CD8+ T cells43 (unlike in the mouse where it is homogenously expressed on CD4+ as well as CD8+ T cells44) as well as regulatory T cells (TREGs), plasma cells, neutrophils, eosinophils, and Natural Killer T (NKT) cells.45–49
Together with the TCR, CD28 is recruited to microclusters in the immunological synapse that forms between T cells and antigen presenting cells (APCs). The main function of CD28 is to augment TCR signalling, resulting in increased cytokine production, clonal proliferation, differentiation and survival. In doing so, CD28 increases TCR sensitivity by lowering the threshold TCR engagement that is required for efficient T cell activation50 and enables T cell responses against weak agonist peptides.51 Although it was thought early on that CD28 may have its own signalling pathway, it is now known that CD28 essentially amplifies TCR signalling,52, 53 affecting cytokine production,54, 55 cell cycle progression,56 apoptosis,57, epigenetic structure58 and metabolism.59, 60
The cytoplasmic tail recruits the PI3K/Akt pathway, as well as the kinases PKCθ, Lck and RAS.20 Lck and PKCθ recruitment induces Ca2+ influx and activation of the transcription factor (TF) NFAT, resulting in upregulation of T-bet, a TF that induces Th1 cytokine production.55 Increased expression of the cytokines IL-2, IFNγ, IL-4 and the macrophage inflammatory protein MIP1α, as well as cytokine and chemokine receptors, is induced.52 CD28 costimulation is critical for the induction of IL-2 secretion.54 IL-2 secretion is induced through GRB2, AP-1 dependent RAS signalling61 as well as via NFAT.20 Additionally, CD28 induces chromatin remodelling via c-Rel (an NF-κB-family TF)62, rendering the IL-2 locus accessible, and thereby allowing for a more rapid secondary response.63, 64 Similar changes are induced to other cytokine loci including IL-4 and IFNγ.65 Cell-cycle progression is enhanced via upregulation of Cyclin-D, facilitating progression to late G1 and S phases.56 The recruitment of the PI3K/Akt pathway enhances the expression of Glut-1, mTor and c-Myc, as well as NFAT and the canonical NF-κB pathway.66 Glut-1, mTor and c-Myc facilitate the required metabolic switch from fatty acid, pyruvate and glutamine oxidation67–69 to aerobic glycolysis and glutaminolysis to meet the energy and biosynthesis requirements associated with T cell activation.70–72 PI3K/Akt also induces the upregulation of Bcl-XL and suppression of p73 (both in an NF-κB dependent manner73, 74) which, combined with inhibition of FasL expression and reduction of caspase 8 activation, enhances cell survival.75, 76 Through ERK/JNK dependent upregulation of the co-stimulatory receptors ICOS, CD40L and 4–1BB, CD28 facilitates a sustained secondary response.77–79 Importantly, CD28 costimulation is not only required during T cell priming, but also contributes to the expansion, cytolytic function, IL-2 production and cell cycle completion of memory T cells.80, 81
CD28 signalling is regulated in multiple ways. First, activation of CD28 requires binding with its ligands CD80 or CD86. Low levels of CD86 are constitutively expressed on B-cells, dendritic cells (DCs), Langerhans cells, monocytes and activated CD4+ T-cells, and are upregulated upon activation, as is CD80.82, 83 Both ligands induce the same signalling cascades, although it has been suggested that CD86 plays a greater role in initiation and CD80 in maintenance of immune responses.66, 82 Second, after repeated TCR and CD28:CD80 stimulation, CTLA-4 and TNF (tumour necrosis factor) down-regulate CD28.84–86 However, stimulation through 4–1BB can reconstitute CD28 expression and promote T cell reactivation and IL-2 production.87, 88 Third, CD28 signalling is inhibited via CTLA-4 and PD-1. Cell surface CTLA-4 is induced upon T-cell activation and is a high affinity receptor for CD80/86. It inhibits CD28 signalling by competing for receptor binding as well as enhancing the internalization and degradation of CD28 and trans-endocytosis of CD80/86.86, 89–91 PD-1 expression is induced within 24hrs after TCR stimulation and induces T cell exhaustion through blockade of CD28-induced PI3K activity.92–94
Due to the importance of CD28 in both T cell priming and memory responses, incorporation of CD28 signals into a second generation CAR is thus a fundamental and logical way to amplify CAR signalling, promote T cell proliferation and persistence, and offset anergy,
4–1BB costimulation
Following T cell priming, a number of costimulatory receptors appear on the surface of T cells in order to sustain activation. This includes members of the tumour necrosis factor receptor superfamily (TNFRSF), such as OX40, CD27, CD30, HVEM and 4–1BB, also known as CD137 or TNFRSF9.95
4–1BB is a type II transmembrane protein that was first found in activated lymphocytes.96–99 It is expressed on a variety of lymphoid cells,100–103 as well as some non-haematopoietic tissues.99, 104, 105 Unbound 4–1BB is expressed as a 30kD monomer on the T cell surface, which forms a trimeric complex upon binding to its ligand. 4–1BBL is expressed on activated APCs as well as on activated T-cells.106–110 4–1BBL is distinct from the other members of the TNFR family by its three-bladed propeller like structure.111 Formation of the trimeric complex results in both signalling downstream of 4–1BB as well as downstream of 4–1BBL.112
4–1BB is transiently induced by TCR and CD28 signalling via the ERK and JNK signalling pathways in both CD4+ and CD8+ T-cells, faster and more durably in the latter.113–115 In memory cells, 4–1BB can be induced by IL-15 in the absence of antigen stimulation.116
4–1BB signalling enhances T-cell proliferation, cell cycle progression, cytokine secretion and cytolytic potential as well as prevention of clonal deletion and AICD, and reduces sensitivity to Transforming Growth Factor (TGF)β suppression.117, 118 4–1BB-mediated costimulation increases IFNγ and IL-2 secretion by CD8+ T cells and IL-2 and IL-4 secretion by CD4+ T cells.119, 120 It enhances TCR signalling through tyrosine phosphorylation of SLP-76, CD3ε, CD3ζ and Lck as well as the recruitment of PKCθ and increase in intracellular Ca2+ levels.117 Signalling is mediated by TNF Receptor Associated Factor (TRAF)−1, −2 and −3.121–124 TRAF1 is essential for the activation of ERK, the upregulation of Bcl-XL and downregulation of Bim.125 Additionally, it restricts the activation of the non-canonical NF-κB pathway in the absence of costimulation by preventing NIK (NF-κB inducing kinase) activation, but enhances the canonical NF-κB pathway upon costimulation.126 TRAF2 is essential for the activation of p38 and the induction of NF-κB dependent activation of Bcl-XL and Bfl-1, and also induces cytokine production via the MAPK pathway, promoting Th1 differentiation and cytokine production in CD4+ T cells.123, 127, 128 TRAF3 moderates activation through suppression of the non-canonical NF-κB pathway.129 In both CD4+ and CD8+ T-cells, 4–1BB induces cell cycle progression in an IL-2 independent manner via ERK1/2 and PI3K and in an IL-2 dependent manner via the PI3K/Akt pathway.130 4–1BB signals are potentiated in the presence of CD28 signalling, but 4–1BB can still be upregulated in CD28null CD8+ memory T cells wherein it exerts CD28-independent costimulation.131–134 Similar to CD28 signalling, 4–1BB is able to induce telomerase activity, enhancing its levels after CD3/CD28 stimulation and induce re-activation of CD28 unresponsive T cells.135
4–1BB signalling affects the size, quality and maintenance of the memory CD8+ T cell pool, and T cell expansion upon secondary challenge.136–138 4–1BB can amplify T cell proliferation in both CD8+ and CD4+ T cells.114, 137 Although 4–1BB−/− and 4–1BBL−/− mice do not show a defect in CD4:CD8 T-cell ratios, 4–1BB−/− mice have a reduced number of primary CD8+ T-cells due to reduced proliferation of naïve cells,139, 140 a reduced CD8+ T cell response and a reduced CD8 memory pool.141, 142
4–1BB stimulation can rescue T cells from anergy and exhaustion, even after down-regulation of CD28.143, 144 However, the timing of 4–1BB signalling is of great importance. 4–1BB activation early after viral infection can have a detrimental effect by inducing AICD through prolonged up-regulation of TNF and Fas.102, 145
The incorporation of 4–1BB signalling domains in second generation CARs is therefore a logical choice to prevent anergy and to promote T cell proliferation and memory, with the anticipation of a greater effect on T cell maintenance than on functional activation relative to CD28.
28ζ and BBζ CARs–structure and signalling functions
Whereas a large amount of biological data on CD28, 4–1BB and some other costimulatory receptors is available, less is known about the function of their costimulatory domains within CARs. It would be mistaken to extrapolate all of the physiological functions of natural receptors to CARs for a number of reasons. First, the recruitment of these domains does not follow interactions with a single or sometimes two physiological ligands but varies, depending on the level of antigen on the target cell, the level of expression of the CAR in the T cell and the affinity of the CAR. There also is a temporal and spatial difference with the expression pattern of the natural receptors, due to the constitutive expression of the vector-encoded CAR and the covalent linkage of the costimulatory and activating domains. Furthermore, receptors such as 4–1BB are monomers that normally trimerize upon activation, but they are forced dimers in most CAR designs. Additionally, the nature of the synapse that second generation CARs form with antigen presenting cells may not be the same as TCR-centred synapses. Finally, CAR functionality is not solely determined by the cytoplasmic signalling domains, as other structural features may affect its overall function (Box 1). All in all, the pharmacology of recombinant costimulatory receptors is an emerging field in need of more experimentation. There are presently few data on CAR signalling or comprehensive comparisons between CARs. Functionality
Box 1 – Structural features of CARs.
The functional properties of CARs are primarily determined by the signalling domains that are arrayed in the cytoplasmic tail. However, other structural components influence the overall signalling properties.
Binding Moiety
The binding moiety enables HLA-independent antigen recognition. The affinity of the CAR for the antigen and the location of the epitope on the targeted antigen affect overall CAR function.26 CAR affinity, along with CAR expression levels, will determine the antigen-binding characteristics of the CAR and the effectiveness of target cell recognition.
Hinge
The hinge or spacer domain provides a separation between the binding moiety and the T cell membrane. The requirement for a hinge is target dependent.222 To establish an optimal effector:target inter-membrane distance, membrane-distal epitopes do not require a large hinge within the CAR; in contrast, membrane-proximal epitopes elicit suboptimal T cell activation in the absence of a hinge.223 Longer flexible hinges also allow for better binding to relatively inaccessible epitopes.206, 224 The hinge can also have detrimental effects. Cross-activation can occur between IgG1 Fc-hinge containing CAR T cells and FcγR+ cells, resulting in unspecific innate immune activation and AICD.222, 225
Transmembrane domain
The transmembrane domain (TM) is considered to be a purely structural requirement. Within first generation CARs, CD4 and CD8 TM domains allowed for similar cytokine secretion.226 However, the CD3ζ TM domain was shown to facilitate interaction between the CAR and endogenous CD3, allowing for hetero-dimerisation with the endogenous TCR, resulting in enhanced anti-tumour function and cytokine production.227

The initial characterization of any new CAR typically consists of functional in vitro assays, including measurements of antigen-specific cytotoxicity, antigen-induced proliferation and cytokine production. The most informative reports utilize primary cells, an experimental setting that requires efficient and non-toxic T cell transduction, such as that afforded by γ-retroviral or lentiviral vectors. Studies in leukaemic cells or hybridomas are less dependable, given the profound genetic and functional alterations that affect the proliferation, apoptosis, differentiation and function of these cells. It should also be appreciated that CAR studies in primary T cell studies are most commonly restimulation studies, in so far that T cells are activated and go through S phase prior to transduction,146, 147 such that the first activation through the CAR (in vitro as well as in vivo) is de facto a second stimulation.
Both 28ζ and BBζ CARs augment cytokine secretion relative to first generation CARs.16, 17, 148 Both induce Th1 cytokines including IL-2, IFNγ, TNF, and GM-CSF, although more briskly for 28ζ CARs and more slowly for BBζ CARs.149–156 Th2 cytokines including IL-4 and IL-10 are also produced, with markedly lower levels induced by BBζ constructs.150, 156, 157 28ζ CARs elicit more IL-2, although it should be noted that the level of IL-2 production elicited by different 28ζ CARs varies, potentially exceeding or under-achieving that afforded by endogenous CD28.16, 148 IL-2 is a critical cytokine for T cell function and adoptive cell therapy.158 The impact of CAR-induced IL-2 secretion is complex. IL-2 promotes CAR T cell proliferation and sustains effector function, and may also affect neighbouring cells such as NK cells or TREGs. While TREG support, if it occurs, is undesirable, 28ζ CAR T cells are less sensitive to TREG inhibition via IL-10 and TGFβ than first generation CAR T cells.159 Furthermore, a modified CD28 domain reducing IL-2 production can partially restore cytolytic function of 28ζ CAR T cells in the presence of TREGs without affecting IFNγ secretion and T cell proliferation.160 TREGs resistance can also be achieved by providing IL-12 to 28ζ CAR T cells161 or through constitutive Akt activity.162
Limited information is available on the signalling cascades induced by CARs. In leukaemia cell lines, Finney et al showed that second generation constructs containing either CD28, ICOS, OX40 or 4–1BB domains, do not show increased tyrosine phosphorylation compared to ζ-chain only constructs.155 Consistent with the known CD28 biology, 28ζ CARs induce signalling via NF-κB, Akt, ERK and NFAT and induce T-bet, EOMES and GATA3.163 28ζ CARs have been shown to activate the PI3K pathway, more consistently than BBζ CARs.152, 155, 164
A key factor in the specificity and safety of CAR T cells is their dependence on antigen stimulation to induce signalling. Some second generation CARs have been reported to possess constitutive or tonic activity. CD19-specific BBζ T cells show increased proliferation in vitro and enhanced in vivo survival in the absence of antigen150, 165 This may be due in part to the unnatural dimeric 4–1BB structure, as TRAF recruitment is highly dependent on 4–1BB conformational changes.166 Tonic signalling has not been observed with CD28 CARs,16, 150 although it may occur when the scFv’s oligomerise and induce CAR clustering and downstream signalling.163 Antigen independent activation due to scFv crosslinking can be reduced by selection of an appropriate Heavy/Light chain orientation, hinge region or use of ligand-derived binding moieties instead of an scFv.
28ζ and BBζ CARs–anti-tumour efficacy in preclinical models
Over a decade ago, we provided the first demonstration that CD19 CAR therapy utilizing human peripheral blood T lymphocytes can eradicate established lymphoma and leukaemia in immunodeficient mice.167 In this study, the use of a first generation CAR required a high T cell dose and optimized T cell expansion in the presence of CD28 costimulation and IL-15.167 We later showed that a 28ζ CAR vastly outperformed a first generation CAR in an aggressive acute lymphoblastic leukaemia (ALL) model148 (and likewise in a syngeneic CD19 mouse model168). An initial comparison of a 28ζ and a BBζ CAR showed similar anti-tumour activity, albeit with higher accumulation of BBζ T cells, even after the tumour had been eradicated.149
Little detail is known about CAR-mediated tumour elimination. The anti-tumour effect of CAR T cells may be mediated by direct tumour cytolysis as well as cytokine secretion. Interestingly, direct cytolysis is not confined to CD8+ T cells, as CD4+ CAR T cells can also acquire strong cytolytic potential.167, 169 IFNγ and TNF produced by CD4+ and CD8+ T cells are able to damage the tumour microvasculature and induce tumour cell cycle arrest and senescence.170–172
Complementing functionality, T cell persistence is another major determinant of anti-tumour activity. Both CD28 and 4–1BB costimulatory domains extend T cell survival compared to first generation CARs,148, 150, 173 albeit with different characteristics. 28ζ CARs program higher functionality while BBζ CAR direct greater longevity. The mechanisms for the latter remain to be fully elucidated. Two studies showed a higher induction of the antiapoptotic factor Bcl-XL by 28ζ compared to BBζ CARs, accompanied by reduced AICD,152, 154 but in vitro Bcl-XL induction may not directly correlate with in vivo persistence. Telomerase activity might also contribute to the differences in persistence. One study showed that a 28ζ CAR induced peak levels of telomerase 2 days after antigen stimulation, which declined by day 4, whereas a BBζ CAR induced more persistent activity.174
Increased in vivo persistence of BBζ CAR T cells may also result from reduced exhaustion and apoptosis upon repeated antigen stimulation. For example, PD-1 upregulation in T cells following CAR stimulation, combined with the induction of PD-L1 on tumour cells by IFNγ, could reduce T cell function and ultimately T cell longevity. Combined 28ζ CAR T cells and PD-1 blocking antibody thus enhances proliferative and functional T cell capacity in vitro and anti-tumour functionality in vivo, associated with reduced infiltration of myeloid derived suppressor cells.175 It was also shown that engineered production of IL-15 by 28ζ CAR T-cells can lead to a reduction in PD-1 expression and enhanced functionality.176 The significance of PD-1 expression, however, needs to be interpreted with caution. In tumour-infiltrating T cells, PD-1+ CD8+ T cells are potent tumour-reactive T cells that produce copious amounts of IFNγ production and up-regulate 4–1BB expression.177 Nonetheless, the combination of PD-1/PD-L1 blockade with CAR T cell therapy, especially 28ζ CARs, is an attractive perspective.
CAR T cell functionality is commonly studied in bulk CD4+ and CD8+ T cells. Recent reports have shown that, in the context of 28ζ constructs targeting Mesothelin169 or CD19,178 CD4+ CAR T cells have a stronger expansion and higher cytokine secretion compared to CD8+ CAR T cells, and are more resistant to AICD. CD4+ CAR T cells alone may achieve tumour control and establish long-term persistence, and their presence enhances the anti-tumour activity of CD8+ CAR T cells.169
The engineering of T cells at different maturational stages is another important determinant of T cell anti-tumour activity, a topic that is beyond the scope of this review and has been recently reviewed elsewhere.179, 180
28ζ and BBζ CARs in the clinic: The CD19 paradigm
The most investigated target for CARs to date is CD19. We initially chose CD19 as a target for CAR-based therapy because of its common expression in most B cell leukaemias and lymphomas and absence in all normal tissues other than the B cell lineage.181, 182 We provided the first proof-of-principle that CAR-modified human peripheral blood T cells targeted to CD19 can eradicate a broad range of B cell malignancies.167 Successful B cell tumour eradication was eventually obtained with different CD19 CARs,148, 150, 173, 183 paving the way for numerous on-going clinical trials. The targeting of CD19 eventually became a paradigm for evaluating CAR technology.184 CD19 malignancies are so far the only setting where both 28ζ and BBζ CARs have been studied in the clinic.
While the first clinical reports on CD19 CAR trial focused on non-Hodgkin lymphoma (NHL)185–187 and chronic lymphocytic leukaemia (CLL),6, 13–15, 188, 189 the most dramatic results have been obtained in ALL.7–11 Three centres made seminal contributions to these clinical studies – Memorial Sloan Kettering Cancer Center (MSKCC) for adult ALL, and the Children’s Hospital of Philadelphia (CHOP) and the National Cancer Institute (NCI) for childhood ALL. The first clinical results obtained with CAR therapy utilized the 19–28z CAR (Fig. 2), which we transduced in autologous peripheral blood T cells collected by apheresis.7, 190 Adult patients with relapsed, chemorefractory disease were infused with 3 million CAR+ T cells/kg following a single infusion of cyclophosphamide (3 g/m2) as conditioning. Four out of four patients with measurable disease went into molecular remission within 4 weeks.7 The groups of Stephen Grupp, Crystal Mackall, and ours subsequently published follow-up studies in adult and paediatric disease.8–11 All three centres reported a highly remarkable complete remission rate – a rare occurrence for phase I studies in oncology and especially for relapsed ALL. All reported the same two main toxicities: B cell aplasia, which is expected when targeting CD19,148, 168 and cytokine responses, sometimes causing severe cytokine release syndrome.8, 9, 191 Similar toxicities have been seen with antibody therapies targeting costimulatory pathways (Box 2).
Figure 2: Second generation anti-CD19-CARs used in clinical trials to treat Acute Lymphoblastic Leukaemia (ALL).
A. CAR tested in MKSCC-trials,6, 7, 9 consisting of SJ25C1 scFv, CD28 spacer, trans-membrane and signalling domain and CD3ζ chain16 B. CAR used in clinical trials at NCI.10 This CAR contains the FMC63 scFv but is otherwise the same as the MSKCC CAR shown in A. C. CAR used and developed by the Baylor College of Medicine team.188 This CAR utilizes the FMC63 scFv, IgG1 hinge, CD4 transmembrane domain, CD28 signalling domain and CD3ζ chain. D. Construct developed at St Jude Children’s Research Hospital17 and used by the CHOP and UPenn teams.8, 11 This CAR contains the FMC63 scFv, CD8α hinge, and 4–1BB signalling domain in tandem with the CD3ζ chain.
Box 2. Costimulation targeted by monoclonal antibodies.
Costimulatory pathways may be recruited to support T cell function via second generation CARs or by using monoclonal antibodies (mAbs) that target costimulatory receptors.228 These mAbs, which may exert agonistic or antagonistic effects, are potent modulators of tumour immunity. Like CAR T cells, they may also induce immune toxicities. The CD28 superagonist TGN1412 mAb activates T cells bypassing TCR stimulation and other costimulatory signals. It may trigger massive cytokine release, known as a cytokine storm, associated with pulmonary infiltrates, acute lung injury, acute renal failure followed by disseminated intravascular coagulation.229 The 4–1BB agonist mAb BMS-663513 has been tested in patients with solid tumours. Objective responses were observed in 17% of melanoma patients and 14% of renal cell carcinoma patients at low doses, but high doses may cause liver toxicity or fatalities.230–232 Agonists of OX40 and CD40 are under investigation for multiple tumours, inducing potent responses, with occasional grade 3–4 toxicity.232–237 Trials investigating targeting of CD27 and GITR are on-going.
Immune checkpoint blockade using antagonists against CTLA-4 (Ipilimumab) and PD-1 (Pembrolizumab) has been approved by the US FDA. The best results have been seen in metastatic melanoma, where anti-CTLA-4 treatment increases 3-year survival rates to 20%, and anti-PD-1 results in 32–38% overall responses.238, 239 Targeting of the PD1/PD-L1 axis can also be achieved through blocking of PD-L1. In a variety of solid tumours, 6–17% OR was achieved.240 Several additional anti-CTLA-4, anti-PD-1 and anti-PD-L1 trials are on-going. Immune checkpoint blockade is also associated with occasional immune related adverse events. This includes fatigue, rash, dermatitis, uveitis, neurotoxicity, hepatotoxicity, pancreatitis, nephritis and pneumonitis. Rare fatalities due to anti-CTLA-4 induced enterocolitis, colonic perforation, hepatotoxicity and cardiac arrests have been reported.239, 241 Fatal pneumonitis has also resulted from anti-PD-1 treatment.242 These outcomes underscore the potency and enormous potential of targeting costimulatory pathways to augment tumour immunity and the need, whether using CAR T cells or mAbs, to tame and carefully manage these powerful drugs.

While these studies follow overall similar steps (apheresis, retroviral CAR transduction, T cell infusion following chemotherapy conditioning), they differ in several regards, including the CAR design (28ζ dual-signalling domain16 utilized at the NCI and MSKCC, BBζ17 utilized at CHOP Fig 2), T cell manufacturing, conditioning chemotherapy, patient age, tumour burden, tumour chemo-sensitivity, and T cell dosage.192 It is striking that irrespective of these variances, the outcomes have been similar, which speaks to the extraordinary robustness of CD19 CAR therapy in ALL. The differences in procedures and patient characteristics, as well as the still low number of patients (approximately 74 published and an estimated 150–200 infused at the time of this writing), makes it difficult to yet draw mechanistic conclusions. Some observations nonetheless emerge. CAR T cells traffic very effectively to bone marrow, whether they express a 28ζ or BBζ CAR.9, 14 They occasionally cause severe cytokine release, especially in patients with higher tumour burden.7, 11 A fever tends to start early on in recipients of 28ζ CAR T cells, later in recipient of BBζ CAR T cells. BBζ CAR T cells may persist many months in paediatric ALL patients,8, 11 but less so in adults, where persistence of 28ζ or BBζ CAR T cells is on the order of 1–3 months.9, 11 More patients need to be studied in detail to better apprehend the function and persistence of CAR T cells in different patient populations.
One next frontier to explore is the activity of second generation CARs against solid tumours. Such clinical studies are just starting in a handful of centres. Expectations notwithstanding, patience will be needed – several years – before sufficient data are gathered to evaluate the therapeutic potential of second generation CARs in multiple tumour types.
Beyond 28ζ and BBζ CARs
It is unlikely that there is one universally optimal costimulatory domain for CAR-induced T cell activation. Optimal signals may differ in different T cell subsets or T cell differention stages or tumour microenvironments. Costimulation is a dynamic process to which multiple receptors contribute. Several of these have already been incorporated into CARs.
ICOS
Like CD28, ICOS (Inducible Costimulator) is a member of the B7 family, although it is not expressed constitutively but upregulated upon TCR/CD28 signalling. ICOS activates the PI3K/AKT pathway, but is unable to recruit GRB2 and Lck and therefore fails to induce strong IL-2 production. However, through activation of C-MAF, it induces higher secretion of other cytokines such as IL-4 and IL-21.36 ICOS is required for Th17 development156, 193, 194 and plays a critical role in the induction of Bcl-6, which is required for the transition from effector to memory phenotype.36, 195 ICOS deficiency affects both T cell responses, resulting in reduced levels of TEFF, TMEM and TREGs, and cytokine deficits.196–198 ICOS has been incorporated in multiple second generation CAR constructs, targeting either CD33, EGFRvIII or mesothelin.155, 156, 199 ICOSζ CARs have higher PI3K activation and IFNγ production than BBζ CARs.155 In Th17 cells, ICOSζ CARs maintain a bipolar Th17/Th1 phenotype and secrete a distinct cytokine profile, including IL17A, IL-17F and IL-22. In one study, the anti-tumour effect of one ICOSζ CAR was not greater than a 28ζ and BBζ CAR, although it resulted in greater CD4+ T cells persistence.156
OX40
Like 4–1BB, OX40 is a member of the TNFRSF, expressed by activated T cells after TCR/CD28 stimulation. OX40 deficiency affects CD8 T cell responses and reduces CD4 T cell proliferation and effector differentiation.200–202 Its signalling is mediated by TRAF2, TRAF3 as well as TRAF5, inducing the canonical and non-canonical NF-κB pathways.203, 204 OX40-based CARs targeting CD33 have been developed and their characteristics appear to be overall similar to BBζ constructs in terms of proliferative support and cytokine secretion.155 Numerous third generation constructs incorporating OX40, targeting GD2, MUC1 or CEA have been assessed.157, 205, 206
CD27
CD27 is also a member of the TNFRSF, but one that is constitutively expressed by naïve and memory T cells. TRAF2 or TRAF5 binding induces the (non)canonical NF-κB pathways, as well as AP-1 downstream of the JNK/MAPK pathway.203 CD27 stimulation results in IL-2, IFNγ and TNF production, as well as the upregulation of Bcl-XL and memory formation. However, chronic CD27 signalling induces T cell apoptosis.203 CD27 is able to induce T cell expansion in an IL-2 independent manner207 and stimulates glycolysis, aiding transition of naïve and memory to effector T cell.208 Absence of CD27 results in reduced T cell expansion and impaired memory responses.136, 202 The incorporation of CD27 into second generation CARs targeting folate receptor-α induces T cell proliferation, Bcl-XL expression and IFNγ production, displaying comparable in vivo efficacy with 28ζ and BBζ CARs in one study.153
NKG2D
In a CAR design that does not utilize an scFv for ligand (antigen) binding, NKG2D CARs utilize the NK-cell receptor NKG2D to provide costimulation via interaction with the endogenous DAP10 co-signalling domain, providing PI3K/AKT stimulation and promoting a Th1 response.209 NKG2D ligands are expressed in a variety of tumour cells as well as normal tissues, especially in the context of inflammation.209, 210 Different types of NKG2D-based CARs have been developed, using either the full length or the extracellular domain of NKG2D, combined with CD3ζ, CD28 and/or 4–1BB domains.211, 212 In vitro, expression of NKG2D-based CARs in T or NK cells induces lysis of multiple tumour targets and displays a high capacity to secrete cytokines including INFγ, TNF and GM-CSF. In vivo, full length NKG2D-CD3ζ CARs provide a better immune response than T cells transduced with NKG2D alone.213, 214
Other combinatorial strategies
Costimulatory signalling domains may also be combined, either in cis (third generation CARs) or in trans (combined CAR and CCR expression). Briefly, third generation CARs incorporate two costimulatory domains within their cytoplasmic tail. Most commonly, CD28 and 4–1BB149–152, 154, 215, 216 or CD28 and OX-40157, 205, 206 domains have been combined Third generation CARs showed similar or improved in vitro and in vivo cytolytic function compared to other CARs. Cytokine production levels vary in comparison to second-generation CARs, with both increases and decreases reported.150, 152 T cell proliferation and in vivo persistence may be increased in some cases.149, 152, 215 Few clinical trials have utilized third generation CARs to date. These did not suggest a clinical advantage over second generation CARs.217, 218 When combining costimulation in trans through two independent antigen-specific receptors such as a CCR and a CAR,32, 219 two antigens are targeted, e.g., ErbB2 and Mucin-1,220 PSMA and PSCA,219 and mesothelin and α–folate receptor.221 Dual antigen targeting may be used to enhance tumour targeting,220, 221 or reinforce tumour selectivity.219
Conclusions
CARs that incorporate costimulatory domains are conceptually distinct from those that only elicit T cell activation. The latter effectively redirect cytotoxicity, but dual-signalling CARs further reprogram T cell function and T cell persistence. These second generation CARs have recently yielded exciting clinical results in patients with B cell malignancies. While more is to be learned about the molecular functions of these CARs, both 28ζ and BBζ CARs have yielded high complete remission rates in B cell malignancies, especially ALL. Emerging data, albeit still incomplete, suggest that 28ζ CARs provide a brisker proliferative response and more vigorous activation of effector functions than BBζ CARs. The latter however persist longer, and achieve comparable anti-tumour responses over a more protracted time period. This suggests that BBζ CAR T cells compensate for their lesser functional potency by gradually building up and sustaining their numbers. These different kinetics may be exploited in different ways to further enhance the efficacy of CAR T cells and tailor CAR therapy to the requirements of different tumour burdens and tumour microenvironments. More studies are needed to better understand CAR function in different T cell types and to adjust CAR signalling to balance its effects on T cell function and persistence. A new field of immune-pharmacology is born.
Glossary Terms
- Anergy
Rapidly acquired state of T cell unresponsiveness occurring after suboptimal activation via the TCR in the absence of costimulation or in the presence of co-inhibitory molecules
- Activation-induced Cell Death (AICD)
apoptosis occurring following T cell activation
- Exhaustion
progressive hyporesponsiveness developed by T cells after repeated exposure to antigen
- T cell Engineering
cell modification intended to alter T cell antigen specificity, proliferation, persistence and/or function
- Chimeric Antigen Receptor
Fusion receptor combining antigen-recognition and T cell activating properties. CARs consist of an antigen binding moiety (scFv or ligand), a hinge, a transmembrane domain and a cytoplasmic signaling domain, which includes a costimulatory domain in second generation CARs
- 1st Gen CAR
CAR with an endodomain consisting of the cytoplasmic signalling domain of CD3ζ or the Fc-γ receptor
- 2nd Gen CAR
CAR design including a single costimulatory element within the endoplasmic domain in trans with the activating CD3z domain
- 3rd Gen CAR
CAR design containing two costimulatory elements within the cytoplasmic domain in trans with the activating CD3z domain
- scFv
single-chain variable fragment; fusion protein consisting of the variable fragments of the immunoglobulin heavy- and light chains, connected via a glycine/serine linker
- Hinge
also referred to as spacer domain, it is an extracellular component connecting the binding moiety to the trans-membrane element
- Chimeric Costimulatory Receptor
Fusion molecule coupling antigen-specificity to T cell costimulatory signalling, without activating domains
- Primary T cell response
response upon the first encounter with foreign antigen, in which naive lymphocytes are primed, resulting in differentiation and clonal expansion
- Secondary T cell response
response upon a repeat encounter with a foreign antigen, in which memory lymphocytes are activated resulting in stronger clonal expansion and enhanced antigen eradication compared to the primary response
- Molecular remission
complete remission and absence of disease based on PCR analysis
- Complete remission
absence of disease based on imaging or biopsy analysis
- Severe cytokine release syndrome
A clinical syndrome comprising fever, hypotension, hypoxia and/or neurologic symptoms, associated with cytokine release occurring following in vivo T cell activation
References
- 1.Sadelain M, Riviere I & Brentjens R Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer 3, 35–45 (2003). [DOI] [PubMed] [Google Scholar]
- 2.Ho WY, Blattman JN, Dossett ML, Yee C & Greenberg PD Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction. Cancer Cell 3, 431–7 (2003). [DOI] [PubMed] [Google Scholar]
- 3.Morgan RA et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314, 126–9 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dossett ML et al. Adoptive immunotherapy of disseminated leukemia with TCR-transduced, CD8+ T cells expressing a known endogenous TCR. Mol Ther 17, 742–9 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sadelain M, Brentjens R & Riviere I The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol 21, 215–23 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Brentjens RJ et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–28 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brentjens RJ et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5, 177ra38 (2013).First report of complete remission in 4 out of 4 subjects with ALL following CD19 CAR T cell therapy
- 8.Grupp SA et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368, 1509–18 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Davila ML et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6, 224ra25 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lee DW et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Maude SL et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371, 1507–17 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Porter DL, Kalos M, Zheng Z, Levine B & June C Chimeric Antigen Receptor Therapy for B-cell Malignancies. J Cancer 2, 331–2 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Porter DL, Levine BL, Kalos M, Bagg A & June CH Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365, 725–33 (2011).First case-report of complete remission in two of 3 subjects with CCL following CD19 CAR T cell therapy
- 14.Kalos M et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3, 95ra73 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kochenderfer JN et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709–20 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Maher J, Brentjens RJ, Gunset G, Riviere I & Sadelain M Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol 20, 70–5 (2002).First report of sustained T cell expansion and functionality upon repeated exposure to antigen using a second generation CAR (28ζ) transduced in human primary T cells
- 17.Imai C et al. Chimeric receptors with 4–1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18, 676–84 (2004).First report of anti-tumour efficacy of a BBζ CAR in an ALL xenograft model
- 18.Zinkernagel RM & Doherty PC Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature 248, 701–2 (1974). [DOI] [PubMed] [Google Scholar]
- 19.Dembic Z et al. Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature 320, 232–8 (1986). [DOI] [PubMed] [Google Scholar]
- 20.Boomer JS & Green JM An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect Biol 2, a002436 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jenkins MK, Chen CA, Jung G, Mueller DL & Schwartz RH Inhibition of antigen-specific proliferation of type 1 murine T cell clones after stimulation with immobilized anti-CD3 monoclonal antibody. J Immunol 144, 16–22 (1990). [PubMed] [Google Scholar]
- 22.Weissman AM et al. Molecular cloning of the zeta chain of the T cell antigen receptor. Science 239, 1018–21 (1988). [DOI] [PubMed] [Google Scholar]
- 23.Irving BA & Weiss A The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways. Cell 64, 891–901 (1991). [DOI] [PubMed] [Google Scholar]
- 24.Romeo C & Seed B Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides. Cell 64, 1037–46 (1991). [DOI] [PubMed] [Google Scholar]
- 25.Letourneur F & Klausner RD T-cell and basophil activation through the cytoplasmic tail of T-cell-receptor zeta family proteins. Proc Natl Acad Sci U S A 88, 8905–9 (1991). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Eshhar Z, Waks T, Gross G & Schindler DG Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A 90, 720–4 (1993).Redirection of T cell hybridomas through the coupling of an scFv to T cell activating CD3ζ or Fc-γR domain.
- 27.Brocker T, Peter A, Traunecker A & Karjalainen K New simplified molecular design for functional T cell receptor. Eur J Immunol 23, 1435–9 (1993). [DOI] [PubMed] [Google Scholar]
- 28.Brocker T & Karjalainen K Signals through T cell receptor-zeta chain alone are insufficient to prime resting T lymphocytes. J Exp Med 181, 1653–9 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Brocker T Chimeric Fv-zeta or Fv-epsilon receptors are not sufficient to induce activation or cytokine production in peripheral T cells. Blood 96, 1999–2001 (2000). [PubMed] [Google Scholar]
- 30.Gallardo HF, Tan C, Ory D & Sadelain M Recombinant retroviruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate both stable gene transfer and pseudotransduction in human peripheral blood lymphocytes. Blood 90, 952–7 (1997). [PubMed] [Google Scholar]
- 31.Gong MC et al. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia 1, 123–7 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Krause A et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med 188, 619–26 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Finney HM, Lawson AD, Bebbington CR & Weir AN Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol 161, 2791–7 (1998). [PubMed] [Google Scholar]
- 34.Hombach A et al. Tumor-specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL-2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol 167, 6123–31 (2001). [DOI] [PubMed] [Google Scholar]
- 35.Shahinian A et al. Differential T cell costimulatory requirements in CD28-deficient mice. Science 261, 609–12 (1993). [DOI] [PubMed] [Google Scholar]
- 36.Chen L & Flies DB Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 13, 227–42 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hutloff A et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397, 263–6 (1999). [DOI] [PubMed] [Google Scholar]
- 38.Watanabe N et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat Immunol 4, 670–9 (2003). [DOI] [PubMed] [Google Scholar]
- 39.Brunet JF et al. A new member of the immunoglobulin superfamily--CTLA-4. Nature 328, 267–70 (1987). [DOI] [PubMed] [Google Scholar]
- 40.Ishida Y, Agata Y, Shibahara K & Honjo T Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 11, 3887–95 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hansen JA, Martin PJ & Nowinski RC Monoclonal-Antibodies Identifying a Novel T-Cell Antigen and Ia Antigens of Human-Lymphocytes. Immunogenetics 10, 247–260 (1980). [Google Scholar]
- 42.Aruffo A & Seed B Molecular cloning of a CD28 cDNA by a high-efficiency COS cell expression system. Proc Natl Acad Sci U S A 84, 8573–7 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yamada H et al. Monoclonal antibody 9.3 and anti-CD11 antibodies define reciprocal subsets of lymphocytes. Eur J Immunol 15, 1164–8 (1985). [DOI] [PubMed] [Google Scholar]
- 44.Gross JA, St John T & Allison JP The murine homologue of the T lymphocyte antigen CD28. Molecular cloning and cell surface expression. J Immunol 144, 3201–10 (1990). [PubMed] [Google Scholar]
- 45.Bour-Jordan H & Bluestone JA Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev 229, 41–66 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Woerly G et al. Human eosinophils express and release IL-13 following CD28-dependent activation. J Leukoc Biol 72, 769–79 (2002). [PubMed] [Google Scholar]
- 47.Kozbor D, Moretta A, Messner HA, Moretta L & Croce CM Tp44 molecules involved in antigen-independent T cell activation are expressed on human plasma cells. J Immunol 138, 4128–32 (1987). [PubMed] [Google Scholar]
- 48.Venuprasad K et al. Immunobiology of CD28 expression on human neutrophils. I. CD28 regulates neutrophil migration by modulating CXCR-1 expression. Eur J Immunol 31, 1536–43 (2001). [DOI] [PubMed] [Google Scholar]
- 49.Williams JA et al. Regulation of thymic NKT cell development by the B7-CD28 costimulatory pathway. J Immunol 181, 907–17 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Viola A & Lanzavecchia A T cell activation determined by T cell receptor number and tunable thresholds. Science 273, 104–6 (1996). [DOI] [PubMed] [Google Scholar]
- 51.Manickasingham SP, Anderton SM, Burkhart C & Wraith DC Qualitative and quantitative effects of CD28/B7-mediated costimulation on naive T cells in vitro. J Immunol 161, 3827–35 (1998). [PubMed] [Google Scholar]
- 52.Diehn M et al. Genomic expression programs and the integration of the CD28 costimulatory signal in T cell activation. Proc Natl Acad Sci U S A 99, 11796–801 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Riley JL et al. Modulation of TCR-induced transcriptional profiles by ligation of CD28, ICOS, and CTLA-4 receptors. Proc Natl Acad Sci U S A 99, 11790–5 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.June CH, Ledbetter JA, Gillespie MM, Lindsten T & Thompson CB T-cell proliferation involving the CD28 pathway is associated with cyclosporine-resistant interleukin 2 gene expression. Mol Cell Biol 7, 4472–81 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Smeets RL et al. Molecular pathway profiling of T lymphocyte signal transduction pathways; Th1 and Th2 genomic fingerprints are defined by TCR and CD28-mediated signaling. BMC Immunol 13, 12 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Boonen GJ et al. CD28 induces cell cycle progression by IL-2-independent down-regulation of p27kip1 expression in human peripheral T lymphocytes. Eur J Immunol 29, 789–98 (1999). [DOI] [PubMed] [Google Scholar]
- 57.Radvanyi LG et al. CD28 costimulation inhibits TCR-induced apoptosis during a primary T cell response. J Immunol 156, 1788–98 (1996). [PubMed] [Google Scholar]
- 58.Weng NP, Araki Y & Subedi K The molecular basis of the memory T cell response: differential gene expression and its epigenetic regulation. Nat Rev Immunol 12, 306–15 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gubser PM et al. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol 14, 1064–72 (2013). [DOI] [PubMed] [Google Scholar]
- 60.Yang K et al. T cell exit from quiescence and differentiation into Th2 cells depend on Raptor-mTORC1-mediated metabolic reprogramming. Immunity 39, 1043–56 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Faris M, Kokot N, Lee L & Nel AE Regulation of interleukin-2 transcription by inducible stable expression of dominant negative and dominant active mitogen-activated protein kinase kinase kinase in jurkat T cells. Evidence for the importance of Ras in a pathway that is controlled by dual receptor stimulation. J Biol Chem 271, 27366–73 (1996). [DOI] [PubMed] [Google Scholar]
- 62.Rao S, Gerondakis S, Woltring D & Shannon MF c-Rel is required for chromatin remodeling across the IL-2 gene promoter. J Immunol 170, 3724–31 (2003). [DOI] [PubMed] [Google Scholar]
- 63.Thomas RM, Gao L & Wells AD Signals from CD28 induce stable epigenetic modification of the IL-2 promoter. J Immunol 174, 4639–46 (2005). [DOI] [PubMed] [Google Scholar]
- 64.Murayama A et al. A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory. EMBO J 25, 1081–92 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Grogan JL et al. Early transcription and silencing of cytokine genes underlie polarization of T helper cell subsets. Immunity 14, 205–15 (2001). [DOI] [PubMed] [Google Scholar]
- 66.Acuto O & Michel F CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol 3, 939–51 (2003). [DOI] [PubMed] [Google Scholar]
- 67.Rathmell JC, Farkash EA, Gao W & Thompson CB IL-7 enhances the survival and maintains the size of naive T cells. J Immunol 167, 6869–76 (2001). [DOI] [PubMed] [Google Scholar]
- 68.Vander Heiden MG et al. Growth factors can influence cell growth and survival through effects on glucose metabolism. Mol Cell Biol 21, 5899–912 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA & Thompson CB In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol Cell 6, 683–92 (2000). [DOI] [PubMed] [Google Scholar]
- 70.Jacobs SR et al. Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J Immunol 180, 4476–86 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Frauwirth KA et al. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769–77 (2002). [DOI] [PubMed] [Google Scholar]
- 72.Kane LP, Andres PG, Howland KC, Abbas AK & Weiss A Akt provides the CD28 costimulatory signal for up-regulation of IL-2 and IFN-gamma but not TH2 cytokines. Nat Immunol 2, 37–44 (2001). [DOI] [PubMed] [Google Scholar]
- 73.Boise LH et al. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity 3, 87–98 (1995). [DOI] [PubMed] [Google Scholar]
- 74.Wan YY & DeGregori J The survival of antigen-stimulated T cells requires NFkappaB-mediated inhibition of p73 expression. Immunity 18, 331–42 (2003). [DOI] [PubMed] [Google Scholar]
- 75.Kirchhoff S, Muller WW, Li-Weber M & Krammer PH Up-regulation of c-FLIPshort and reduction of activation-induced cell death in CD28-costimulated human T cells. Eur J Immunol 30, 2765–74 (2000). [DOI] [PubMed] [Google Scholar]
- 76.Gimmi CD, Freeman GJ, Gribben JG, Gray G & Nadler LM Human T-cell clonal anergy is induced by antigen presentation in the absence of B7 costimulation. Proc Natl Acad Sci U S A 90, 6586–90 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Beier KC et al. Induction, binding specificity and function of human ICOS. Eur J Immunol 30, 3707–17 (2000). [DOI] [PubMed] [Google Scholar]
- 78.Johnson-Leger C, Christensen J & Klaus GG CD28 co-stimulation stabilizes the expression of the CD40 ligand on T cells. Int Immunol 10, 1083–91 (1998). [DOI] [PubMed] [Google Scholar]
- 79.Kim JO, Kim HW, Baek KM & Kang CY NF-kappaB and AP-1 regulate activation-dependent CD137 (4–1BB) expression in T cells. FEBS Lett 541, 163–70 (2003). [DOI] [PubMed] [Google Scholar]
- 80.Borowski AB et al. Memory CD8+ T cells require CD28 costimulation. J Immunol 179, 6494–503 (2007). [DOI] [PubMed] [Google Scholar]
- 81.Fuse S, Zhang W & Usherwood EJ Control of memory CD8+ T cell differentiation by CD80/CD86-CD28 costimulation and restoration by IL-2 during the recall response. J Immunol 180, 1148–57 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hathcock KS, Laszlo G, Pucillo C, Linsley P & Hodes RJ Comparative analysis of B7–1 and B7–2 costimulatory ligands: expression and function. J Exp Med 180, 631–40 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fleischer J et al. Differential expression and function of CD80 (B7–1) and CD86 (B7–2) on human peripheral blood monocytes. Immunology 89, 592–8 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Eck SC, Chang D, Wells AD & Turka LA Differential down-regulation of CD28 by B7–1 and B7–2 engagement. Transplantation 64, 1497–9 (1997). [DOI] [PubMed] [Google Scholar]
- 85.Lewis DE, Merched-Sauvage M, Goronzy JJ, Weyand CM & Vallejo AN Tumor necrosis factor-alpha and CD80 modulate CD28 expression through a similar mechanism of T-cell receptor-independent inhibition of transcription. J Biol Chem 279, 29130–8 (2004). [DOI] [PubMed] [Google Scholar]
- 86.Berg M & Zavazava N Regulation of CD28 expression on CD8+ T cells by CTLA-4. J Leukoc Biol 83, 853–63 (2008). [DOI] [PubMed] [Google Scholar]
- 87.Habib-Agahi M, Jaberipour M & Searle PF 4–1BBL costimulation retrieves CD28 expression in activated T cells. Cell Immunol 256, 39–46 (2009). [DOI] [PubMed] [Google Scholar]
- 88.Topp MS et al. Restoration of CD28 expression in CD28− CD8+ memory effector T cells reconstitutes antigen-induced IL-2 production. J Exp Med 198, 947–55 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Peach RJ et al. Complementarity determining region 1 (CDR1)- and CDR3-analogous regions in CTLA-4 and CD28 determine the binding to B7–1. J Exp Med 180, 2049–58 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Linsley PS et al. Human B7–1 (CD80) and B7–2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1, 793–801 (1994). [DOI] [PubMed] [Google Scholar]
- 91.Qureshi OS et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 332, 600–3 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Chemnitz JM, Parry RV, Nichols KE, June CH & Riley JL SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 173, 945–54 (2004). [DOI] [PubMed] [Google Scholar]
- 93.Barber DL et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–7 (2006). [DOI] [PubMed] [Google Scholar]
- 94.Riley JL PD-1 signaling in primary T cells. Immunol Rev 229, 114–25 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Croft M Costimulation of T cells by OX40, 4–1BB, and CD27. Cytokine Growth Factor Rev 14, 265–73 (2003). [DOI] [PubMed] [Google Scholar]
- 96.Kwon BS et al. Isolation and initial characterization of multiple species of T-lymphocyte subset cDNA clones. Proc Natl Acad Sci U S A 84, 2896–900 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kwon BS & Weissman SM cDNA sequences of two inducible T-cell genes. Proc Natl Acad Sci U S A 86, 1963–7 (1989). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schwarz H, Tuckwell J & Lotz M A receptor induced by lymphocyte activation (ILA): a new member of the human nerve-growth-factor/tumor-necrosis-factor receptor family. Gene 134, 295–8 (1993). [DOI] [PubMed] [Google Scholar]
- 99.Schwarz H, Valbracht J, Tuckwell J, von Kempis J & Lotz M ILA, the human 4–1BB homologue, is inducible in lymphoid and other cell lineages. Blood 85, 1043–52 (1995). [PubMed] [Google Scholar]
- 100.Lee SJ et al. 4–1BB signal stimulates the activation, expansion, and effector functions of gammadelta T cells in mice and humans. Eur J Immunol 43, 1839–48 (2013). [DOI] [PubMed] [Google Scholar]
- 101.Melero I, Johnston JV, Shufford WW, Mittler RS & Chen L NK1.1 cells express 4–1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4–1BB monoclonal antibodies. Cell Immunol 190, 167–72 (1998). [DOI] [PubMed] [Google Scholar]
- 102.Zhang X et al. CD137 promotes proliferation and survival of human B cells. J Immunol 184, 787–95 (2010). [DOI] [PubMed] [Google Scholar]
- 103.Wilcox RA et al. Cutting edge: Expression of functional CD137 receptor by dendritic cells. J Immunol 168, 4262–7 (2002). [DOI] [PubMed] [Google Scholar]
- 104.Drenkard D et al. CD137 is expressed on blood vessel walls at sites of inflammation and enhances monocyte migratory activity. FASEB J 21, 456–63 (2007). [DOI] [PubMed] [Google Scholar]
- 105.von Kempis J, Schwarz H & Lotz M Differentiation-dependent and stimulus-specific expression of ILA, the human 4–1BB-homologue, in cells of mesenchymal origin. Osteoarthritis Cartilage 5, 394–406 (1997). [DOI] [PubMed] [Google Scholar]
- 106.Futagawa T et al. Expression and function of 4–1BB and 4–1BB ligand on murine dendritic cells. Int Immunol 14, 275–86 (2002). [DOI] [PubMed] [Google Scholar]
- 107.Goodwin RG et al. Molecular cloning of a ligand for the inducible T cell gene 4–1BB: a member of an emerging family of cytokines with homology to tumor necrosis factor. Eur J Immunol 23, 2631–41 (1993). [DOI] [PubMed] [Google Scholar]
- 108.Pollok KE et al. 4–1BB T-cell antigen binds to mature B cells and macrophages, and costimulates anti-mu-primed splenic B cells. Eur J Immunol 24, 367–74 (1994). [DOI] [PubMed] [Google Scholar]
- 109.Alderson MR et al. Molecular and biological characterization of human 4–1BB and its ligand. Eur J Immunol 24, 2219–27 (1994). [DOI] [PubMed] [Google Scholar]
- 110.Polte T, Jagemann A, Foell J, Mittler RS & Hansen G CD137 ligand prevents the development of T-helper type 2 cell-mediated allergic asthma by interferon-gamma-producing CD8+ T cells. Clin Exp Allergy 37, 1374–85 (2007). [DOI] [PubMed] [Google Scholar]
- 111.Won EY et al. The structure of the trimer of human 4–1BB ligand is unique among members of the tumor necrosis factor superfamily. J Biol Chem 285, 9202–10 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Vinay DS, Cha K & Kwon BS Dual immunoregulatory pathways of 4–1BB signaling. J Mol Med (Berl) 84, 726–36 (2006). [DOI] [PubMed] [Google Scholar]
- 113.Dawicki W & Watts TH Expression and function of 4–1BB during CD4 versus CD8 T cell responses in vivo. Eur J Immunol 34, 743–51 (2004). [DOI] [PubMed] [Google Scholar]
- 114.Takahashi C, Mittler RS & Vella AT Cutting edge: 4–1BB is a bona fide CD8 T cell survival signal. J Immunol 162, 5037–40 (1999). [PubMed] [Google Scholar]
- 115.Lee SW et al. Functional dichotomy between OX40 and 4–1BB in modulating effector CD8 T cell responses. J Immunol 177, 4464–72 (2006). [DOI] [PubMed] [Google Scholar]
- 116.Pulle G, Vidric M & Watts TH IL-15-dependent induction of 4–1BB promotes antigen-independent CD8 memory T cell survival. J Immunol 176, 2739–48 (2006). [DOI] [PubMed] [Google Scholar]
- 117.Nam KO et al. Cross-linking of 4–1BB activates TCR-signaling pathways in CD8+ T lymphocytes. J Immunol 174, 1898–905 (2005). [DOI] [PubMed] [Google Scholar]
- 118.Daniel-Meshulam I, Horovitz-Fried M & Cohen CJ Enhanced antitumor activity mediated by human 4–1BB-engineered T cells. Int J Cancer 133, 2903–13 (2013). [DOI] [PubMed] [Google Scholar]
- 119.Cannons JL et al. 4–1BB ligand induces cell division, sustains survival, and enhances effector function of CD4 and CD8 T cells with similar efficacy. J Immunol 167, 1313–24 (2001). [DOI] [PubMed] [Google Scholar]
- 120.Wen T, Bukczynski J & Watts TH 4–1BB ligand-mediated costimulation of human T cells induces CD4 and CD8 T cell expansion, cytokine production, and the development of cytolytic effector function. J Immunol 168, 4897–906 (2002). [DOI] [PubMed] [Google Scholar]
- 121.Jang IK, Lee ZH, Kim YJ, Kim SH & Kwon BS Human 4–1BB (CD137) signals are mediated by TRAF2 and activate nuclear factor-kappa B. Biochem Biophys Res Commun 242, 613–20 (1998). [DOI] [PubMed] [Google Scholar]
- 122.Arch RH & Thompson CB 4–1BB and Ox40 are members of a tumor necrosis factor (TNF)-nerve growth factor receptor subfamily that bind TNF receptor-associated factors and activate nuclear factor kappaB. Mol Cell Biol 18, 558–65 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Saoulli K et al. CD28-independent, TRAF2-dependent costimulation of resting T cells by 4–1BB ligand. J Exp Med 187, 1849–62 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zheng C, Kabaleeswaran V, Wang Y, Cheng G & Wu H Crystal structures of the TRAF2: cIAP2 and the TRAF1: TRAF2: cIAP2 complexes: affinity, specificity, and regulation. Mol Cell 38, 101–13 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sabbagh L, Pulle G, Liu Y, Tsitsikov EN & Watts TH ERK-dependent Bim modulation downstream of the 4–1BB-TRAF1 signaling axis is a critical mediator of CD8 T cell survival in vivo. J Immunol 180, 8093–101 (2008). [DOI] [PubMed] [Google Scholar]
- 126.McPherson AJ, Snell LM, Mak TW & Watts TH Opposing roles for TRAF1 in the alternative versus classical NF-kappaB pathway in T cells. J Biol Chem 287, 23010–9 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Pollok KE et al. Inducible T cell antigen 4–1BB. Analysis of expression and function. J Immunol 150, 771–81 (1993). [PubMed] [Google Scholar]
- 128.Cannons JL, Choi Y & Watts TH Role of TNF receptor-associated factor 2 and p38 mitogen-activated protein kinase activation during 4–1BB-dependent immune response. J Immunol 165, 6193–204 (2000). [DOI] [PubMed] [Google Scholar]
- 129.Hauer J et al. TNF receptor (TNFR)-associated factor (TRAF) 3 serves as an inhibitor of TRAF2/5-mediated activation of the noncanonical NF-kappaB pathway by TRAF-binding TNFRs. Proc Natl Acad Sci U S A 102, 2874–9 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Lee HW, Nam KO, Park SJ & Kwon BS 4–1BB enhances CD8+ T cell expansion by regulating cell cycle progression through changes in expression of cyclins D and E and cyclin-dependent kinase inhibitor p27kip1. Eur J Immunol 33, 2133–41 (2003). [DOI] [PubMed] [Google Scholar]
- 131.DeBenedette MA, Shahinian A, Mak TW & Watts TH Costimulation of CD28- T lymphocytes by 4–1BB ligand. J Immunol 158, 551–9 (1997). [PubMed] [Google Scholar]
- 132.Chu NR, DeBenedette MA, Stiernholm BJ, Barber BH & Watts TH Role of IL-12 and 4–1BB ligand in cytokine production by CD28+ and CD28- T cells. J Immunol 158, 3081–9 (1997). [PubMed] [Google Scholar]
- 133.Bukczynski J, Wen T & Watts TH Costimulation of human CD28- T cells by 4–1BB ligand. Eur J Immunol 33, 446–54 (2003). [DOI] [PubMed] [Google Scholar]
- 134.Fann M et al. Gene expression characteristics of CD28null memory phenotype CD8+ T cells and its implication in T-cell aging. Immunol Rev 205, 190–206 (2005). [DOI] [PubMed] [Google Scholar]
- 135.Ahmad HT, Mansooreh J, Fereshteh M & Mojtaba H Changes in lymphocytes’ telomerase activity by 4–1BB costimulation. J Cancer Res Ther 10, 998–1003 (2014). [DOI] [PubMed] [Google Scholar]
- 136.Hendriks J et al. During viral infection of the respiratory tract, CD27, 4–1BB, and OX40 collectively determine formation of CD8+ memory T cells and their capacity for secondary expansion. J Immunol 175, 1665–76 (2005). [DOI] [PubMed] [Google Scholar]
- 137.Shuford WW et al. 4–1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med 186, 47–55 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sabbagh L, Snell LM & Watts TH TNF family ligands define niches for T cell memory. Trends Immunol 28, 333–9 (2007). [DOI] [PubMed] [Google Scholar]
- 139.DeBenedette MA et al. Analysis of 4–1BB ligand (4–1BBL)-deficient mice and of mice lacking both 4–1BBL and CD28 reveals a role for 4–1BBL in skin allograft rejection and in the cytotoxic T cell response to influenza virus. J Immunol 163, 4833–41 (1999). [PubMed] [Google Scholar]
- 140.Kwon BS et al. Immune responses in 4–1BB (CD137)-deficient mice. J Immunol 168, 5483–90 (2002). [DOI] [PubMed] [Google Scholar]
- 141.Watts TH TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol 23, 23–68 (2005). [DOI] [PubMed] [Google Scholar]
- 142.Humphreys IR et al. Biphasic role of 4–1BB in the regulation of mouse cytomegalovirus-specific CD8(+) T cells. Eur J Immunol 40, 2762–8 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wang C et al. Loss of the signaling adaptor TRAF1 causes CD8+ T cell dysregulation during human and murine chronic infection. J Exp Med 209, 77–91 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Habib-Agahi M, Phan TT & Searle PF Co-stimulation with 4–1BB ligand allows extended T-cell proliferation, synergizes with CD80/CD86 and can reactivate anergic T cells. Int Immunol 19, 1383–94 (2007). [DOI] [PubMed] [Google Scholar]
- 145.Zhang B, Zhang Y, Niu L, Vella AT & Mittler RS Dendritic cells and Stat3 are essential for CD137-induced CD8 T cell activation-induced cell death. J Immunol 184, 4770–8 (2010). [DOI] [PubMed] [Google Scholar]
- 146.Frecha C et al. Stable transduction of quiescent T cells without induction of cycle progression by a novel lentiviral vector pseudotyped with measles virus glycoproteins. Blood 112, 4843–52 (2008). [DOI] [PubMed] [Google Scholar]
- 147.Unutmaz D, KewalRamani VN, Marmon S & Littman DR Cytokine signals are sufficient for HIV-1 infection of resting human T lymphocytes. J Exp Med 189, 1735–46 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Brentjens RJ et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res 13, 5426–35 (2007). [DOI] [PubMed] [Google Scholar]
- 149.Carpenito C et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A 106, 3360–5 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Milone MC et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 17, 1453–64 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Tammana S et al. 4–1BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood T cells against B-cell malignancies. Hum Gene Ther 21, 75–86 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Zhong XS, Matsushita M, Plotkin J, Riviere I & Sadelain M Chimeric antigen receptors combining 4–1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther 18, 413–20 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Song DG et al. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 119, 696–706 (2012). [DOI] [PubMed] [Google Scholar]
- 154.Santoro SP et al. T Cells Bearing a Chimeric Antigen Receptor against Prostate-Specific Membrane Antigen Mediate Vascular Disruption and Result in Tumor Regression. Cancer Immunol Res (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Finney HM, Akbar AN & Lawson AD Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J Immunol 172, 104–13 (2004). [DOI] [PubMed] [Google Scholar]
- 156.Guedan S et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 124, 1070–80 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Hombach AA & Abken H Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int J Cancer 129, 2935–44 (2011). [DOI] [PubMed] [Google Scholar]
- 158.Rosenberg SA IL-2: the first effective immunotherapy for human cancer. J Immunol 192, 5451–8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Loskog A et al. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia 20, 1819–28 (2006). [DOI] [PubMed] [Google Scholar]
- 160.Kofler DM et al. CD28 costimulation Impairs the efficacy of a redirected t-cell antitumor attack in the presence of regulatory t cells which can be overcome by preventing Lck activation. Mol Ther 19, 760–7 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Pegram HJ et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 119, 4133–41 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Sun J et al. T cells expressing constitutively active Akt resist multiple tumor-associated inhibitory mechanisms. Mol Ther 18, 2006–17 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Frigault MJ et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol Res (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gargett T, Fraser CK, Dotti G, Yvon ES & Brown MP BRAF and MEK inhibition variably affect GD2-specific chimeric antigen receptor (CAR) T-cell function in vitro. J Immunother 38, 12–23 (2015). [DOI] [PubMed] [Google Scholar]
- 165.Song DG et al. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4–1BB). Cancer Res 71, 4617–27 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Chattopadhyay K et al. Sequence, structure, function, immunity: structural genomics of costimulation. Immunol Rev 229, 356–86 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Brentjens RJ et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 9, 279–86 (2003).First report demonstrating the therapeutic efficacy of human CD19 CAR T cells in mice with systemic B cell malignancies.
- 168.Davila ML, Kloss CC, Gunset G & Sadelain M CD19 CAR-targeted T cells induce long-term remission and B Cell Aplasia in an immunocompetent mouse model of B cell acute lymphoblastic leukemia. PLoS One 8, e61338 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Adusumilli PS et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med 6, 261ra151 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Zhang B, Karrison T, Rowley DA & Schreiber H IFN-gamma- and TNF-dependent bystander eradication of antigen-loss variants in established mouse cancers. J Clin Invest 118, 1398–404 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Braumuller H et al. T-helper-1-cell cytokines drive cancer into senescence. Nature 494, 361–5 (2013). [DOI] [PubMed] [Google Scholar]
- 172.Matsushita H et al. Cytotoxic T Lymphocytes Block Tumor Growth Both by Lytic Activity and IFNgamma-Dependent Cell-Cycle Arrest. Cancer Immunol Res 3, 26–36 (2015). [DOI] [PubMed] [Google Scholar]
- 173.Kowolik CM et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res 66, 10995–1004 (2006). [DOI] [PubMed] [Google Scholar]
- 174.Plunkett FJ et al. The loss of telomerase activity in highly differentiated CD8+CD28-CD27- T cells is associated with decreased Akt (Ser473) phosphorylation. J Immunol 178, 7710–9 (2007). [DOI] [PubMed] [Google Scholar]
- 175.John LB et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res 19, 5636–46 (2013). [DOI] [PubMed] [Google Scholar]
- 176.Hoyos V et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24, 1160–70 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Gros A et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J Clin Invest 124, 2246–59 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Liadi I et al. Individual Motile CD4+ T Cells Can Participate in Efficient Multikilling through Conjugation to Multiple Tumor Cells. Cancer Immunol Res (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Crompton JG, Sukumar M & Restifo NP Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy. Immunol Rev 257, 264–76 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Riddell SR et al. Adoptive therapy with chimeric antigen receptor-modified T cells of defined subset composition. Cancer J 20, 141–4 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Li YS, Hayakawa K & Hardy RR The regulated expression of B lineage associated genes during B cell differentiation in bone marrow and fetal liver. J Exp Med 178, 951–60 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Li YS, Wasserman R, Hayakawa K & Hardy RR Identification of the earliest B lineage stage in mouse bone marrow. Immunity 5, 527–35 (1996). [DOI] [PubMed] [Google Scholar]
- 183.Cooper LJ et al. T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect. Blood 101, 1637–44 (2003). [DOI] [PubMed] [Google Scholar]
- 184.Kohn DB et al. CARs on track in the clinic. Mol Ther 19, 432–8 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Jensen MC et al. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant 16, 1245–56 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kochenderfer JN et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099–102 (2010).First case-report of a remission following CD19 CAR T cell therapy in a subject with a B cell lymphoma
- 187.Savoldo B et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 121, 1822–6 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Cruz CR et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122, 2965–73 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kochenderfer JN et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122, 4129–39 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Hollyman D et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother 32, 169–80 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Lee DW et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124, 188–95 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Davila ML, Brentjens R, Wang X, Riviere I & Sadelain M How do CARs work?: Early insights from recent clinical studies targeting CD19. Oncoimmunology 1, 1577–1583 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Paulos CM et al. The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells. Sci Transl Med 2, 55ra78 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Maciolek JA, Pasternak JA & Wilson HL Metabolism of activated T lymphocytes. Curr Opin Immunol 27, 60–74 (2014). [DOI] [PubMed] [Google Scholar]
- 195.Oestreich KJ et al. Bcl-6 directly represses the gene program of the glycolysis pathway. Nat Immunol 15, 957–64 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Dong C et al. ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature 409, 97–101 (2001). [DOI] [PubMed] [Google Scholar]
- 197.Tafuri A et al. ICOS is essential for effective T-helper-cell responses. Nature 409, 105–9 (2001). [DOI] [PubMed] [Google Scholar]
- 198.Burmeister Y et al. ICOS controls the pool size of effector-memory and regulatory T cells. J Immunol 180, 774–82 (2008). [DOI] [PubMed] [Google Scholar]
- 199.Shen CJ et al. Chimeric antigen receptor containing ICOS signaling domain mediates specific and efficient antitumor effect of T cells against EGFRvIII expressing glioma. J Hematol Oncol 6, 33 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Kopf M et al. OX40-deficient mice are defective in Th cell proliferation but are competent in generating B cell and CTL Responses after virus infection. Immunity 11, 699–708 (1999). [DOI] [PubMed] [Google Scholar]
- 201.Salek-Ardakani S, Moutaftsi M, Crotty S, Sette A & Croft M OX40 drives protective vaccinia virus-specific CD8 T cells. J Immunol 181, 7969–76 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nolte MA, van Olffen RW, van Gisbergen KP & van Lier RA Timing and tuning of CD27-CD70 interactions: the impact of signal strength in setting the balance between adaptive responses and immunopathology. Immunol Rev 229, 216–31 (2009). [DOI] [PubMed] [Google Scholar]
- 203.Croft M The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol 9, 271–85 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Hildebrand JM et al. Roles of tumor necrosis factor receptor associated factor 3 (TRAF3) and TRAF5 in immune cell functions. Immunol Rev 244, 55–74 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Pule MA et al. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther 12, 933–41 (2005). [DOI] [PubMed] [Google Scholar]
- 206.Wilkie S et al. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J Immunol 180, 4901–9 (2008). [DOI] [PubMed] [Google Scholar]
- 207.Carr JM et al. CD27 mediates interleukin-2-independent clonal expansion of the CD8+ T cell without effector differentiation. Proc Natl Acad Sci U S A 103, 19454–9 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Fox CJ, Hammerman PS & Thompson CB The Pim kinases control rapamycin-resistant T cell survival and activation. J Exp Med 201, 259–66 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Sentman CL & Meehan KR NKG2D CARs as cell therapy for cancer. Cancer J 20, 156–9 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Chang YH et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res 73, 1777–86 (2013). [DOI] [PubMed] [Google Scholar]
- 211.Lehner M et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS One 7, e31210 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Song DG et al. Chimeric NKG2D CAR-expressing T cell-mediated attack of human ovarian cancer is enhanced by histone deacetylase inhibition. Hum Gene Ther 24, 295–305 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Barber A & Sentman CL Chimeric NKG2D T cells require both T cell- and host-derived cytokine secretion and perforin expression to increase tumor antigen presentation and systemic immunity. J Immunol 183, 2365–72 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Zhang T & Sentman CL Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J Immunol 190, 2455–63 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Zhao Y et al. A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol 183, 5563–74 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Wang J et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther 18, 712–25 (2007). [DOI] [PubMed] [Google Scholar]
- 217.Till BG et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4–1BB domains: pilot clinical trial results. Blood 119, 3940–50 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Morgan RA et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 18, 843–51 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Kloss CC, Condomines M, Cartellieri M, Bachmann M & Sadelain M Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol 31, 71–5 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Wilkie S et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol 32, 1059–70 (2012). [DOI] [PubMed] [Google Scholar]
- 221.Lanitis E et al. Chimeric antigen receptor T Cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Res 1, 43–53 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Hudecek M et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res 3, 125–35 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.James SE et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol 180, 7028–38 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Guest RD et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother 28, 203–11 (2005). [DOI] [PubMed] [Google Scholar]
- 225.Almasbak H et al. Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther (2015). [DOI] [PubMed] [Google Scholar]
- 226.Fitzer-Attas CJ, Schindler DG, Waks T & Eshhar Z Harnessing Syk family tyrosine kinases as signaling domains for chimeric single chain of the variable domain receptors: optimal design for T cell activation. J Immunol 160, 145–54 (1998). [PubMed] [Google Scholar]
- 227.Bridgeman JS et al. The optimal antigen response of chimeric antigen receptors harboring the CD3zeta transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex. J Immunol 184, 6938–49 (2010). [DOI] [PubMed] [Google Scholar]
- 228.Miller JF & Sadelain M The Journey from Discoveries in Fundamental Immunology to Cancer Immunotherapy. Cancer Cell (2015). [DOI] [PubMed] [Google Scholar]
- 229.Suntharalingam G et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 355, 1018–28 (2006). [DOI] [PubMed] [Google Scholar]
- 230.Melero I, Hirschhorn-Cymerman D, Morales-Kastresana A, Sanmamed MF & Wolchok JD Agonist antibodies to TNFR molecules that costimulate T and NK cells. Clin Cancer Res 19, 1044–53 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Yao S, Zhu Y & Chen L Advances in targeting cell surface signalling molecules for immune modulation. Nat Rev Drug Discov 12, 130–46 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Gangadhar TC & Vonderheide RH Mitigating the toxic effects of anticancer immunotherapy. Nat Rev Clin Oncol 11, 91–9 (2014). [DOI] [PubMed] [Google Scholar]
- 233.Curti BD et al. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res 73, 7189–98 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.de Vos S et al. A phase II study of dacetuzumab (SGN-40) in patients with relapsed diffuse large B-cell lymphoma (DLBCL) and correlative analyses of patient-specific factors. J Hematol Oncol 7, 44 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Hussein M et al. A phase I multidose study of dacetuzumab (SGN-40; humanized anti-CD40 monoclonal antibody) in patients with multiple myeloma. Haematologica 95, 845–8 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Furman RR, Forero-Torres A, Shustov A & Drachman JG A phase I study of dacetuzumab (SGN-40, a humanized anti-CD40 monoclonal antibody) in patients with chronic lymphocytic leukemia. Leuk Lymphoma 51, 228–35 (2010). [DOI] [PubMed] [Google Scholar]
- 237.Advani R et al. Phase I study of the humanized anti-CD40 monoclonal antibody dacetuzumab in refractory or recurrent non-Hodgkin’s lymphoma. J Clin Oncol 27, 4371–7 (2009). [DOI] [PubMed] [Google Scholar]
- 238.Topalian SL et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32, 1020–30 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Hodi FS et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363, 711–23 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Brahmer JR et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366, 2455–65 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Hodi FS et al. Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: a randomized clinical trial. JAMA 312, 1744–53 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Topalian SL et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366, 2443–54 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]


