Skip to main content
Immunotherapy Advances logoLink to Immunotherapy Advances
. 2024 Jun 19;4(1):ltae004. doi: 10.1093/immadv/ltae004

Regulation of temporal cytokine production by co-stimulation receptors in TCR-T cells is lost in CAR-T cells

Ashna Patel 1, Mikhail A Kutuzov 2, Michael L Dustin 3, P Anton van der Merwe 4, Omer Dushek 5,
PMCID: PMC11228853  PMID: 38978751

Abstract

CD8+ T cells contribute to immune responses by producing cytokines when their T-cell receptors (TCRs) recognise peptide antigens on major-histocompability-complex class I. However, excessive cytokine production can be harmful. For example, cytokine release syndrome is a common toxicity observed in treatments that activate T cells, including chimeric antigen receptor (CAR)-T-cell therapy. While the engagement of costimulatory receptors is well known to enhance cytokine production, we have limited knowledge of their ability to regulate the kinetics of cytokine production by CAR-T cells. Here we compare early (0–12 h) and late (12–20 h) production of IFN-gg, IL-2, and TNF-a production by T cells stimulated via TCR or CARs in the presence or absence ligands for CD2, LFA-1, CD28, CD27, and 4-1BB. For T cells expressing TCRs and 1st-generation CARs, activation by antigen alone was sufficient to stimulate early cytokine production, while co-stimulation by CD2 and 4-1BB was required to maintain late cytokine production. In contrast, T cells expressing 2nd-generation CARs, which have intrinsic costimulatory signalling motifs, produce high levels of cytokines in both early and late periods in the absence of costimulatory receptor ligands. Losing the requirement for costimulation for sustained cytokine production may contribute to the effectiveness and/or toxicity of 2nd-generation CAR-T-cell therapy.

Keywords: T cells, chimeric antigen receptors, T-cell receptors, co-stimulation receptors, cytokine production, immmunotherapy

Graphical Abstract

Graphical Abstract.

Graphical Abstract

Introduction

CD8+ T cells play central roles in immunity to intracellular pathogens and cancer. They can become activated to respond when their cell surface T-cell receptors (TCRs) recognise peptide antigens displayed on major-histocompatibility-complex (pMHC) class I gene products on the surface of nearly all cells. The binding of pMHC to the TCR initiates a signalling cascade that can lead to multiple functional responses. These include the production of cytokines such as IFN-g, IL-2, and TNF-a that collectively contribute to the ability of CD8+ T cells to execute immune responses [1, 2].

Because excessive cytokines levels can damage tissues, their production needs to be tightly regulated. One mechanism of regulation is engagement of co-stimulatory and/or co-inhibitory receptors on the T-cell surface, which increases and decreases the production of cytokines, respectively [3–6]. For example, cytokine production by T cells in cancer and chronic infections requires engagement of CD28 [7]. We have limited information on the relative abilities of other co-stimulatory receptors, such as the adhesion receptors CD2 and LFA-1 and the TNFSF receptors CD27 and 4-1BB, to sustain T-cell cytokine production.

The use of T cells in cell therapies to treat cancers, autoimmunity, and intractable chronic infections is showing great promise [8–10]. In these therapies, T cells are transduced to express a new TCR (TCR-T) or chimeric antigen receptor (CAR-T) that recognises an appropriate target antigen before being infused back into patients. While TCR-T and 1st-generation CAR-T lack intrinsic co-stimulatory activity, 2nd-generation CAR-T contains co-stimulatory signalling motifs in their cytoplasmic domains. The role of costimulatory receptors and intrinsic co-stimulatory signalling motifs in CAR-T-cell activation has previously been investigated [11–17] but their role in regulating sustained cytokine production by TCR-T and CAR-T cells remains poorly characterised. In the case of CAR-T cells, this is particularly important because patients often experience cytokine release syndrome, which can lead to organ failure and death [18].

We have previously developed and validated a reductionist system using TCR/CAR and co-stimulatory receptor ligands, and used it to investigate antigen sensitivity of TCRs and CARs [15, 17, 19–22] (Fig. 1A). In this work, we use this system to investigate production of the cytokines IFN-g, IL-2, and TNF-a (Fig. 1B). We found that, while cytokine production by TCR-T and 1st generation CAR-T cells was dependent on engagement of co-stimulation receptors, cytokine production by 2nd generation CAR-T cells was largely independent of co-stimulation receptors.

Figure 1.

Figure 1.

Regulated production of cytokine by co-stimulation receptors for the TCR. (A) Schematic of costimulation receptors and their ligands. (B) Schematics of potential cytokine production kinetics when T cells recognise antigen alone or with ligands to co-stimulation receptors. (C, D) Schematic of experiments to determine early, late, and total cytokine production. (E) Primary human CD8+ T cells transduced with the 1G4 TCR were stimulated by streptavidin surfaces coated with biotinylated pMHC alone (250 ng/well) or with the indicated biotinylated ligand (250 ng/well). The supernatant level of the indicated cytokine was determined after 20 h of stimulation (total production), after 12 h (early production), or between 12 and 20 h (late production). The rat CD4 ligand is used as a non-binding ligand control. Statistical significance of each ligand is compared to pMHC alone condition. (F) The production rate of each cytokine is calculated by dividing the supernatant cytokine data from (E) by the stimulation duration: 20, 12, or 8 h for total, early, or late, respectively. Statistical significance was determined by multiple t-tests on log-transformed data with Holm–Sidak correction for multiple comparisons. Abbreviations: *P-value < 0.05, **P-value < 0.01, ***P-value < 0.005, ****P-value < 0.001, ns = not significant. Data is shown as a mean ± SEM of six independent experiments (i.e. six independent human donors).

Materials and methods

Protein production and purification

Ligands for co-stimulation receptors

The soluble extracellular domain (ECD) fused to a c-terminus His-tag and AviTag of human CD58, ICAM-1, CD86, 4-1BBL, and CD70 were produced in adherent HEK 293T cells. Plasmids encoding TNFSR ligands (4-1BBL and CD70) were a kind gift from Harald Wajant (Wurzburg, Germany) and incorporate a Flag¨ tag and tenascin-C trimerisation domain [50]. Adherent HEK 293T cells were transfected with Xtremegene HP reagent (Roche) according to manufacturer’s protocol. Supernatants were filtered using a 0.45 µM filter and proteins purified on a Ni-NTA agarose column. Biotinylation of proteins was performed either in vitro using the BirA enzyme (Avidity, USA) or in situ through co-transfection of BirA encoding plasmid (10%) with 100 µM D-biotin. Additional purification with removal of excess biotin was performed through size exclusion chromatography in HBS-EP. Purified proteins were stored at −80°C.

Peptide-MHC monomers

The 9V variant (SLLMWITQV) derived from the wild-type NY-ESO-1157 165 9C peptide was synthesised at a purity of >95% (Peptide Protein Research, UK). Class 1 pMHCs were produced by expressing Human HLA-A*0201 heavy chain and human -2microglobulin with a C-terminal BirA tag in Escherichia coli as inclusion bodies and refolded with the 9V peptide in vitro as described previously [22, 24]. Protein was biotinylated using the BirA enzyme (Avidity, USA) and purified via size-exclusion chromatography (Superdex S75 column, GE Healthcare, USA), in HBS-EP buffer (10 mM M HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% v/v Tween-20). Purified protein was aliquoted and stored at −80°C until use.

Peptide-MHC tetramers

Fluorescently conjugated pMHC tetramers were produced to enable the detection of antigen receptors (TCR/ CAR). Streptavidin-PE was gradually added to biotinylated 9V pMHC at a 4:1 molar ratio while shaking. Tetramers were stored for up to 3 months at 4°C in the dark.

Production of TCR or CAR transduced primary human CD8+ T cells

Primary human CD8+ T-cell isolation

Human CD8+ T cells were isolated from leukocyte cones purchased from the National Health Service’s (UK) Blood and Transplantation service. Isolation was performed using negative selection. Briefly, blood samples were incubated with Rosette-Sep Human CD8+ enrichment cocktail (Stemcell) at 150 µl/ml for 20 min. This was followed by a 3.1-fold dilution with PBS before layering on Ficoll Paque Plus (GE) at a 0.8:1.0 ficoll to sample ratio. Ficoll-sample preparation was spun at 1200g for 20 min at room temperature. Buffy coats were collected, washed, and isolated cells counted. Cells were resuspended in complete RMPI (RPMI supplemented with 10% v/v FBS, 1% penicillin/ streptomycin) with 50 Units/ml of IL-2 (PeproTech) and CD3/CD28 human T-activator Dynabeads (Thermo Fisher) at a 1:1 bead to cell ratio. At all times isolated human CD8+ T cells were cultured at 37 and 5% CO2.

Lentivirus production

HEK 293T cells were seeded in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin in six-well plates to reach 60–80% confluency on the following day. Cells were transfected with 0.25 pRSV-Rev (Addgene, 12253), 0.53 µg pMDLg/pRRE (Addgene, 12251), 0.35 µg pMD2.G (Addgene, 12259), and 0.8 µg of transfer plasmid using 5.8 X-tremeGENE HP (Roche). Media was replaced after 16 h and supernatant harvested after a further 24 h by filtering through a 0.45 cellulose acetate filter. Supernatant from one well of a six-well plate was used to transduce 1 million T cells.

TCR and CAR transduction

One million cells in 1 ml of complete RPMI were grown overnight in TC-treated 12 well plates. On the following day, T cells were transduced using lentivirus encoding for 1G4 TCR or CAR constructs. On Days 2 and 4 post-transduction, 1 ml of media was exchanged and IL-2 was added to a final concentration of 50 Units/ml. Dynabeads were magnetically removed on Day 5 post-transduction. T cells were then cultured at a density of 1 million cells/ml and supplemented with 50 Units/ml IL-2 every other day. T cells were used between 10 and 16 days after transduction.

Plate stimulation assay

Biotinylated pMHC and biotinylated costimulatory ligand (CD58, ICAM-1, CD86, CD70, or 4-1BBL) were diluted to the required concentration and added to Pierce Streptavidin Coated High Capacity 96 well plates (Thermo Fisher) in 50 µl PBS and incubated for 45 min at room temperature or left overnight at 4°C.

The 96-well plates were washed twice in PBS to remove excess protein. CD8+ T-cell blasts T cells transduced with TCR or CAR, were counted and washed before being added to wells in the 96 well plate. 75 000 T cells were added in 200 µl complete RPMI unless otherwise stated. Cells were spun briefly at 50g, 37°C for 1 min to allow them to settle at the bottom of each well and make contact with relevant protein, before incubation for the required time at 37°C 5% CO2. At the end of the stimulation assay, the supernatant was carefully removed and saved for ELISA analysis. 10 mM EDTA in PBS was then added to detach the T cells. The cells were then aspirated and transferred to a v-bottom plate and washed once in 200 µl PBS 1% BSA (500g, 4°C, 5 min). Antibodies against T-cell activation markers were diluted in PBS 1% BSA at a 1:200 dilution. To detect TCR-CAR expression fluorescently conjugated peptide-MHC tetramers were added to the staining antibodies at a 1:1000 dilution. A viability dye was also added at a dilution of 1:2500 to distinguish live cells from dead cells. The cells were treated with 50 µl of the staining solution before incubating them for 20 min at 4°C in the dark. The cells were washed twice in PBS, and resuspended in 75 µl PBS, before running on a flow cytometer. Flow cytometry data was analysed using FlowJo (BD Biosciences).

Cytokine detection

IL-2 human uncoated ELISA kit, TNF-a human uncoated ELISA kit, IFN-g human uncoated ELISA kit, and Nunc MaxiSorp 96-well plates were used according to the manufacturer’s instructions. The supernatant from stimulation assays was diluted 1 in 15 for ELISAs. The absorbance at 450 nm and 570 nm were measured using a SpectraMax M5 plate reader (Molecular Devices).

Data analysis

All statistical tests were performed in PRISM V.9 (GraphPad Software). ELISA standard curves were fitted using linear regression.

CAR and TCR sequences

The CARs we have used have been previously described [17]. The 2nd generation CAR contains the following architecture:

D52N scFv—FLAG − CD28 hinge [P10747112 152] − CD28 TM [P10747153 179] − CD28 ICD [P10747180 220]. − CD247 z-chain ICD [P2096352 164] with UniProtKB accession numbers indicated and the D52N scFv sequence is

EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYQMSWVRQAPGKGLEW

VSGIVSSGGSTAYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY

YCAGELLPYYGMDVWGQGTTVTVSSAKTTPKLEEGEFSEARVQSELT

QPRSVSGSPGQSVTISCTGTERDVGGYNYVSWYQQHPGKAPKLIIHN

VIERSSGVPDRFSGSKSGNTASLTISGLQAEDEADYYCWSFAGGYYV FGTGTDVTVLG

The 1st generation CAR is identical to the above except without the CD28 ICD.

The sequence of the 1G4 wild-type TCR we have used containing human constant domains is as follows

1G4 alpha-chain:

METLLGLLILWLQLQWVSSKQEVTQIPAALSVPEGENLVLNCS

FTDSAIYNLQWFRQDPGKGLTSLLLIQSSQREQTSGRLNASLD KSSGRSTLYIAASQPGDSATYLCAVRPTSGGSYIPTFGRGTSL

IVHPYIQNPDPAVYQLRDSKSSDKSVCLFTDFDSQTNVSQSKD

SDVYITDKTVLDMRSMDFKSNSAVAWSNKSDFACANAFNNSII

PEDTFFPSPESSCDVKLVEKSFETDTNLNFQNLSVIGFRILLL KVAGFNLLMTLRLWSS

1G4 beta-chain:

MSIGLLCCAALSLLWAGPVNAGVTQTPKFQVLKTGQSMTLQCA

QDMNHEYMSWYRQDPGMGLRLIHYSVGAGITDQGEVPNGYNVS RSTTEDFPLRLLSAAPSQTSVYFCASSYVGNTGELFFGEGSRL

TVLEDLKNVFPPEVAVFEPSEAEISHTQKATLVCLATGFYPDH

VELSWWVNGKEVHSGVSTDPQPLKEQPALNDSRYCLSSRLRVS

ATFWQNPRNHFRCQVQFYGLSENDEWTQDRAKPVTQIVSAEAW

GRADCGFTSESYQQGVLSATILYEILLGKATLYAVLVSALVLM

AMVKRKDSRG

Results

The temporal production of cytokines is tuned differently by different T-cell co-stimulation receptors

To study the impact of T-cell co-stimulation on the kinetics of cytokine production, we transduced primary human CD8+ T cells with the 1G4 TCR recognising a modified peptide from the NY-ESO-1157 165 cancer testis antigen (SLLMWITQV, 9V) displayed on HLA-A*02:01 [23, 24]. We stimulated these T cells with purified biotinylated pMHC antigen alone or together with the purified biotinylated extracellular domain of ligands to the co-stimulation receptors CD2 (CD58), LFA-1 (ICAM-1), CD28 (CD86), CD27 (CD70), and 4-1BB (4-1BBL) (Fig. 1A). We included purified biotinylated rat CD4 as a negative control protein to ensure that addition of ligands did not sterically impact the ability of T cells to recognise pMHC. We coupled biotinylated antigens and ligands to streptavidin surfaces because this stimulation platform allowed for precisely defined surfaces that could persistently activate T cells without other compensatory/redundant molecules. We have previously observed [15, 21] that co-stimulation can prevent T-cell desensitisation to TCR signals over a period of 20 h leading to different outcomes for cytokine accumulation and production rates (Fig. 1B). To parse cytokine production kinetics the amount of IFN-g, IL-2, and TNF-a was measured after 12 h of stimulation (early production), the media was then replaced and the amount of the same cytokines were measured over the next 8 h (late production) (Fig. 1C). As a control, we also measured the total cytokine produced after 20 h without replacing the media (total production) (Fig. 1D).

Consistent with their known costimulatory role, all tested costimulatory ligands increased total production of almost all cytokines (Fig. 1E). The single exception was that CD70 did not increase TNF-a production. The CD2 costimulatory ligand CD58 had the largest impact on the production of all three cytokines. The second most potent costimulatory ligand varied with cytokine: it was the LFA-1 ligand ICAM-1 for IFN-g and the CD28 ligand CD86 for IL-2 and TNF-a.

Interestingly, distinct effects were seen when comparing early versus late production (Fig. 1E), especially for IL-2 and TNF-a. While most ligands increased early production of each cytokine, only some increased late production. For example, CD2, LFA-1, CD28, and 4-1BB engagement increased early production of TNF-a but only 4-1BB engagement increased late production.

To directly compare early and late production, we calculated the cytokine production rate by dividing by the stimulation duration (Fig. 1F). The rate of cytokine production decreased between early and late production for all three cytokines when T cells were stimulated with pMHC antigen alone. Interestingly, co-stimulatory ligands differed in their effect on late cytokine production. 4-1BB co-stimulation maintained the production rate of IL-2 and TNF-a, and increased the production rate of IFN-g. CD2 and LFA-1 costimulation maintained the production rate of IFN-g, but not IL-2 and TNF-a. Finally, CD28 and CD27 co-stimulation failed to maintain the production rate of any of the three cytokines.

Previous work has suggested that T cells can tune their cytokine production based on feedback from extracellular cytokine levels [25, 26]. To exclude this we examine the effect of varying the density of T cells in the culture (Supplementary Fig. S1). Our finding that cytokine levels scaled linearly with cell numbers shows that cytokine production is not affected by extracellular cytokine levels, ruling out feedback effects in this experimental system.

Taken together, these results indicate that sustained (20 h) cytokine production induced by TCR engagement requires co-stimulation and that co-stimulatory receptors differ in their impact on cytokine production.

Unregulated cytokine production by 2nd generation CAR-T cells

We next compared the early and late production of cytokines by CAR-T cells. We used a previously described 1st generation CAR containing the z-chain cytoplasmic domain and a 2nd generation CARs contain both the z-chain and CD28 cytoplasmic domains [17]. These CARs use the D52N scFv, which like the 1G4 TCR, recognise the NY-ESO-1 pMHC (Fig. 2A). The D52N scFv is derived from the 3M4EF Fab, which binds pMHC in the same orientation as the TCR [27, 28]. In previous work we found that these CARs required antigen for cytokine production and that CAR expression did not induce T-cell exhaustion, suggesting that these CARs produce little (or no) tonic signalling [17]. Using pMHC tetramers, we confirmed that both CARs expressed at similar levels to the TCR (Fig. 2B).

Figure 2.

Figure 2.

Regulated production of the cytokine IFN-g by co-stimulation receptors for 1st generation CAR-T cells is reduced for 2nd generation CAR-T cells. (A) Schematic of 1st and 2nd generation CARs that use the D52N scFv to recognize the NY-ESO-1 pMHC. (B) Surface expression of each antigen receptor on transduced primary human CD8+ T cells is detected using pMHC tetramers in flow cytometry. (C) Rate of IFN-g production for the indicated conditions. Statistical significance was determined by multiple t-test on log-transformed data relative to the pMHC alone condition with a Holm–Sidak multiple comparisons correction. Abbreviations: *P-value < 0.05, **P-value < 0.01, ***P-value < 0.001, ****P-value < 0.0001, ns = not significant. Data is shown as a mean ± SEM of five independent experiments (i.e. five independent human donors).

We measured total, early, and late cytokine production by TCR and CAR-expressing CD8+ T cells in response to pMHC antigen alone or with co-stimulation ligands. As before, we measured cytokine levels (Supplementary Fig. S2) and calculated production rates by dividing by the stimulation duration (Figs. 2C and 3). Only CD2 engagement increased early IFN-g production induced by CAR stimulation, while CD2 and 4-1BB engagement increased late IFN-g production (Fig. 2C). CD2, CD28, and 4-1BB engagement enhanced early IL-2 and TNF-a production induced by stimulation via TCR and both CARs (Fig. 3). CD2, CD28, and 4-1BB engagement also enhanced late IL-2 and TNF-a production induced by stimulation via TCR and the 1st generation CAR (Fig. 3). However, only 4-1BB engagement enhanced late IL-2 and TNF-a production induced by stimulation of the 2nd generation CAR (Fig. 3).

Figure 3.

Figure 3.

Regulated production of the cytokine IL-2 and TNF-a by co-stimulation receptors for 1st generation CAR-T cells is reduced for 2nd generation CAR-T cells. The production rate of IL-2 (top) and TNF-a (bottom) for the indicated antigen receptor, production period, and co-stimulation ligand. Statistical significance was determined by multiple t-test on log-transformed data relative to the pMHC alone condition with a Holm–Sidak multiple comparisons correction. Abbreviations: *P-value < 0.05, **P-value < 0.01, ***P-value < 0.001, ****P-value < 0.0001, ns = not significant. Data is shown as a mean ± SEM of five independent experiments (i.e. five independent human donors).

Interestingly T cells expressing a 2nd generation CAR had a striking ability to maintain a high rate of late cytokine production in response to antigen alone (Figs. 23). This is more easily observed when plotting the data as heatmaps (Fig. 4) or when plotting early cytokine production against late cytokine production (Supplementary Fig. S3). In the case of the TCR and 1st generation CARs, the production rate of all cytokines decreased between early and late periods in response to antigen alone. Engagement of CD2 or 4-1BB was required to maintain late cytokine production. In striking contrast, 2nd generation CARs were able to maintain a high rate of cytokine production in response to antigen alone, even without co-stimulation.

Figure 4.

Figure 4.

Cytokine production rates are maintained by 2nd generation CAR-T cells largely independent of extrinsic co-stimulation. The mean cytokine production rates from Figs. 2 and 3 are displayed in heatmaps.

As a control experiment, we examined whether the sum of early and late production was equal to the total production for each cytokine (Supplementary Fig. S4). We found that the sum of early and late exhibited a modest increase over total production for all three cytokines. A likely explanation for this is increased degradation/consumption of cytokines after secretion in the total versus the early/late production experiments. In the early/late experiments, supernatant is removed after 12 h and stored at low temperatures, which would halt degradation/consumption. An alternative explanation is that the secreted cytokine-induced negative feedback reduces cytokine production rates preferentially in the total stimulation experiment. However, we failed to find evidence for secreted cytokine levels influencing production rates in our cell titration experiments (Supplementary Fig. S1).

Discussion

Using a reductionist system, we have found that cytokine production by T cells is highly regulated by extrinsic co-stimulation ligands when recognising antigens through the TCR or 1st generation CAR. However, 2nd generation CARs displayed largely unregulated cytokine production that is maintained independent of extrinsic co-stimulation receptors. The analysis we have performed can be extended in a number of ways, including the study of co-inhibitory receptors and combinations thereof.

The ability of T cells to orchestrate immune responses relies on finely tuned production of cytokines. This may be important to enable the clearance of pathogens without excessive tissue damage [7]. We found that the rate of cytokine production decreases when T cells recognise antigens in isolation, as in our previous work [15, 21]. As a result, T cells become increasingly reliant on signalling from co-stimulation receptors to sustain cytokine production. The sustained production of IFN-g, IL-2, and TNF-a over 20 h could be achieved by 4-1BB co-stimulation whereas co-stimulation by CD2 and LFA-1 could only sustain IFN-g production. Although CD28 and CD27 co-stimulation increased the early rate of cytokine production (except TNF-a for CD27), they could not sustain this higher rate with CD27 co-stimulation completely absent over the late production period. The impact of all of these co-stimulation receptors has previously been shown to be TCR dependent [15, 21, 29–32], although in the case of 4-1BB there is also evidence for a TCR independent signalling function [33, 34], which may explain its unique ability to sustain cytokine production. We have used expanded primary human CD8+ T cells and a similar ‘wave’ of cytokine production has been observed in a variety of un-expanded primary T cells, including naive, central memory, and effector memory when stimulated by the TCR and CD28 [35]. Taken together, these results suggest that sustained cytokine production by T cells relies on extrinsic co-stimulation receptors.

In contrast to the TCR, we found that antigen recognition by 2nd generation CARs produced largely unregulated cytokine production. Whereas the TCR and 1st generation CARs required extrinsic ligation of co-stimulation receptors to increase and sustain cytokine production, 2nd generation CARs maintained (IL-2, TNF-a) or increased (IFN-g) their rate of cytokine production over time independent of extrinsic co-stimulation. The only detectable regulation was by 4-1BB which led to an even higher production rate of all cytokines, which may be a result of the non-overlapping signalling pathways induced by CD28 and 4-1BB [3, 33]. We hypothesise that this regulation by the 4-1BB co-stimulation receptor will be lost in 2nd generation CARs that rely on the 4-1BB rather than the CD28 co-stimulation domain. Moreover, we predict that 3rd generation CARs that contain both CD28 and 4-1BB co-stimulation domains would not exhibit higher cytokine production by engagement of CD28 and 4-1BB receptors, as their signalling pathways would be fully activated by the CAR [36, 37].

The ability of co-stimulation receptors to increase cytokine production by a population of cells may result from increasing the fraction of activated T cells and/or increasing the amount of cytokine produced by each T cell [38–40]. However, this dichotomy can be blurred in situations where T cells can be activated (e.g. as measured by surface CD69) but where cytokine cannot be detected. In these situations, co-stimulation receptors may amplify intracellular signalling to engage cytokine production pathways in already activated T cells. Indeed, we have used a high concentration of pMHC in the present study (250 ng/well), which we previously determined activates the majority of T cells in this experimental system as determined by single-cell flow cytometry for surface CD69 [17]. T cells produce only modest levels of the cytokine IL-2 in this condition in the absence of co-stimulation. It follows that engaging co-stimulation receptors is stimulating IL-2 production from already activated T cells. Similarly, the early production of IFN-g induced by antigen alone is lost in late production for the TCR unless co-stimulation is included, suggesting that T cells were initially activated. Taken together, these results suggest that co-stimulation receptors act to induce cytokine production in already activated T cells.

Ligand engagement of co-stimulation receptors could enhance cytokine production in part by increasing cell adhesion. We note that CD70 and 4-1BBL are both high-affinity trimers [41] yet only 4-1BBL is able to sustain late cytokine production. Conversely, CD86, ICAM-1, and CD58 are lower-affinity monomers [42–44] yet CD58 generally has a larger impact in late production compared to CD86 and ICAM-1. Thus ligand binding strength/valency does not correlate with costimulatory activity. More likely explanations include differences in signalling pathways or expression patterns of costimulation receptors. In previous work, we have suggested that the TCR-signalling induced expression of 4-1BB but not CD27 can explain the ability of 4-1BB but not CD27 to sustain cytokine production [21].

A key difference between TCRs and CARs is their affinity for their target antigens. While TCRs typically bind their antigens with KD values larger than 1 µM [45], antibody-derived CARs bind antigens with KD values lower than 1 µM and often into the nM regime [46, 47]. Our finding that 1st generation CAR displayed a similar pattern of regulated cytokine production to the TCR, despite binding with a 50-fold higher affinity [17], suggests that differences in antigen affinity do not explain the difference reported in this study between the TCR and 2nd generation CARs. The CAR/antigen affinity has been shown to control antigen sensitivity [48, 49], including for the D52N CAR used in this study [17].

The unregulated production of cytokines by CAR-T cells is a double-edged sword. On the one hand, cancer cells are known to reduce the expression of ligands to co-stimulation receptors to escape T-cell immunity. As a result, reducing the dependence on co-stimulation ligands for cytokine production is a useful feature in reducing the ability of cancers to evade CAR-T cell responses. On the other hand, the inability to extrinsically control CAR-T cells means that the normal regulatory processes of adaptive immunity are lost increasing the possibility of cytokine storms leading to excessive inflammatory responses and tissue damage.

Supplementary Material

ltae004_suppl_Supplementary_Material

Acknowledgements

We thank Johannes Pettmann and John Nguyen for their assistance in protein production. The Editor-in-Chief, Tim Elliott, and handling editor, Stephanie Dougan, would like to thank the two anonymous reviewers for their contribution to the publication of this article.

Glossary

Abbreviations

CAR

Chimeric antigen receptor

CRS

Cytokine release syndrome;

pMHC

peptide major-histocompatibilitycomplex

TCR

T cell receptor

TNFSF

Tumour necrosis factor superfamily

Contributor Information

Ashna Patel, The Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.

Mikhail A Kutuzov, The Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.

Michael L Dustin, The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK.

P Anton van der Merwe, The Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.

Omer Dushek, The Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.

Author contributions

Ashna Patel (data curation, formal analysis, investigation, methodology, visualisation, writing—original draft, writing—review & editing), Mikhail A. Kutuzov (investigation), Mike Dustin (investigation, supervision, writing—review & editing), P. Anton van der Merwe (investigation, supervision, writing—review & editing), and Omer Dushek (conceptualisation, data curation, formal analysis, funding acquisition, investigation, project administration, supervision, visualisation, writing—original draft, writing—review & editing)

Funding

The work was funded by a Wellcome Trust Senior Fellowship in Basic Biomedical Sciences (207537/Z/17/Z to OD), by a Biotechnology and Biological Sciences Research Council (BBSRC) studentship (grant number BB/M011224/1 to AP), and by a Kennedy Trust for Rheumatology Research Professorship (to MLD).

Conflict of interest

Omer Dushek and P. Anton van der Merwe have financial interests MatchBio Ltd.

Ethics approval

Not applicable.

Data availability

The data underlying this article are available in the article and in its online supplementary material.

References

  • 1. Smith-Garvin JE, Koretzky G, Jordan MS.. T cell activation. Annu Rev Immunol 2009; 27:591–619. 10.1146/annurev.immunol.021908.132706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Lever M, Maini PK, van der Merwe PA. et al. Phenotypic models of T cell activation. Nat Rev Immunol 2014; 14:619–29. 10.1038/nri3728 [DOI] [PubMed] [Google Scholar]
  • 3. Chen L, Flies DB.. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013; 13:227–42. 10.1038/nri3405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Attanasio J, Wherry EJ.. Costimulatory and coinhibitory receptor pathways in infectious disease. Immunity 2016; 44:1052–68. 10.1016/j.immuni.2016.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Callahan MK, Postow MA, Wolchok JD.. Targeting T cell co-receptors for cancer therapy. Immunity 2016; 44:1069–78. 10.1016/j.immuni.2016.04.023 [DOI] [PubMed] [Google Scholar]
  • 6. Zhang Q, Vignali DA.. Co-stimulatory and co-inhibitory pathways in autoimmunity. Immunity 2016; 44:1034–51. 10.1016/j.immuni.2016.04.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Hashimoto M, Kamphorst AO, Im SJ. et al. CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions. Annu Rev Med 2018; 69:301–18. 10.1146/annurev-med-012017-043208 [DOI] [PubMed] [Google Scholar]
  • 8. Baulu E, Gardet C, Chuvin N. et al. TCR-engineered T cell therapy in solid tumors: state of the art and perspectives., Technical report. Sci Adv 2023; 9:eadf3700. 10.1126/sciadv.adf3700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Baker DJ, Arany Z, Baur JA. et al. CAR T therapy beyond cancer: the evolution of a living drug. Nature 2023; 619:707–15. 10.1038/s41586-023-06243-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Labanieh L, Mackall CL.. CAR immune cells: design principles, resistance and the next generation. Nature 2023; 614:635–48. 10.1038/s41586-023-05707-3 [DOI] [PubMed] [Google Scholar]
  • 11. Chmielewski M, Hombach AA, Abken H.. CD28 cosignalling does not affect the activation threshold in a chimeric antigen receptor-redirected T-cell attack. Gene Ther 2011; 18:62–72. 10.1038/gt.2010.127 [DOI] [PubMed] [Google Scholar]
  • 12. Cheadle EJ, Rothwell DG, Bridgeman JS. et al. Ligation of the CD2 co-stimulatory receptor enhances IL-2 production from first-generation chimeric antigen receptor T cells. Gene Ther 2012; 19:1114–20. 10.1038/gt.2011.192 [DOI] [PubMed] [Google Scholar]
  • 13. Majzner RG, Frank MJ, Mount C. et al. CD58 aberrations limit durable responses to CD19 CAR in large B cell lymphoma patients treated with axicabtagene ciloleucel but can be overcome through novel CAR engineering. Blood 2020; 136:53–4. 10.1182/blood-2020-139605 [DOI] [Google Scholar]
  • 14. Salter AI, Rajan A, Kennedy JJ. et al. Comparative analysis of TCR and CAR signaling informs CAR designs with superior antigen sensitivity and in vivo function. Sci Signaling 2021; 14:eabe2606. 10.1126/scisignal.abe2606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Trendel N, Kruger P, Gaglione S. et al. Perfect adaptation of CD8+ T cell responses to constant antigen input over a wide range of affinity is overcome by costimulation. Sci Signal 2021; 666:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Larson RC, Kann MC, Bailey SR. et al. CAR T cell killing requires the IFN R pathway in solid but not liquid tumours. Nature 2022; 604:563–70. 10.1038/s41586-022-04585-5 [DOI] [PubMed] [Google Scholar]
  • 17. Burton J, Siller-Farfán JA, Pettmann J. et al. Inefficient exploitation of accessory receptors reduces the sensitivity of chimeric antigen receptors. Proc Natl Acad Sci USA 2023; 120:e2216352120. 10.1073/pnas.2216352120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Morris EC, Neelapu SS, Giavridis T. et al. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol 2021; 22(22):85–96. 10.1038/s41577-021-00547-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lever M, Lim H-S, Kruger P. et al. A minimal signalling architecture explains the T cell response to a 1,000,000-fold variation in antigen affinity and dose. Proc Natl Acad Sci USA 2016; 113:E6630–8. 10.1073/pnas.1608820113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Abu-Shah E, Trendel N, Kruger P. et al. Human CD8 + T cells exhibit a shared antigen threshold for different effector responses. J Immunol 2020; 205:1503–12. 10.4049/jimmunol.2000525 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Nguyen J, Pettmann J, Kruger P. et al. Quantitative contributions of TNF receptor superfamily members to CD8 + T-cell responses. Mol Syst Biol 2021; 17:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pettmann J, Huhn A, Abu Shah E. et al. The discriminatory power of the t cell receptor. eLife 2021; 10:1–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Chen JL, Stewart-Jones G, Bossi G. et al. Structural and kinetic basis for heightened immunogenicity of T cell vaccines. J Exp Med 2005; 201:1243–55. 10.1084/jem.20042323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Aleksic M, Dushek O, Zhang H. et al. Dependence of T cell antigen recognition on T cell receptor-peptide MHC confinement time. Immunity 2010; 32:163–74. 10.1016/j.immuni.2009.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Tkach KE, Barik D, Voisinne G. et al. T cells translate individual, quantal activation into collective, analog cytokine responses via time-integrated feedbacks. eLife 2014; 3:e01944. 10.7554/eLife.01944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Mayya V, Dustin ML.. What scales the T cell response? Trends Immunol 2016; 37:513–22. 10.1016/j.it.2016.06.005 [DOI] [PubMed] [Google Scholar]
  • 27. Stewart-jones G, Wadle A, Hombach A. et al. Rational development of high-affinity T-cell receptor-like antibodies. Proc Natl Acad Sci USA 2009; 106:5784–8. 10.1073/pnas.0901425106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Maus MV, Plotkin J, Jakka G. et al. An MHC-restricted antibody-based chimeric antigen receptor requires TCR-like affinity to maintain antigen specificity. Mole Therapy Oncol 2016; 3:16023. 10.1038/mto.2016.23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Martin PJ, Ledbetter JA, Morishita Y. et al. A 44 kilodalton cell surface homodimer regulates interleukin 2 production by activated human T lymphocytes. J Immunol 1986; 136:3282–7. 10.4049/jimmunol.136.9.3282 [DOI] [PubMed] [Google Scholar]
  • 30. Bockenstedt LK, Goldsmith MA, Dustin M. et al. The CD2 ligand LFA-3 activates T cells but depends on the expression and function of the antigen receptor. J Immunol 1988; 141:1904–11. [PubMed] [Google Scholar]
  • 31. Beinke S, Phee H, Clingan JM. et al. Proline-rich tyrosine kinase-2 is critical for CD8 T-cell short-lived effector fate. Proc Natl Acad Sci USA 2010; 107:16234–9. 10.1073/pnas.1011556107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Dadas O, Ertay A, Cragg MS.. Delivering co-stimulatory tumor necrosis factor receptor agonism for cancer immunotherapy: past, current and future perspectives. Front Immunol 2023; 14:1147467. 10.3389/fimmu.2023.1147467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Muller J, Baeyens A, Dustin ML.. Tumor necrosis factor receptor superfamily in T cell priming and effector function. Adv Immunol 2018; 140:21–57. 10.1016/bs.ai.2018.08.001 [DOI] [PubMed] [Google Scholar]
  • 34. Otano I, Azpilikueta A, Glez-Vaz J. et al. CD137 (4-1BB) costimulation of CD8+ T cells is more potent when provided in cis than in trans with respect to CD3-TCR stimulation. Nat Commun 2021; 12:7296. 10.1038/s41467-021-27613-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Legat A, Speiser DE, Pircher H. et al. Inhibitory receptor expression depends more dominantly on differentiation and activation than ”exhaustion” of human CD8 T cells. Front Immunol 2013; 4:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ritmeester-Loy SA, Draper IH, Bueter EC. et al. Differential protein–protein interactions underlie signaling mediated by the TCR and a 4-1BB domain-containing CAR. Sci Signaling 2024; 17:eadd4671. 10.1126/scisignal.add4671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Karlsson H, Svensson E, Gigg C. et al. Evaluation of intracellular signaling downstream chimeric antigen receptors. PLoS One 2015; 10:e0144787. 10.1371/journal.pone.0144787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Das J, Ho M, Zikherman J. et al. Digital signaling and hysteresis characterize ras activation in lymphoid cells. Cell 2009; 136:337–51. 10.1016/j.cell.2008.11.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Huang J, Brameshuber M, Zeng X. et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4+ T cells. Immunity 2013; 39:846–57. 10.1016/j.immuni.2013.08.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. James JR. Tuning ITAM multiplicity on T cell receptors can control potency and selectivity to ligand density. Sci Signaling 2018; 11:eaan1088. 10.1126/scisignal.aan1088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Lang I, Füllsack S, Wyzgol A. et al. Binding studies of TNF receptor superfamily (TNFRSF) receptors on intact cells. J Biol Chem 2016; 291:5022–37. 10.1074/jbc.M115.683946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. van der Merwe PA, Barclay AN, Mason DW. et al. Human cell-adhesion molecule CD2 binds CD58 (LFA-3) with a very low affinity and an extremely fast dissociation rate but does not bind CD48 or CD59. Biochemistry 1994; 33:10149–60. 10.1021/bi00199a043 [DOI] [PubMed] [Google Scholar]
  • 43. Tominaga Y, Kita Y, Satoh A. et al. Affinity and kinetic analysis of the molecular interaction of ICAM-1 and leukocyte function-associated antigen-1. J Immunol (Baltimore, Md. : 1950) 1998; 161:4016–22. [PubMed] [Google Scholar]
  • 44. Collins AV, Brodie DW, Gilbert RJC. et al. The interaction properties of costimulatory molecules revisited. Immunity 2002; 17:201–10. 10.1016/s1074-7613(02)00362-x [DOI] [PubMed] [Google Scholar]
  • 45. Yin Y, Li Y, Mariuzza RA.. Structural basis for self-recognition by autoimmune T-cell receptors. Immunol Rev 2012; 250:32–48. 10.1111/imr.12002 [DOI] [PubMed] [Google Scholar]
  • 46. Mao R, Kong W, He Y.. The affinity of antigen-binding domain on the antitumor efficacy of CAR T cells: moderate is better. Front Immunol 2022; 13:1032403. 10.3389/fimmu.2022.1032403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. He C, Mansilla-Soto J, Khanra N. et al. CD19 CAR antigen engagement mechanisms and affinity tuning. Sci Immunol 2023; 8:eadf1426. 10.1126/sciimmunol.adf1426 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Caruso HG, Hurton LV, Najjar A. et al. Tuning sensitivity of CAR to EGFR density limits recognition of normal tissue while maintaining potent antitumor activity. Cancer Res 2015; 75:3505–18. 10.1158/0008-5472.CAN-15-0139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Liu X, Jiang S, Fang C. et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res 2015; 75:3596–607. 10.1158/0008-5472.CAN-15-0159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Wyzgol A, Müller N, Fick A. et al. Trimer stabilization, oligomerization, and antibody-mediated cell surface immobilization improve the activity of soluble trimers of CD27L, CD40L, 41BBL, and glucocorticoidinduced TNF receptor ligand. J Immunol 2009; 183:1851–61. 10.4049/jimmunol.0802597 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ltae004_suppl_Supplementary_Material

Data Availability Statement

The data underlying this article are available in the article and in its online supplementary material.


Articles from Immunotherapy Advances are provided here courtesy of Oxford University Press

RESOURCES