Adoptive cell immunotherapy with chimeric antigen receptor (CAR) showed limited potency in solid tumors, despite durable remissions for hematopoietic malignancies. Therefore, an investigation of ways to enhance the efficacy of CARs' antitumor response has been engaged upon. We previously examined the interplay between the biophysical parameters of CAR binding (i.e., affinity, avidity, and antigen density), as regulators of CAR T-cell activity and detected nonmonotonic behaviors of affinity and antigen density and an interrelation between avidity and antigen density. Here, we built an evolving phenotypic model of CAR T-cell regulation, which suggested that receptor downmodulation is a key determinant of CAR T-cell function. We verified this assumption by measuring and manipulating receptor downmodulation and intracellular signaling processes. CAR downmodulation inhibition, via actin polymerization inhibition, but not inhibition of regulatory inhibitory phosphatases, was able to increase CAR T-cell responses. In addition, we documented trogocytosis in CAR T cells that depends on actin polymerization. In summary, our study modeled the parameters that govern CAR T-cell engagement and revealed an underappreciated mechanism of T-cell regulation. These results have a potential to predict and therefore advance the rational design of CAR T cells for adoptive cell treatments.

See related article on p. 872

This article is featured in Highlights of This Issue, p. 761

The notable success of cancer-specific adoptive immunotherapies raised much interest in recent years, as these methodologies have shown durable clinical responses for treating hematopoietic malignancies and recently also for solid malignancies (1–3). These new immune-based methodologies, such as bispecific antibodies (bsAb) and chimeric antigen receptors (CARs), emerged as tools to redirect T cells to efficiently eliminate tumor cells (4, 5). Still, the limited outcome in solid malignancies highlights the need for better understanding of the molecular mechanisms that lead to optimal response and of how these mechanisms can be manipulated to rationally design such techniques.

CAR T cells are engineered to express hybrid receptors composed of extracellular high-affinity variable fragments of an antibody (scFv) linked to intracellular activatory signaling domains. Typically, scFvs with high affinity are selected; however, increased affinity might lead to nonbeneficial or reduced efficacy (6–9), or to reduced specificity (10, 11). Likewise, we now demonstrated that CAR T-cell response is nonmonotonic, and medium affinity CAR T cells outperformed high-affinity CAR T cells (12). We compared between the contribution of affinity, avidity, and antigen density in a unique and quantitative experimental system, and described the relation between these parameters. Antigen density also has a nonmonotonic behavior, whereas avidity had a monotonic behavior. Although each of these parameters seems to affect T-cell functionality separately, avidity seems to be related to antigen density, thereby increasing sensitivity and the magnitude of response.

The rational design of optimal CAR properties for adoptive cell therapy can be aided by a model of CAR T-cell activation that can predict responses of CARs of varying affinities and avidities to a range of antigen density. Classical T-cell receptor (TCR) activation that occurs upon engagement with its cognate antigen depends on the affinity, avidity, and antigen density, and consequentially initiates a signaling cascade that induces activation. Models that predict how the binding parameters determine T-cell response include regulatory elements such as kinetic proofreading, serial triggering, signaling network motifs, and receptor downmodulation (13–15).

Based on our phenotypical observations (12), we set to model CAR activation using classical mathematical TCR models; however, these models only partially explained CAR T-cell response. We therefore generated an improved dynamical model that addresses the number of receptors. This revised model suggests that receptor downmodulation provides an impetus for CAR T-cell regulation. To test this hypothesis, we measured and compared receptor downmodulation and intracellular inhibitory signaling processes. CAR downmodulation was evident rapidly following encounter with target cells. We observed similar kinetics of intracellular activatory and inhibitory processes, contrary to the kinetics described following TCR activation (16). Finally, CAR internalization inhibition, but not inhibition of regulatory phosphatases (Shp1/Shp2), increased CAR T-cell responses.

Cell growth and drug treatment

Unless otherwise mentioned, cells were cultured in RPMI 1640 medium containing 10% FBS, 100 units/mL penicillin, 100 μg/mL streptomycin, 0.5 mmol/L HEPES, 1 mmol/L sodium pyruvate, nonessential amino acids, and 2 mmol/L glutamine at 37°C in 5% CO2.

Peripheral blood mononuclear cells (PBMCs) and T cells were cultured in T-cell culture medium (T-cell CM) that additionally was supplemented with 0.05 mmol/L 2-mercaptoethanol and the described units of IL2 at 37°C in 5% CO2.

For peptide loading of APCs, cells were incubated with BioGro medium, which contained RPMI 1640 medium, 2% BioGro (Biological Industries), 100 units/mL penicillin, 100 μg/mL streptomycin, 0.5 mmol/L HEPES, 1 mmol/L sodium pyruvate, nonessential amino acids, and 2 mmol/L glutamine at 37°C in 5% CO2.

For drug inhibition of actin polymerization, cells were incubated with either 5 μmol/L or 1 μmol/L Cytochalasin D (CytD, Sigma Aldrich) or control 0.1% DMSO in T-cell CM for 45 minutes prior to beginning of functional assay.

For drug inhibition of Shp1/Shp2 phosphatases, cells were incubated with either 20 or 10 μg/mL of freshly dissolved Sodium Stibogluconate (SSG, Cayman) in T-cell CM for 16 hours prior to beginning of functional assay.

Reagents and antibodies

Unless otherwise mentioned, all media and cell-growth reagents were purchased from Thermo Fisher Scientific. Oligonucleotides were manufactured at high-performance liquid chromatography quality by Sigma-Aldrich. All peptides were ordered from LifeTein.

The following commercial antibodies were used in this work: Anti-mouse Ig kappa light chain (BioLegend), Anti–HLA-A2 (clone BB7.2, Bio-Rad), Anti-CD8 (BD Biosciences), Anti-CD3 (clone SK7, BD Biosciences), Anti-CD3 (clone OKT3, eBioscience), Anti-IFNγ (eBioscience), Anti-IL2 (eBioscience), Anti-TNFα (eBioscience), Anti-Biotin (BioLegend), Anti-CD19 (eBioscience), Anti–p-LAT (pY-226, BD Biosciences), Anti–p-Zap70 (pY-292, BD Biosciences), Anti–p-Slp76 (pY-128, BD Biosciences), Anti–p-cCbl (pY-700, BD Biosciences), and Anti–p-Shp2 (pY-542, BD Biosciences).

The TCR-like antibodies described in the work were received from Adicet Bio and were previously characterized (12).

Production of MHC–peptide complexes

Single-chain MHC–peptide complexes were produced by in vitro refolding of inclusion bodies produced in Escherichia coli upon isopropyl β-d-1-thiogalactopyranoside induction, and MHC tetramers and monomers were generated using labeled streptavidin, as described previously (12, 42).

Peptide loading

Peptides were ordered from LifeTein and dissolved in DMSO. Epstein-Barr virus–transformed JY B-cell lymphoblastoid lines were incubated overnight with the indicated peptides and concentrations in BioGro medium at 37°C as described previously (12). After pulsing, the cells were washed to remove free peptide.

Transduction of retroviral CAR constructs into primary T cells and expansion

Primary PBMCs were obtained from donors, after obtaining their informed consent according to the Technion Institutional Review Board, using density-gradient centrifugation according to the Ficoll-Hypaque technique. CAR constructs described previously (12) were transduced to primary T cells, using the Phoenix amphotropic packaging cells, as described previously (7, 12).

pMHC and receptor quantification

pMHC and receptor were analyzed using TCR-like Fabs or pMHC monomers, respectively, as described previously (43).

Intracellular cytokines detection

Transduced CAR T cells and JY cells loaded with the mentioned peptides were added at 1 × 105 cells/well at 1:1 effector-to-target (E:T) ratio (effector cell number was calculated as the number of live Tyr-MHC pentamer-positive cells). Cells were cultured in sterile 96-well plates in 100 μL of T-cell CM containing brefeldin A and monensin(eBioscience). After 5.5 hours at 37°C with 5% CO2, cells were fixed and permeabilized according to the manufacturer's protocol (Cytofix/Cytoperm Kit; BD Biosciences), and intracellular cytokines staining (ICS) was performed as described in the text. Anti-CD8 or anti-CD3 antibodies were used to gate effector cells from target JY cells. Finally, cells were washed and analyzed by an LSR II flow cytometer (BD Biosciences). The transduced T cells were gated by size and granulation (FSC/side scatter), GFP-positive, and CD3/8-positive cells, as demonstrated in Supplementary Fig. S5B. Reactivity was assessed by measuring percentage of reactive cells and the median fluorescence intensity (MFI) of the reactive cells (reactive-MFI).

Mathematical modeling

We used a phenotypical model, composed of ordinary differential equations reflecting fast processes from receptor–ligand interaction to intracellular signaling cascades, and a slow receptor downmodulation process (see Supplementary Information for detailed model development). As the time scale for the downmodulation (minutes) is much larger than the time scale (seconds) of the receptor–ligand and intracellular signaling reactions, we used a quasi-steady-state approximation where the receptor–ligand and intracellular signaling reactions are assumed to reach local steady state, whereas the downmodulation of receptors evolves adiabatically. The final model is thus solved for the steady-state solution of receptor–ligand and intracellular signaling reactions:

where R and L reflect the number of receptors and ligands, respectively; |\ {C_N}$|⁠, Y, and P reflect the signaling receptor–ligand complexes, the activatory mediators, and the final cytokine product, respectively; and |{k_{{a}}},\ \lambda ,\ \mu ,\ \alpha ,\ {\rm{and}}\ \gamma \ $| reflect activation and deactivation constants (see Supplementary Information). The final model consists of these steady-state solutions together with the evolution equation for receptor downmodulation:

where |\chi $| is the downmodulation rate, and ϵ is a small parameter that represents the slow downmodulation time scale.

The model equations are solved using Matlab program (Mathworks) and are available upon request. Graphs show activity (P) as a function of log(L) for each different receptor affinity, for initial number of receptors of 70,000 or 700,000 per cell, corresponding to low and high CAR expression levels.

The model calculations in Fig. 1B were generated using parameters of ka = 1, γ = 1, α = 100, λ = 1, PT = 10,000, YT = 100, and kp as indicated. For Fig. 1CF, the same parameters were used with kp = 0.05. In Fig. 1E and F, we used χF = 0.35. All other parameters (μ, δ, and χL) were used as indicated in Fig. 1C–F. We note that parameters of χF = 0.35 and χL = 0.005 [χ = 0.35 + 0.005 × (L)] gave similar downmodulation rates to the experimental data (Supplementary Fig. S3E). We used the koff and kD values calculated from the Fab surface plasmon resonance experiments (12). Because the variables L and R are given in terms of numbers (per cell), whereas kD values are in (M) units, there was a need to convert kD to be dimensionless. In the code, we used kD values that give reasonable and comparable outcomes (using the original koff values), thus maintaining the differences between affinities. To achieve this, we multiplied the original kD by a factor of 104. This multiplication was carried on all kD values.

Figure 1.

Gradual construction of CAR activation model reveals that models that include receptor downmodulation can recapitulate the avidity effect. A, Schematic representation of major phenomena observed in our experimental system. B–F, Mathematical models to predict CAR activity. The models are constructed with increasing complexity from simple kinetic proofreading (KP, B) to kinetic proofreading with intracellular IFF signaling motive (KP-IFF, C), to KP-IFF with limited signaling (KPL-IFF, D), to KP-IFF with receptor downmodulation (E), to KPL-IFF with receptor downmodulation (F) models. Top plots schematically represent interaction of receptor (R) and target (L) with affinity constants Koff and Kon and with the kinetic constants that participate in each model. Graphs (bottom plots) demonstrate model prediction for cell activation (P) as a function of increasing Ag density for receptor affinities of 434 nmol/L (black), 35 nmol/L (gold), 16 nmol/L (purple), and 4 nmol/L (blue) for low expression level (70,000 receptors per cell, green) or high expression level (700,000 receptors per cell, red). The contribution of each new complexity in the model is demonstrated by changing the relevant constants. λ, activation constant; μ, intracellular inhibitory signaling constant; δ, nonsignaling receptor constant; χ, receptor downmodulation constant.

Figure 1.

Gradual construction of CAR activation model reveals that models that include receptor downmodulation can recapitulate the avidity effect. A, Schematic representation of major phenomena observed in our experimental system. B–F, Mathematical models to predict CAR activity. The models are constructed with increasing complexity from simple kinetic proofreading (KP, B) to kinetic proofreading with intracellular IFF signaling motive (KP-IFF, C), to KP-IFF with limited signaling (KPL-IFF, D), to KP-IFF with receptor downmodulation (E), to KPL-IFF with receptor downmodulation (F) models. Top plots schematically represent interaction of receptor (R) and target (L) with affinity constants Koff and Kon and with the kinetic constants that participate in each model. Graphs (bottom plots) demonstrate model prediction for cell activation (P) as a function of increasing Ag density for receptor affinities of 434 nmol/L (black), 35 nmol/L (gold), 16 nmol/L (purple), and 4 nmol/L (blue) for low expression level (70,000 receptors per cell, green) or high expression level (700,000 receptors per cell, red). The contribution of each new complexity in the model is demonstrated by changing the relevant constants. λ, activation constant; μ, intracellular inhibitory signaling constant; δ, nonsignaling receptor constant; χ, receptor downmodulation constant.

Close modal

Cytotoxicity assay

CAR T-cell cytotoxicity was measured using conventional [35S] methionine release assay. Target melanoma cell lines 1938, 624.38, and 501A cells were incubated with 12 μCimL [35S] methionine overnight, washed, and seeded at 5 × 103 cells/well coincubated with CAR-positive T cells at 10:1, E:T ratio for 7 hours, and the concentrations of [35S] methionine released from target cells were assayed. The percent-specific cytotoxicity was calculated as follows:

Relative cytotoxicity was determined as: (specific lysis/maximal specific lysis) × 100.

CAR downmodulation assay

CAR downmodulation was detected similarly to previously described methods, with some changes to prevent target cell interference with pMHC monomer staining (44). Briefly, 105 peptide-loaded JY cells were washed and added to each Eppendorf tube at 106 cells/mL in complete T-cell CM. 105 CAR T cells were then added to each Eppendorf tubes at 106 cells/mL, shortly centrifuged, and incubated at 37°C for 5 minutes. Receptor downmodulation was terminated by the addition of a double volume of ice-cold FACS buffer. All following procedures were done at 4°C. The contents of each tube were then removed, pelleted at 3,000 rpm for 1 minute, and resuspended in 100 μL of ice-cold FACS buffer. As JY:T-cell interaction could interfere with MHC staining, JY cells were removed prior to staining using negative selection with Dynabeads utilizing the CELLection pan mouse IgG kit conjugated to anti-CD19 Ab (HIB19), according to the manufacturer's instructions. The cells were stained with excess monomer–MHC–PE for 30 minutes prior to CD8-APC and CD3-PerCP staining for 15 minutes, and then washed and analyzed at the Calibur flow cytometer (BD Biosciences). CAR expression separation was determined posteriori according to GFP signal (which is correlative to CAR expression levels), gating on low, medium, and high GFP signal, as demonstrated in Supplementary Fig. S5D.

Phospho-flow assay

To calibrate a time point sufficient for detection of phosphorylation induction, and the antibodies concentrations to be used, we initially determined the maximal phosphorylation levels of antibodies against p-Zap70, p-LAT, p-Slp76, p-cCbl, and p-Shp2 (BD Biosciences) using flow cytometry. CAR T cells were activated using saturated levels of the anti-CD3 antibody (clone OKT3) for different incubation periods, and cells were stained at different antibody concentrations. Once the setting was calibrated, phospho-flow staining experiments were conducted similarly to previously described methods (16, 33), with the addition of labeled pMHC staining in order to separate by avidity. Briefly, 1.5 × 105 CAR T cells and 1.5 × 105 peptide-loaded JY cells were cultured in serum-starved medium prior to experiment beginning. CAR T cells were then treated for 2 minutes at 37°C with either coculturing with the JY cells, stimulating with OKT3 anti-CD3ϵ antibody (5 μg/mL), or left unstimulated. The reaction was stopped immediately by addition of an equal volume of ice-cold FACS buffer containing pMHC monomers (10 μg/mL) and phosphatase-inhibitor cocktail (PhosSTOP, Sigma), to allow surface staining of CARs, for 5 minutes on ice. Following washing, cells were fixed and permeabilized according to the manufacturer's protocol (BD Phosflow) and stained with streptavidin-BV421 (BioLegend), anti-CD8 antibodies, and BD Phosflow antibodies, as described in text. Finally, cells were washed and analyzed by an LSR II flow cytometer (BD Biosciences). CAR T cells were gated by size and granulation (FCS/side scatter), GFP-positive, pMHC-positive, and CD8-positive cells. CAR expression separation was determined posteriori according to pMHC staining signal, gating on low, medium, and high pMHC staining signal, as demonstrated in Supplementary Fig. S5C. Relative phosphorylation for each phospho-protein was determined as [(MFI – minimal MFI) / (maximal MFI – minimal MFI)] × 100, whereas minimal and maximal MFIs were the lowest and highest MFI values for each experiment.

Antigen trogocytosis assays

Trogocytosis flow cytometry was analyzed by measuring antigen transfer using antigen-specific staining. CAR T cells (5 × 104) that were treated with 0.3, 1, or 5 μmol/L CytD inhibitor or with DMSO cocultured with melanoma cells or loaded JY cells at 1:1 E:T ratio in V-shaped 96-well plates. Unless specified differently, cells were incubated for 15 minutes at 37°C. Antigen transfer was measured by staining of cells with the high-affinity TCR-like Abs that recognize specifically the Tyr-HLA-A2 antigen, followed by labeling with secondary antibodies and measured by flow cytometry. Antigen transfer was determined by calculating the MFI of the effector CAR+ cells (by gating on GFP+ cells). Initial tests were carried on in order to compare whether the target cell presence can alter the staining of the antigen on the effector cells.

Trogocytosis imaging was analyzed using antigen-specific staining. CAR T cells (5 × 105) that were pretreated with 1 or 5 μmol/L CytD inhibitor or with DMSO for 45 minutes were washed and cocultured with loaded JY cells at 1:1 E:T ratio in V-shaped 96-well plates and incubated for 15 minutes at 37°C. Antigen trogocytosis was measured by staining cells with the high-affinity TCR-like Abs that recognize specifically the Tyr-HLA-A2 antigen, followed by labeling with secondary antibodies and anti-CD8 antibody for surface staining. Samples were seeded with 1% agar-PBS in 15μ-Slide VI 0.4 chambers (IBIDI) and imaging at confocal microscope, and z-stacks were taken at optimal imaging parameters with a LSM710 confocal microscope with 40× water immersion objective (Carl Zeiss Microimaging). IMARIS software was used to generate the figures. Antigen trogocytosis was determined by calculating the MFI on the surface of effector CAR+ T cells, determined by CD8+ and GFP+ cells.

Quantification and statistical analysis

FACS data were analyzed using FCS Express 5 (De Novo). MFIs were calculated after subtraction of the background staining calculated from isotype-control staining (for primary conjugated antibodies) or from secondary antibody staining (for nonconjugated primary antibodies). Reactive-MFI was measured in a similar fashion on positive cells, with a threshold of at least 3% positive cells. In case of less than 3%, the reactive-MFI was determined as zero.

A standard principal component analysis (PCA) analysis was performed using the Perseus software (45). The MFI data matrix was arranged such that each CAR T-cell line (e.g., CAR T of 16 nmol/L affinity with high expression) was represented in rows and the averages of all markers p-Zap70, p-LAT, p-Slp76, p-cCbl, and p-Shp2, or on four markers excluding p-LAT. Following replacement of negative values by zeros, the matrices were log transformed, normalized, and then analyzed by PCA.

All statistical analyses of the described measurements (MFIs, reactive-MFIs, percent-reactive cells, receptor downmodulation, and relative killing) were performed using The R Project for Statistical Computing (46). P values were corrected for FDR using the Benjamini–Hochberg procedure (47). According to the correction, for the ICS analysis, P values under 0.021 were considered significant; for the phospho-flow analysis, P values under 0.023 were considered significant; for the downmodulation analysis, P values under 0.023 were considered significant. Significant values are represented with stars as follows: *, 0.05–0.01; **, 0.01–0.001; ***, <0.001 adjusted values. A three-way ANOVA (CAR expression × CAR affinity × antigen density) was performed on MFI (phosphoflow assay), reactive-MFI (ICS assay), or percent-reactive cells (ICS assay) of the CAR T cells, using the aov function. For two-way ANOVA calculation of the variance explained by affinity or CAR expression, we divided each effect sum of squares by the total sum of squares.

Statistical analysis of cytotoxicity was performed using The R Project for Statistical Computing. A two-way ANOVA (CAR affinity × tumor cell line) was performed using the aov function, followed by Tukey Honest Significant Differences (TukeyHSD).

Statistical analysis of receptor downmodulation at coincubation with 100 μmol/L peptide-loaded JY cells was performed using The R Project for Statistical Computing. A one-way ANOVA (drug treatment) was performed on MFI, using the aov function, followed by TukeyHSD with FDR correction using the Benjamini–Hochberg procedure.

Statistical analysis of ICS following drug treatment (reactive-MFI and percent-reactive) was performed using The R Project for Statistical Computing. Two-way ANOVAs (drug treatment x antigen density) were performed using the aov function, followed by TukeyHSD with FDR correction using the Benjamini–Hochberg procedure.

Mathematical modeling of CAR T-cell response

We have previously generated an experimental system in which CAR affinity, avidity and target cell's antigen density can be manipulated. This system revealed several key features regarding the effect of affinity, avidity, and antigen density on CAR T-cell function (Fig. 1A):

  • A bell-shaped response as a function of antigen density.

  • Different affinities had similar antigen densities inducing peak response.

  • Peak response was different between affinities, with mid-affinities yielding the highest responses.

  • High avidity induced a higher response.

  • High avidity had a wider window of maximal response.

To explain these findings from a mechanistic perspective, we mathematically modeled T-cell activation. Although the T-cell response is an intricate network of many elements, it can be phenotypically modeled through fewer parameters representing the complex signaling cascade (13). Based on previous approaches to formulate a mathematical model of native TCR-induced activation and response (13, 18), we tested several models while gradually increasing the model's complexity. We started with a kinetic proofreading model. This model considers the antigen (ligand, L) and CAR (receptor, R) binding kinetic constants (kon and koff). The complex that is formed upon binding (C0), however, does not immediately trigger signaling, and only after sufficient binding, the complex becomes a signaling complex (CN; kp is the kinetic proofreading constant) that induces a downstream signaling cascade and consequentially cytokine production (P). Higher kp reduces the difference between sensitivities of the various affinities (Fig. 1B). This model however does not predict a ligand-dependent bell-shaped curve. We therefore tested a model incorporating also an intracellular incoherent-feedforward (IFF) motif (Fig. 1C), similarly to previous works (14, 17), to reflect the inhibitory intracellular signaling molecules (such as Shp1 and Shp2 phosphatases) that can induce ligand-dependent inhibition (14, 16). As expected, increasing the inhibition constant (μ) produced lower responses at high ligand concentrations. Yet, this model predicts highest sensitivity for the highest affinity receptor (4 nmol/L affinity, blue lines in Fig. 1). High-affinity–induced inhibition was described in TCRs. High-affinity interactions are thought to prevent continuous signaling due to slower dissociation constants (13). This assumption, the limited signaling model, assumes a state of a nonsignaling complex (CN+1, formed by a δ constant from CN; Fig. 1D). Incorporating this element into the kinetic proofreading with IFF model (KPL-IFF) predicts reduced responses for high affinities and for high antigen densities as well. Nonetheless, the model was negated, as the peak amplitude of cells with high receptor expression (700,000 receptors per cell) is similar to cells with low expression level (70,000 receptors per cell), consistent with previous models that compared receptor expression levels (17). Equally, none of the above-described models displayed different magnitudes of response as a function of receptor expression levels. We also note that all the described models show complete symmetry between the receptor and ligand. Because the numbers of receptors per effector cell are several orders of magnitude higher than the number of antigens (ligands) per target cell, it stands to reason that further increasing receptor number cannot increase activity, as all ligands are already fully occupied. We therefore examined a model without R and L symmetry, in which T-cell responses were more dependent on avidity. Previous works have shown that both TCRs and CARs are internalized and degraded following target cell encounter, thus regulating T-cell activation (18–21). We created a model that incorporates a slow downmodulation of the receptor by a constant χ, which increases as a function of the number of ligands. This model predicts higher response when receptor expression increases. Surprisingly, due to the ligand-induced receptor downmodulation, the response at high ligand concentrations is reduced, thus creating a bell-shaped curve, even without the IFF motif (Fig. 1E). Moreover, this model now predicts a similar ligand concentration that induces peak response across all different affinities, and induces a wider window of high response in high avidity cells, corresponding to our experimental data. Integrating all the above-described elements (proofreading, limited signaling, IFF, and receptor downmodulation), the model predicts a bell-shaped response as a function of antigen density, and as a function of affinity, with similar antigen densities between affinities that induce maximal responses, where high avidity contributes to a higher and wider maximal response (Fig. 1F). This model describes best our observed data. We note that in both downmodulation-incorporating models (Fig. 1E and F), the inhibitory part of IFF had negligible effect on the outcome.

Antigen-dependent proximal signaling is correlated with avidity and basal level of activity

Our model predicted the antigen-dependent bell-shaped curve to be mostly dependent on CAR downmodulation, whereas works on TCR signaling attribute this feature mainly to intracellular inhibitory molecules, such as Shp1 and Shp2 (16). To address this discrepancy, we examined both processes, looking at the differential phosphorylation pattern of known proximal TCR signaling molecules, and at ligand-dependent receptor downmodulation. First, we assessed the level of phosphorylation of three activatory signaling molecules (p-Zap70, p-LAT, and p-Slp76) and two inhibitory signaling molecules (p-cCbl and p-Shp2) using flow cytometry. We incubated CAR T cells of different affinities (kDs of 4, 16, 35, and 434 nmol/L) with peptide-loaded APC target cells for 2 minutes, and analyzed the relative phosphorylation of each molecule (i.e., normalized by the maximal phosphorylation). We used unstimulated T cells for basal activation measurements, and anti-CD3 Ab (OKT3) for CAR-independent activation measurements. We post hoc separated the cells by expression level, gating according to CAR surface staining. All five phosphorylated molecules showed an antigen-dependent increase in phosphorylation, reaching highest phosphorylation at the highest antigen density. The averages of relative phosphorylation for representative signaling molecules are shown graphically in Fig. 2 arranged either by expression level in each individual CAR affinity (Fig. 2A and C) or by their avidity (Fig. 2B and D; see Supplementary Fig. S1 for the remaining signaling molecules and separated graphs for each CAR affinity, and Supplementary Table S1 for statistical analysis). The phosphorylation seemed to depend mainly on receptor avidity, and less on receptor affinity, as seen when results are arranged by avidity (Fig. 2B and D). Higher avidity cells displayed higher phosphorylation level (including higher phosphorylation to nonspecific antigens, as seen for unloaded target cells).

Figure 2.

High avidity CAR T cells display stronger proximal signaling. A–D, Contour plots showing phosphorylation levels of representative downstream proximal signaling components in CAR T cells of varying affinity and CAR expression levels that were incubated with target APCs of different Ag densities. The dependency of the activation marker Zap70 (A and B) and inhibition marker Shp2 (C and D) on Ag density (y axis) and CAR expression and CAR affinity (x axis; A and C) or on avidity (x axis; B and D) is indicated by color. Values represent averages of n = 4 independent experiments for each CAR T-cell population. E–H, Average phosphorylation levels + SE of all the activatory molecules (blue lines) compared with the average phosphorylation of all the inhibitory molecules (orange lines) in each CAR affinity.

Figure 2.

High avidity CAR T cells display stronger proximal signaling. A–D, Contour plots showing phosphorylation levels of representative downstream proximal signaling components in CAR T cells of varying affinity and CAR expression levels that were incubated with target APCs of different Ag densities. The dependency of the activation marker Zap70 (A and B) and inhibition marker Shp2 (C and D) on Ag density (y axis) and CAR expression and CAR affinity (x axis; A and C) or on avidity (x axis; B and D) is indicated by color. Values represent averages of n = 4 independent experiments for each CAR T-cell population. E–H, Average phosphorylation levels + SE of all the activatory molecules (blue lines) compared with the average phosphorylation of all the inhibitory molecules (orange lines) in each CAR affinity.

Close modal

Our previous work on antigen-induced inhibition in TCRs revealed that activatory signaling molecules respond sharply at low and medium antigen densities, whereas inhibitory signaling molecules respond gradually, reaching maximal phosphorylation only at high antigen densities (16), thus signal summation of these modules creates a bell-shaped response of the T cells. However, when we compared activation of the inhibitory and the activatory molecules, both modules responded gradually to antigens, reaching maximal phosphorylation only at the high antigen density, and with no detectable gap between the two modules (Fig. 2EH). Although we clearly see activation of the inhibitory molecules (reinforcing the existence of an IFF motive), we cannot attribute the antigen-dependent bell-shaped curve to a gap between the activatory and inhibitory modules.

Next, we applied a systematical analysis to assess the contribution of affinity and avidity to proximal signaling responses, by applying a two-way ANOVA for each antigen density, and analyzed the percentage of variance attributed to each effect (the effect's sum of squares) out of the total variance (the total sum of squares), thereby denoting the percentage of variance they could explain for each antigen density. As observed above, apart from the p-LAT molecule, the level of phosphorylation was strongly dependent on receptor avidity (Fig. 3AC). p-LAT showed high dependence on avidity only at high antigen densities. We then applied PCA on all five phosphorylated molecules over all affinities and avidities (Fig. 3D). The first principal component resembles avidity and corresponds to more than 70% of the variance. The second component seems to reflect the different affinities. As the phosphorylation rates of p-LAT displayed different trends from all other four molecules (Supplementary Fig. S1G), we repeated the PCA, this time excluding the p-LAT molecule (Supplementary Fig. S2F). Now the avidity component explained more than 80% of variance. The first principal component strongly correlated with CAR expression levels (Fig. 3E).

Figure 3.

Variance in proximal signaling levels is affected mainly by CAR expression level. A–C, The effect of affinity and expression level on phosphorylation levels of downstream proximal signaling components was analyzed by a two-way ANOVA for each antigen density. The percentage of response variance that could be explained by affinity (A), expression level (B), or their interaction (C) is calculated by dividing the variance attributed to each effect (the effect's sum of squares) out of the total variance (the total sum of squares). D, PCA of phosphorylation levels over all activation markers and all CAR affinities and expression levels (“All” represents the whole CAR T-cell population, without post hoc separation). PCA indicates that the main component reflects changes in expression levels (different colors), whereas the second and more minor component reflects the different CAR affinities (different shapes). E, Principal component 1 values (of PCA performed on p-Zap70, p-Slp76, p-cCbl, and p-Shp2, excluding p-LAT), which describes 86.1% of variance, is correlative to CAR expression levels of the cells.

Figure 3.

Variance in proximal signaling levels is affected mainly by CAR expression level. A–C, The effect of affinity and expression level on phosphorylation levels of downstream proximal signaling components was analyzed by a two-way ANOVA for each antigen density. The percentage of response variance that could be explained by affinity (A), expression level (B), or their interaction (C) is calculated by dividing the variance attributed to each effect (the effect's sum of squares) out of the total variance (the total sum of squares). D, PCA of phosphorylation levels over all activation markers and all CAR affinities and expression levels (“All” represents the whole CAR T-cell population, without post hoc separation). PCA indicates that the main component reflects changes in expression levels (different colors), whereas the second and more minor component reflects the different CAR affinities (different shapes). E, Principal component 1 values (of PCA performed on p-Zap70, p-Slp76, p-cCbl, and p-Shp2, excluding p-LAT), which describes 86.1% of variance, is correlative to CAR expression levels of the cells.

Close modal

Whereas CARs are activated solely by their antigen targets, the native TCR requires the recruitment of additional coreceptors and the formation of the TCR complex, including the various CD3 subunits (21). We were therefore surprised that the OKT3 antibody, which activates TCR complexes and not CARs, induced discrepant levels of phosphorylation between different CAR affinities and avidities (Supplementary Fig. S2A–S2E). The deviations across different avidities and affinities to OKT3 activation resembled differences at the basal activation state. As basal TCR or CAR activity can tune cell function and sensitivity toward its cognate antigen (8, 22, 23), we compared the basal level of phosphorylation with the PCA's first principal component that describes most of the variance in phosphorylation, and detected a strong correlation to the basal level of p-Zap70, p-Slp76, and p-cCbl phosphorylation levels (Supplementary Fig. S2G). This suggests that the differences in phosphorylation following an antigen encounter can be predicted by the basal phosphorylation level prior to that encounter. When comparing phosphorylation levels between activated and resting cells, high Pearson correlations are observed (Supplementary Fig. S2H). Because the basal level of phosphorylation can be affected by avidity (24), it is yet to be concluded which parameter is causative for the differential phosphorylation levels. In aggregate, we demonstrated a rapid antigen-dependent activation of both inhibitory and activatory modules, which is correlated with basal activation levels and avidity; however, it could not explain signal summation that results in nonmonotonous behavior.

CAR downmodulation is correlated with avidity, but not affinity

Next, we set out to characterize receptor downmodulation occurring immediately after receptor and ligand contact. CAR T cells that were shortly incubated (5 minutes) with peptide-loaded APCs target were assessed for the surface expression of CD3, CD8, and the CAR. Surprisingly, we detected a significant CAR downmodulation (i.e., reduction in CAR surface staining) even after this short incubation period (Fig. 4A; Supplementary Fig. S3D). The amount of CAR downmodulation was higher when initial CAR expression was higher. All CARs, regardless of affinity, showed minor CAR downmodulation in response to nonspecific antigens (unloaded APCs). Smaller changes were detected in both CD8 and CD3 coreceptors (Supplementary Fig. S3A and S3B). As the CAR expression is proportional to GFP expression (Supplementary Fig. S3C), we post hoc gated each CAR population into three CAR expression levels according to GFP expression level (Fig. 4B), consequently highlighting the dependence of CAR downmodulation on initial CAR expression levels. In fact, the amount of CAR downmodulation is proportional to the initial CAR expression (Fig. 4C). In other words, the fraction of downmodulated CARs depends solely on antigen density, whereas the total amount of downmodulated CARs depends also on the initial number of receptors. CAR affinity had no evident effect over receptor downmodulation. CAR downmodulation occurred rapidly, and no further downmodulation was observed at longer incubation times (Supplementary Fig. S3D). Taken together, we examined and detected both receptor downmodulation and intracellular proximal signaling processes. Both processes occurred rapidly and were dependent on CAR expression levels, and are plausible mechanisms that can result in antigen-dependent nonmonotonic behavior.

Figure 4.

CAR downmodulation is associated with avidity. A, Surface expression of CARs per cell is downmodulated as Ag density increases. Values represent averages + SE of 4 ≤ n ≤ 6 independent experiments. B–D, Surface expression of CARs after post hoc separation by avidity. E, Reduction in CAR surface expression levels (y axis) at different Ag densities is highly correlated with initial CAR surface expression (x axis).

Figure 4.

CAR downmodulation is associated with avidity. A, Surface expression of CARs per cell is downmodulated as Ag density increases. Values represent averages + SE of 4 ≤ n ≤ 6 independent experiments. B–D, Surface expression of CARs after post hoc separation by avidity. E, Reduction in CAR surface expression levels (y axis) at different Ag densities is highly correlated with initial CAR surface expression (x axis).

Close modal

Actin-dependent receptor downmodulation, and not Shp1/Shp2 inhibition, mostly induces hyporesponsiveness

Finally, to determine which mechanism is associated with the hyporesponsiveness at high antigen densities, we compared functional avidity of CAR T cells following drug-induced Shp1/Shp2 phosphatase inhibition (using SSG; ref. 25) or drug-induced internalization inhibition (using the actin polymerization inhibitor CytD; ref. 26). As actin depolymerization can interfere with immunologic synapse formation and T-cell activation (27, 28), we worked with the lowest CytD concentrations which could detectably inhibit antigen-induced CAR downmodulation (Fig. 5A; Supplementary Fig. S4A; Supplementary Table S2). We examined CAR downmodulation of the 35 nmol/L CAR following CytD treatment, or treatment with 10 or 20 μg/mL SSG, concentrations that inhibit both Shp1 and Shp2 activities (25). The CAR surface expression of treated cells was compared with surface expression of the unstimulated (i.e., without target cells encounter) control cells (Supplementary Fig. S4A), or with each treatment's unstimulated surface expression (Fig. 5A). We noticed that all treatments reduced CAR expression in the basal unstimulated state, thus lowering initial avidity of the CAR T cells; however, cells treated with CytD had lowered antigen-induced CAR downmodulation, in a drug dose–dependent manner. Alongside downmodulation, trogocytosis is a process that depends on actin polymerization and even on receptor internalization, and therefore can also be the cause of the hypofunctionality at high antigen densities due to antigen removal or even fratricide. CAR T-cell fratricide could not be detected using reciprocal killing assay. Nonetheless, using antigen-specific staining, we observed trogocytosis that occurred rapidly following encounter with peptide-loaded cells (Fig 5B–D; Supplementary Fig. S4A–S4C; Supplementary Tables S5 and S6). However, this phenomenon is evident at the higher antigen densities, and was reduced following CytD treatment (Fig 5B–D), but not following SSG treatment (Supplementary Fig. S4C).

Figure 5.

Actin-dependent receptor downmodulation regulates CAR T-cell responsiveness. A–D, 35 nmol/L CAR T-cells that were pretreated with either SSG, CytD, or control (DMSO) drugs and incubated with target APCs of different Ag densities were assayed for CAR surface expression (A) or antigen expression on effector cells (for antigen trogocytosis detection, B) by flow cytometry. Values represent MFI averages + SE of 4 ≤ n ≤ 8 independent experiments. C and D, Antigen trogocytosis following CytD treatment was measured by confocal microscopy. Values represent average intensity mean + S.E of 7 ≤ n ≤ 10 images. E–G, Intracellular expressions of the cytokines IFNγ (E), TNFα (F), and IL2 (G) were measured on CAR T-cells pretreated with the indicated inhibitors drugs. Values represent reactive-MFIs averages + SE of 4 ≤ n ≤ 8 independent experiments. H–J, CytD inhibits melanoma-induced CAR downmodulation and enhances antitumor activity. T cells expressing the 4 nmol/L CAR that had been pretreated with CytD were incubated with target melanoma cell lines and were assayed for downmodulation (H), trogocytosis (I), and cytolytic activity (J). Values represent MFI averages + SE of n = 7, 6 ≤ n ≤ 8 and n = 4 independent experiments, respectively. *, 0.05–0.01; **, <0.01–0.001; ***, <0.001 adjusted values.

Figure 5.

Actin-dependent receptor downmodulation regulates CAR T-cell responsiveness. A–D, 35 nmol/L CAR T-cells that were pretreated with either SSG, CytD, or control (DMSO) drugs and incubated with target APCs of different Ag densities were assayed for CAR surface expression (A) or antigen expression on effector cells (for antigen trogocytosis detection, B) by flow cytometry. Values represent MFI averages + SE of 4 ≤ n ≤ 8 independent experiments. C and D, Antigen trogocytosis following CytD treatment was measured by confocal microscopy. Values represent average intensity mean + S.E of 7 ≤ n ≤ 10 images. E–G, Intracellular expressions of the cytokines IFNγ (E), TNFα (F), and IL2 (G) were measured on CAR T-cells pretreated with the indicated inhibitors drugs. Values represent reactive-MFIs averages + SE of 4 ≤ n ≤ 8 independent experiments. H–J, CytD inhibits melanoma-induced CAR downmodulation and enhances antitumor activity. T cells expressing the 4 nmol/L CAR that had been pretreated with CytD were incubated with target melanoma cell lines and were assayed for downmodulation (H), trogocytosis (I), and cytolytic activity (J). Values represent MFI averages + SE of n = 7, 6 ≤ n ≤ 8 and n = 4 independent experiments, respectively. *, 0.05–0.01; **, <0.01–0.001; ***, <0.001 adjusted values.

Close modal

Next, we investigated the antigen-induced nonmonotonous curve following treatment with these drugs, by measuring production of the IFNγ, TNFα, and IL2 cytokines (Fig. 5EG; see Supplementary Fig. S4B–S4D for percent-reactive cells and Supplementary Table S3 for statistics). Surprisingly, we detected only minor changes following SSG treatment, however with a trend of increased response at high antigen densities. CytD-treated cells responded differently for the two close concentrations. CytD treatment (1 μmol/L) leads to higher production of the three cytokines, resembling the effect of increasing CAR expression levels, whereas 5 μmol/L CytD treatment showed similar or even lower cytokine production, along with lower percentages of responding cells (Supplementary Fig. S4D–S4F). This might be attributed to interruption of synapse formation. These data suggest that reducing internalization and trogocytosis processes and increasing CAR expression level have a stronger effect than interfering with the intracellular inhibitory signaling module. We speculate that there is a narrow window of CytD concentrations that can interfere with trogocytosis and internalization feedback mechanisms without significantly inhibiting synapse formation. Nonetheless, this treatment could not prevent a bell-shaped response, which corresponds to the observation that CAR downmodulation was still evident at this CytD concentration.

Finally, we corroborated the effect of actin polymerization inhibition on target lysis of tumor cell lines. We compared responses of high-affinity CAR T cells treated with CytD toward two Tyr-HLA-A2–positive and one Tyr-HLA-A2–negative melanoma cell lines (501A, 624.38, and 1938, respectively, see Supplementary Fig S4G). Receptor downmodulation was evident on antigen-presenting melanoma cell lines (Fig. 5H), whereas trogocytosis was hardly evident (Fig. 5I; Supplementary Fig. S4B and S4C). Congruently, CytD interfered with CAR downmodulation but not with trogocytosis. Accordingly, we measured CytD effect on antitumor activity, as it interfered only with one process, thus allowing to distinguish between receptor downmodulation and trogocytosis effects. Treatment with 1 or 0.3 μmol/L CytD that prevented CAR downmodulation could enhance antitumor response (Fig. 5J; Supplementary Table S5). Altogether, reducing receptor downmodulation is equivalent to increasing avidity (as reported by us previously, ref. 12) and to increasing CAR T-cell response.

Mechanistic understanding of CAR T-cell regulation and activity is currently limited, despite being clinically approved and even expedited for additional clinical treatments. Adjusting CAR T cells design by the biophysical properties of binding can furthermore lead to optimal T-cell responsiveness. We previously described the binding properties that lead to differential T-cell responses. Here, impelled by a theoretical model, we investigated the underlying mechanism of CAR T cells response. We first built a novel model to describe the dependence of CAR T-cell activity on antigen density, CAR expression, and CAR affinity. Surprisingly, this model predicted that CAR downmodulation is more dominant in shaping the antigen-dependent nonmonotonous behavior. We experimentally corroborated this hypothesis, using drug-induced Shp1/Shp2 inhibition or actin polymerization inhibition, and demonstrated that reducing CAR internalization could increase cytokine production. These observations stress the fundamental regulatory role of CAR internalization, which can therefore be targeted for manipulating CAR T-cell response.

There is an increased interest in manipulating CAR affinity in order to optimize antitumor response. Recently, works aimed at understanding the dependency of functional avidity on affinity described different outcomes for increased receptor affinity, ranging from (i) an inhibitory outcome and noneffective effect (7–9, 11), (ii) a neither beneficial nor disadvantageous response (6, 29), and (iii) an increased response (10, 30). We previously measured CAR T-cell activity and documented that CARs of medium affinities outperformed the high-affinity CAR (12), supporting the first group of studies. Nevertheless, our new proposed model can predict the responses observed by groups who documented increased affinity effect or no additional beneficial effect (6, 10, 29). Hence, the effect of increased affinity depends on the avidity and antigen density, which can alter the affinity effect from beneficial to disadvantageous. This strengthens the importance of considering all of these binding properties when designing a CAR for treatment.

Manipulation of CAR T-cell regulatory pathways can serve to tune their activity. We noted that high avidity cells have a wider window of antigen densities that induce maximal response, as they are less affected by the internalization inhibitory process. CAR downmodulation has been observed and suggested to inhibit CAR T-cell responses to secondary challenges (18, 31). We tested here a drug-based manipulation of receptor expression levels, which resulted in improved responses. Likewise, genetically controlling CAR expression showed the dependence of CAR T cell exhaustion and efficacy on both the basal and dynamic CAR expression levels (18). Altogether, accumulating evidence demonstrates the eminence of receptor expression levels on CAR T-cell activity and can be considered an effective candidate for manipulating and adjusting CAR T cell responses.

Based upon validated models describing mathematically the T-cell activation through its TCR, we modeled CAR T cell activation. Similar regulatory mechanisms could explain T-cell activation, supported by the observation that similar antigen densities between CAR and TCR targets lead to maximal response. Correspondingly, a recent study (17) compared CAR and TCR activities and described their antigen-dependent nonmonotonic response. Through modeling TCR and CAR activation, reduced activity at high antigen densities was explained by a negative feedback mechanism in the form of an intracellular IFF motive. Contrarily, we used here a system that manipulated and quantified several binding properties of CAR binding, and we consequently inferred that these models were insufficient to fully describe CAR activation because they predict similar peak responses to different CAR expression levels. We resolved this by introducing an avidity-dependent regulatory mechanism. The model demonstrated that receptor downmodulation regulation is more dominant in shaping CAR T-cell activation, and could furthermore describe an antigen density range that induces peak response across all CAR affinities and avidities, in accordance with our experimental data.

The observation that antigen sensitivity and hypofunctionality at high antigen densities were affected by CAR avidity encouraged us to search for the mechanism that regulates antigen responses, a mechanism that would depend on CAR avidity. Our study described two regulating mechanisms that are affected by avidity—receptor downmodulation and proximal signaling. Proximal inhibitory signaling is known to inhibit T-cell function due to high antigen densities or high TCR affinities (16, 32, 33). Although CARs are built using intracellular signaling moieties of the TCR complex and are thought to activate similar downstream signaling, the proximal signaling of CAR T cells remains understudied. Compared with TCRs, CARs display rapid kinetics of proximal activatory signaling molecules, along with fast actin accumulation (34). Different signaling kinetics are observed even when comparing CARs constructed from different signaling domains (35). Our study further characterized CAR activation and described the involvement of several inhibitory and activatory signaling molecules. Moreover, we observed correlation between avidity, the basal proximal signaling activity, and the amount of proximal signaling following stimuli. Basal signaling is known to correlate with T-cell sensitivity and magnitude of response (35, 36). However, increasing basal phosphorylation is not sufficient to induce full T-cell response. For example, inhibition of the Lck-negative regulator Csk resulted in higher basal proximal signaling even without TCR engagement, yet recapitulation of a full TCR activation was accomplished only after CytD drug treatment, indicating the essential role of actin remodeling for T-cell stimulation (28).

We recognize that proximal signaling and receptor internalization processes are not separated, but in fact interconnected. Clustering of proximal signaling molecules such as PLCγ, LAT, and Slp76 in the immunologic synapse depends on actin polymerization (37, 38), and on the other hand, induction of proximal signaling can downregulate the number of receptors (39). Nonetheless, using the CytD and SSG drugs, we could see reduced CAR downmodulation only following CytD treatment and not following SSG treatment. This implies that we can find drugs that can affect, at least in part, one mechanism without altering the other. Induction of receptor downmodulation occurs through its ubiquitination via cCbl, and not through Shp1/Shp2 (40, 41); therefore, the SSG drug should not interfere with receptor downmodulation. Another possibility to differentiate between proximal inhibitory signaling and receptor downmodulation is to target receptor degradation. It was demonstrated in TCRs that internalization occurs constantly, but only internalized and activated TCRs are degraded, resulting in receptor downmodulation (19). When we compared CAR-T cell responses following CytD and SSG treatments, we observed an increased response in CytD-treated cells alone. Peak response of treated cells was higher, with wider window of maximal response, similar to the differences seen for increasing avidity. We suggest that actin rearrangement, and specifically receptor internalization, is the mechanism underlying the distinct responses between different avidities, as predicted from our mathematical model.

We acknowledge that both receptor downmodulation and antigen trogocytosis rely on a similar molecular basis, which depends on actin rearrangements, and therefore the distinction of the two processes can never be completely clear. Hamieh and colleagues described the association between antigen trogocytosis, receptor downmodulation, T:T interaction and activation, CAR T-cell exhaustion, reduction of the tumor's antigen expression, and the consequent tumor escape (48). Nevertheless, we observed very low levels of trogocytosis after incubation with solid tumor cells. Likewise, low CytD concentrations impinged on receptor downmodulation and not antigen trogocytosis, and could increase CAR T-cell antitumor activity. Nevertheless, both trogocytosis and receptor downmodulation processes provide valid mechanisms that might regulate CAR T-cell response. The clinical relevance of trogocytosis in CAR T-cell activity, and its distinction from receptor internalization should be further characterized.

Collectively, we propose a new model to assess the outcome of CAR T cells, as a function of receptor affinity, receptor expression levels, and antigen density. This novel knowledge could impinge upon adoptive cell therapy therapies, as we might better control CAR T-cell activity and tumor eradication, for example, by regulating receptor downmodulation.

Y. Reiter reports other from Adicet Bio outside the submitted work. G. Denkberg reports personal fees from Adicet Bio Israel during the conduct of the study and outside the submitted work. S. Shen-Orr reports personal fees and other from CytoReason outside the submitted work. No disclosures were reported by the other authors.

R. Greenman: Conceptualization, validation, investigation, visualization, methodology, writing–original draft. Y. Pizem: Validation, investigation, visualization, methodology, writing–original draft. M. Haus-Cohen: Validation, investigation, methodology. G. Horev: Software, methodology. G. Denkberg: Investigation, visualization, methodology. S. Shen-Orr: Software, methodology. J. Rubinstein: Resources, software, methodology. Y. Reiter: Conceptualization, supervision, writing–original draft.

The authors are grateful to Professor Zelig Eshhar (Weizmann Institute of Science) for the gift of Phoenix-Ampho cells and the pBullet vector. Dr. Yair Lewis and Professor Izhak Kehat kindly provided the CytD reagent. The authors also thank Drs. Stewart Abbot, Daulet (Dau) Satpayev (Adicet Bio), and Arie Admon (Technion) for helpful comments on the article.

This work was supported by a grant from the Israel Science Foundation (ISF-461/15) to Y. Reiter.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

1.
Wang
M
,
Yin
B
,
Wang
HY
,
Wang
R-F
. 
Current advances in T-cell-based cancer immunotherapy
.
Immunotherapy
2014
;
6
:
1265
78
.
2.
Hege
KM
,
Bergsland
EK
,
Fisher
GA
,
Nemunaitis
JJ
,
Warren
RS
,
McArthur
JG
, et al
Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer
.
J Immunother Cancer
2017
;
5
:
22
.
3.
Brown
CE
,
Alizadeh
D
,
Starr
R
,
Weng
L
,
Wagner
JR
,
Naranjo
A
, et al
Regression of glioblastoma after chimeric antigen receptor T-cell therapy
.
N Engl J Med
2016
;
375
:
2561
9
.
4.
Maus
MV
,
Grupp
SA
,
Porter
DL
,
June
CH
. 
Antibody-modified T cells: CARs take the front seat for hematologic malignancies
.
Blood
2014
;
123
:
2625
35
.
5.
Huehls
AM
,
Coupet
TA
,
Sentman
CL
. 
Bispecific T-cell engagers for cancer immunotherapy
.
Immunol Cell Biol
2015
;
93
:
290
6
.
6.
Chmielewski
M
,
Hombach
A
,
Heuser
C
,
Adams
GP
,
Abken
H
. 
T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity
.
J Immunol
2004
;
173
:
7647
53
.
7.
Oren
R
,
Hod-Marco
M
,
Haus-Cohen
M
,
Thomas
S
,
Blat
D
,
Duvshani
N
, et al
Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody–based chimeric antigen receptors indicates affinity/avidity thresholds
.
J Immunol
2014
;
193
:
5733
43
.
8.
Long
AH
,
Haso
WM
,
Shern
JF
,
Wanhainen
KM
,
Murgai
M
,
Ingaramo
M
, et al
4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors
.
Nat Med
2015
;
21
:
581
90
.
9.
Ghorashian
S
,
Kramer
AM
,
Onuoha
S
,
Wright
G
,
Bartram
J
,
Richardson
R
, et al
Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR
.
Nat Med
2019
;
25
:
1408
14
.
10.
Caruso
HG
,
Hurton
LV
,
Najjar
A
,
Rushworth
D
,
Ang
S
,
Olivares
S
, 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
.
11.
Liu
X
,
Jiang
S
,
Fang
C
,
Yang
S
,
Olalere
D
,
Pequignot
EC
, 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
.
12.
Greenman
R
,
Pizem
Y
,
Haus-Cohen
M
,
Goor
A
,
Horev
G
,
Denkberg
G
, et al
Shaping functional avidity of CAR-T cells: affinity, avidity and antigen density that regulate response
.
Mol Caner Ther
2021
.
13.
Lever
M
,
Maini
PK
,
Merwe
PA
,
van der Dushek
O
. 
Phenotypic models of T cell activation
.
Nat Rev Immunol
2014
;
14
:
619
.
14.
Lever
M
,
Lim
H-S
,
Kruger
P
,
Nguyen
J
,
Trendel
N
,
Abu-Shah
E
, et al
Architecture of a minimal signaling pathway explains the T-cell response to a 1 million-fold variation in antigen affinity and dose
.
Proc Natl Acad Sci U S A.
2016
;
113
:
E6630
8
.
15.
Coombs
D
,
Kalergis
AM
,
Nathenson
SG
,
Wofsy
C
,
Goldstein
B
. 
Activated TCRs remain marked for internalization after dissociation from pMHC
.
Nat Immunol
2002
;
3
:
1109
.
16.
Wolchinsky
R
,
Hod-Marco
M
,
Oved
K
,
Shen-Orr
S
,
Bendall
SC
,
Nolan
GP
, et al
Antigen-dependent integration of opposing proximal TCR-signaling cascades determines the functional fate of T lymphocytes
.
J Immunol
2014
;
192
:
2109
19
.
17.
Harris
DT
,
Hager
MV
,
Smith
SN
,
Cai
Q
,
Stone
JD
,
Kruger
P
, et al
Comparison of T cell activities mediated by human TCRs and CARs that use the same recognition domains
.
J Immunol
2018
;
200
:
1088
100
.
18.
Eyquem
J
,
Mansilla-Soto
J
,
Giavridis
T
,
Stegen
SJC
,
van der Hamieh
M
,
Cunanan
KM
, et al
Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection
.
Nature
2017
;
543
:
113
.
19.
Liu
H
,
Rhodes
M
,
Wiest
DL
,
Vignali
DA
. 
On the dynamics of TCR:CD3 complex cell surface expression and downmodulation
.
Immunity
2000
;
13
:
665
75
.
20.
Lee
K-H
,
Dinner
AR
,
Tu
C
,
Campi
G
,
Raychaudhuri
S
,
Varma
R
, et al
The immunological synapse balances T cell receptor signaling and degradation
.
Science
2003
;
302
:
1218
22
.
21.
Harris
DT
,
Kranz
DM
. 
Adoptive T cell therapies: a comparison of T cell receptors and chimeric antigen receptors
.
Trends Pharmacol Sci
2016
;
37
:
220
30
.
22.
Quintarelli
C
,
Orlando
D
,
Boffa
I
,
Guercio
M
,
Polito
VA
,
Petretto
A
, et al
Choice of costimulatory domains and of cytokines determines CAR T-cell activity in neuroblastoma
.
Oncoimmunology
2018
;
7
:
e1433518
.
23.
Roose
JP
,
Diehn
M
,
Tomlinson
MG
,
Lin
J
,
Alizadeh
AA
,
Botstein
D
, et al
T cell receptor-independent basal signaling via Erk and Abl kinases suppresses RAG gene expression
.
PLOS Biol
2003
;
1
:
e53
.
24.
Mackall
CL
. 
Enhancing the efficacy of CAR T cells
.
Blood
2017
;
130
:
SCI–15–SCI–15
.
25.
Pathak
MK
,
Yi
T
. 
Sodium Stibogluconate is a potent inhibitor of protein tyrosine phosphatases and augments cytokine responses in hemopoietic cell lines
.
J Immunol
2001
;
167
:
3391
7
.
26.
Lal
G
,
Shaila
MS
,
Nayak
R
. 
Activated mouse T cells downregulate, process and present their surface TCR to cognate anti-idiotypic CD4+ T cells
.
Immunol Cell Biol
2006
;
84
:
145
53
.
27.
Rivas
FV
,
O'Keefe
JP
,
Alegre
M-L
,
Gajewski
TF
. 
Actin cytoskeleton regulates calcium dynamics and NFAT nuclear duration
.
Mol Cell Biol
2004
;
24
:
1628
39
.
28.
Xim Tan
Y
,
Manz
BN
,
Freedman
T
,
Zhang
C
,
Shokat
KM
,
Weiss
A
. 
Inhibition of Csk in thymocytes reveals a requirement for actin remodeling in the initiation of full T cell receptor signaling
.
Nat Immunol
2014
;
15
:
186
94
.
29.
Turatti
F
,
Figini
M
,
Balladore
E
,
Alberti
P
,
Casalini
P
,
Marks
JD
, et al
Redirected activity of human antitumor chimeric immune receptors is governed by antigen and receptor expression levels and affinity of interaction
.
J Immunother
2007
;
30
:
684
93
.
30.
Zhao
Q
,
Ahmed
M
,
Tassev
DV
,
Hasan
A
,
Kuo
T-Y
,
Guo
H
, et al
Affinity maturation of T-cell receptor-like antibodies for Wilms tumor 1 peptide greatly enhances therapeutic potential
.
Leukemia
2015
;
29
:
2238
47
.
31.
James
SE
,
Greenberg
PD
,
Jensen
MC
,
Lin
Y
,
Wang
J
,
Budde
LE
, et al
Mathematical modeling of chimeric TCR triggering predicts the magnitude of target lysis and its impairment by TCR downmodulation
.
J Immunol
2010
;
184
:
4284
94
.
32.
Hebeisen
M
,
Baitsch
L
,
Presotto
D
,
Baumgaertner
P
,
Romero
P
,
Michielin
O
, et al
SHP-1 phosphatase activity counteracts increased T cell receptor affinity
.
J Clin Invest
2013
;
123
:
1044
56
.
33.
Presotto
D
,
Erdes
E
,
Duong
MN
,
Allard
M
,
Regamey
P-O
,
Quadroni
M
, et al
Fine-tuning of optimal TCR signaling in tumor-redirected CD8 T cells by distinct TCR affinity-mediated mechanisms
.
Front Immunol
2017
;
8
:
1564
.
34.
Davenport
AJ
,
Cross
RS
,
Watson
KA
,
Liao
Y
,
Shi
W
,
Prince
HM
, et al
Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity
.
Proc Natl Acad Sci U S A
2018
;
115
:
E2068
76
.
35.
Salter
AI
,
Ivey
RG
,
Kennedy
JJ
,
Voillet
V
,
Rajan
A
,
Alderman
EJ
, et al
Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function
.
Sci Signal
2018
;
11
:
eaat6753
.
36.
Tan
YX
,
Zikherman
J
,
Weiss
A
. 
Novel tools to dissect the dynamic regulation of TCR signaling by the kinase Csk and the phosphatase CD45
.
Cold Spring Harb Symp Quant Biol
2013
;
78
:
131
9
.
37.
Hashimoto-Tane
A
,
Saito
T
. 
Dynamic regulation of TCR–microclusters and the microsynapse for T cell activation
.
Front Immunol.
2016
;
7
:
255
.
38.
Kumari
S
,
Depoil
D
,
Martinelli
R
,
Judokusumo
E
,
Carmona
G
,
Gertler
FB
, et al
Actin foci facilitate activation of the phospholipase C-γ in primary T lymphocytes via the WASP pathway
.
Elife
2015
;
4
:
e04953
.
39.
D'Oro
U
,
Vacchio
MS
,
Weissman
AM
,
Ashwell
JD
. 
Activation of the Lck tyrosine kinase targets cell surface T cell antigen receptors for lysosomal degradation
.
Immunity
1997
;
7
:
619
28
.
40.
Naramura
M
,
Jang
I-K
,
Kole
H
,
Huang
F
,
Haines
D
,
Gu
H
. 
c-Cbl and Cbl-b regulate T cell responsiveness by promoting ligand-induced TCR down-modulation
.
Nat Immunol
2002
;
3
:
1192
9
.
41.
Davanture
S
,
Leignadier
J
,
Milani
P
,
Soubeyran
P
,
Malissen
B
,
Malissen
M
, et al
Selective defect in antigen-induced TCR internalization at the immune synapse of CD8 T cells bearing the ZAP-70(Y292F) mutation
.
J Immunol
2005
;
175
:
3140
9
.
42.
Denkberg
G
,
Cohen
CJ
,
Segal
D
,
Kirkin
AF
,
Reiter
Y
. 
Recombinant human single-chain MHC-peptide complexes made from E. coli by in vitro refolding: functional single-chain MHC-peptide complexes and tetramers with tumor associated antigens
.
Eur J Immunol
2000
;
30
:
3522
32
.
43.
Oved
K
,
Ziv
O
,
Jacob-Hirsch
J
,
Noy
R
,
Novak
H
,
Makler
O
, et al
A novel postpriming regulatory check point of effector/memory T cells dictated through antigen density threshold-dependent anergy
.
J Immunol
2007
;
178
:
2307
17
.
44.
D'Oro
U
,
Munitic
I
,
Chacko
G
,
Karpova
T
,
McNally
J
,
Ashwell
JD
. 
Regulation of constitutive TCR internalization by the ζ-chain
.
J Immunol
2002
;
169
:
6269
78
.
45.
Tyanova
S
,
Temu
T
,
Sinitcyn
P
,
Carlson
A
,
Hein
MY
,
Geiger
T
, et al
The Perseus computational platform for comprehensive analysis of (prote)omics data
.
Nat Methods
2016
;
13
:
731
40
.
46.
R Core Team
. 
R: a language and environment for statistical computing
. 
2016
.
Available from
: https://www.r-project.org/.
47.
Benjamini
Y
,
Hochberg
Y
. 
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J R Stat Soc Ser B Methodol
1995
;
57
:
289
300
.
48.
Hamieh
M
,
Dobrin
A
,
Cabriolu
A
,
van der Stegen
SJC
,
Giavridis
T
,
Mansilla-Soto
J
, et al
CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape
.
Nature
2019
;
568
:
112
16
.