In this issue of Blood, Selli et al1 have discovered that whereas CD28-bearing chimeric antigen receptor (CAR) T cells become dysfunctional and exhausted via the classical events occurring with chronic stimulation from tumors or chronic infections, 4-1BB–bearing CAR T cells become dysfunctional in a different way that is driven by the transcription factor FOXO3.

A little over a decade ago, CAR T cells were shown to have remarkable efficacy in B-cell malignancies.2-4 All of them targeted the B-cell molecule CD19 and bore the intracellular signaling domain of 1 of 2 costimulatory molecules, either CD28 or 4-1BB. The incorporation of a costimulatory domain into the CAR was hypothesized to improve persistence and function and, therefore, antitumor effects. Owing to the remarkable initial success of these initial products, all 6 constructs that are approved by the US Food and Drug Administration, including those targeting the B-cell maturation antigen expressed by myeloma plasma cells, and most other CARs in clinical development across a variety of indications, incorporate 1 of these 2 signaling domains.

Because CARs recognize antigen with high affinity and are expressed by constitutive promoters, investigators noted early on that continuous stimulation with antigen, and sometimes even just high-level CAR expression, could drive T-cell dysfunction. These dysfunctional T cells had reduced cytotoxicity, cytokine production, and proliferation after antigen stimulation; this could be overcome by intermitting rest5 from signal transduction. In patients, high tumor burden has been correlated with reduced response and increased T-cell dysfunction.6 Until now, this dysfunction was thought to be essentially the same phenomenon as T-cell “exhaustion,” which has been extensively described in the setting of chronic viral infection and cancers treated with checkpoint blockade.7 

Here, Selli et al modeled chronic activation by repetitive in vitro stimulation of CD19-directed CAR T cells bearing either CD28 or 4-1BB intracellular signaling domains to generate dysfunction, and then performed a comprehensive analysis of the transcriptional, epigenetic, and phenotypic programs. The CAR T cells were stimulated with new tumor cells at a low effector-to-target ratio every 48 hours. After 2 weeks, the CAR T cells lost their ability to kill antigen-positive targets, could no longer make cytokines, and could no longer proliferate. Next, they interrogated these dysfunctional CAR T cells phenotypically, using multiparameter cytometry by time-of-flight. They found that CAR T cells bearing CD28-based CARs bore the hallmarks of classic T-cell exhaustion, including resurgent expression of PD-1, TIGIT, LAG3, TIM3, and CTLA4 (see figure). In contrast, dysfunctional T cells bearing 4-1BB CARs occupied different t-distributed stochastic neighbor embedding space by nearest neighbor analysis and expressed higher CD62L and CD25 on their surface. Interrogation of the transcriptional programs of these dysfunctional CAR T cells using RNA sequencing revealed that CD28-based CAR T cells were highly enriched for exhaustion-associated genes, whereas 4-1BB–based dysfunctional CAR T cells did not have the classic exhaustion signatures. Recent studies of T-cell exhaustion have emphasized that the state is defined by specific epigenetic alterations.8 The investigators also confirmed these differences using assay for transposase-accessible chromatin with sequencing (ATAC-seq), particularly of the chromatin accessibility of the gene encoding PD1 (PDCD1), which looked like classic exhaustion in the CD28-based CAR T cells but not the 4-1BB–based CAR T cells. The authors then used longitudinal single-cell RNA sequencing and identified that most of the dysfunctional 4-1BB CAR T cells were defined by a distinct set of genes involved in cytotoxicity (GNLY, CCL5, PRF1, GZMA, GZMK, CTSW), natural killer cell identity (KLRK1, KLRC2), and T-cell differentiation (ID2), which they termed the TBBD signature (see figure). Intriguingly, their TBBD signature was confirmed in a single patient who had received tisagenlecleucel (an anti-CD19/4-1BB CAR construct) and whose lymphoma had progressed despite persisting CAR T cells in their blood.

CAR T cells bearing different costimulation domains exhibit different transcriptional, epigenetic, and phenotypic signatures when they become dysfunctional after chronic stimulation. CAR T cells bearing a 4-1BB costimulation domain reactivate FOXO3, which drives a novel program of dysfunction that is distinct from classic T-cell exhaustion.

CAR T cells bearing different costimulation domains exhibit different transcriptional, epigenetic, and phenotypic signatures when they become dysfunctional after chronic stimulation. CAR T cells bearing a 4-1BB costimulation domain reactivate FOXO3, which drives a novel program of dysfunction that is distinct from classic T-cell exhaustion.

Close modal

To understand the origin of this novel molecular program of dysfunction, they interrogated transcription factor binding accessibility over time. As expected, CD28-CAR dysfunction had increased accessibility for Jun:Fos (AP1) binding, but in contrast, 4-1BB-CAR dysfunction opened homeobox (HOX) and forkhead box (FOX) sites, and the small conditional RNA-sequencing data revealed high activity of FOXO3. Furthermore, in mechanistic studies, disruption of FOXO3 by CRISPR/Cas resulted in resistance to dysfunction after repetitive antigen stimulation, and, conversely, FOXO3 overexpression dramatically reduced the expansion of 4-1BB (but not CD28) CAR T cells in the setting of repetitive antigen stimulation. In murine xenograft stress models, in which very low numbers of CAR T cells are injected to treat higher tumor burdens, FOXO3 knockout CAR T cells bearing a 4-1BB intracellular signaling domain improved survival.

Remarkably, the authors discovered a new molecular mechanism by deep technological interrogation, spanning from high-dimensional cytometry to longitudinal single-cell RNA sequencing, and while still using a relatively simple in vitro model (repetitive antigen stimulation). The molecular program itself, and reactivation of FOXO3, is interesting and perhaps not typically found in nature. In natural biology, 4-1BB stimulation is temporally dissociated from T-cell receptor (TCR) stimulation, and TCR expression itself cycles with antigen exposure, thus reducing the possibility of tonic signaling. Future studies could include further validation of the TBBD transcriptional signature and FOXO3 activity in more patients who received tisagenlecleucel and did not respond. Likewise, a deeper understanding of whether tisagenlecleucel gets exhausted in patients based on chronic exposure to nascent CD19+ B cells, regardless of disease status, should be explored. Interestingly, chronic exposure to nascent B cells was thought to have a role in the decade-long persistence of tisagenlecleucel in the first 2 patients treated, both of whom had prolonged remissions.9 In addition, it will be interesting for the field to see whether 4-1BB–bearing CAR T cells that are specific to other antigens, such as both of the B-cell maturation antigen (BCMA)–targeted CAR T cells that are approved for multiple myeloma, also exhibit the TBBD signature and FOXO3 reactivation when patients with BCMA-positive disease recur despite persistent BCMA-directed CAR T cells. Finally, at the mechanistic level, it will be interesting to see whether FOXO3 knockout or c-Jun overexpression10 is more potent in reducing CAR T cell exhaustion.

Conflict-of-interest disclosure: M.V.M. is an inventor on patents related to adoptive cell therapies, held by Massachusetts General Hospital (some licensed to Promab) and University of Pennsylvania (some licensed to Novartis); receives grant/research support from Kite Pharma; has served as a consultant for multiple companies involved in cell therapies; holds equity in 2SeventyBio, Century Therapeutics, Neximmune, Oncternal, and TCR2; and serves on the board of directors of 2Seventy Bio.

1.
Selli
ME
,
Landmann
JH
,
Terekhova
M
, et al
.
Costimulatory domains direct distinct fates of CAR-driven T-cell dysfunction
.
Blood
.
2023
;
141
(
26
):
3153
-
3165
.
2.
Kochenderfer
JN
,
Dudley
ME
,
Feldman
SA
, et al
.
B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells
.
Blood
.
2012
;
119
(
12
):
2709
-
2720
.
3.
Porter
DL
,
Levine
BL
,
Kalos
M
,
Bagg
A
,
June
CH
.
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia
.
N Engl J Med
.
2011
;
365
(
8
):
725
-
733
.
4.
Brentjens
RJ
,
Davila
ML
,
Riviere
I
, et al
.
CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia
.
Sci Transl Med
.
2013
;
5
(
177
):
177ra38
.
5.
Weber
EW
,
Parker
KR
,
Sotillo
E
, et al
.
Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling
.
Science
.
2021
;
372
(
6537
):
eaba1786
.
6.
Locke
FL
,
Rossi
JM
,
Neelapu
SS
, et al
.
Tumor burden, inflammation, and product attributes determine outcomes of axicabtagene ciloleucel in large B-cell lymphoma
.
Blood Adv
.
2020
;
4
(
19
):
4898
-
4911
.
7.
Blank
CU
,
Haining
WN
,
Held
W
, et al
.
Defining 'T cell exhaustion'
.
Nat Rev Immunol
.
2019
;
19
(
11
):
665
-
674
.
8.
Sen
DR
,
Kaminski
J
,
Barnitz
RA
, et al
.
The epigenetic landscape of T cell exhaustion
.
Science
.
2016
;
354
(
6316
):
1165
-
1169
.
9.
Melenhorst
JJ
,
Chen
GM
,
Wang
M
, et al
.
Decade-long leukaemia remissions with persistence of CD4(+) CAR T cells
.
Nature
.
2022
;
612
(
7941
):
E22
.
10.
Lynn
RC
,
Weber
EW
,
Sotillo
E
, et al
.
c-Jun overexpression in CAR T cells induces exhaustion resistance
.
Nature
.
2019
;
576
(
7786
):
293
-
300
.
Sign in via your Institution