Weekly Digests
‹ Back to March

NR4A transcription factors control T cell dysfunction programs

March 13, 2019

T cell dysfunction – in the form of exhaustion, anergy, or tolerance – is known to limit the effector functions of T cells, but the molecular mechanisms that induce dysfunctional programs are not fully understood. Two recent studies, published back-to-back in Nature, investigated the mechanisms underlying T cell dysfunction, and through different strategies, both studies identified NR4A transcription factors as essential to induction of dysfunction.

Focused on studying T cell exhaustion, particularly in the context of CAR T cells, Chen et al. transferred CD19-specific CAR T cells into mice bearing CD19-expressing cancers. After a period of time, the transferred CAR T cells developed hallmarks of exhaustion including high PD-1 and TIM3 expression, and decreased effector function. Analysis of chromatin accessibility profiles revealed that regions selectively accessible in PD-1hi T cells were enriched for NR4A, NFAT, and NFκB motifs. RNAseq data further supported these observations, pointing to a role for NR4A family transcription factors (NR4A1, NR4A2, and NR4A3) as transcriptional effectors of T cell dysfunction.

Liu, Wang, and Lu et al. similarly identified upregulation of NR4A1 after a genome-wide genetic and epigenetic analysis of in vitro-induced tolerant T cells. When they induced overexpression of NR4A1 in naive T cells, they observed upregulation of genes related to anergy and exhaustion, downregulation of genes related to effector programs, reduced TH1 and TH17 differentiation in CD4+ T cells, and reduced IFNγ production by CD8+ T cells. Similarly, ablation of NR4A1 enhanced effector functions of CD4+ and CD8+ T cells, increased expansion, and blocked the formation of tolerance.

To study the effective role of NR4A transcription factors in vivo, Liu, Wang, and Lu et al. treated tumor-bearing mice with tumor-specific CD8+ T cells with NR4A1 knocked out. NR4A1-/- T cells outperformed their wild-type counterparts, expressing less PD-1 and TIM3, producing more IFNγ and TNF, infiltrating tumors more readily, and mediating more pronounced tumor regression. In the context of both acute and chronic viral infection, NR4A1-/-CD8+ T cells showed enhanced effector function, Tbet expression, and proliferation, and reduced expression of exhaustion markers.

In a similar series of adoptive transfer experiments, Chen et al. compared the antitumor efficacy of wild-type CAR T cells to NR4A triple knockout (TKO; NR4A1, NR4A2, and NR4A3) CAR T cells. In both immunodeficient and immunocompetent tumor-bearing mice, wild-type CAR T cells showed limited efficacy, while NR4A TKO CAR T cells induced pronounced tumor regression and enhanced survival. NR4A TKO CAR T cells isolated from tumors 8 days after cell transfer showed reduced expression of PD-1 and TIM3, and the percentage of cells producing IFNγ and TNF upon restimulation was higher compared to wild-type CAR TILs. Compared to the TKO cells, CAR T cells with individual knockouts of NR4A1, NR4A2, or NR4A3 showed weaker antitumor efficacy, increased expression of inhibitory receptors PD-1 and TIM3, and decreased ability to produce cytokines upon restimulation.

Transcriptional and chromatin accessibility comparisons performed by Chen et al. showed that NR4A-expressing cells promoted the expression of dysfunction programs by favoring the accessibility and expression of exhaustion-related genes, and that each individual NR4A transcription factor, functioning downstream of NFAT, played a similar role in maintaining the accessibility of an enhancer for the Pdcd1 transcription start site. Similarly, Liu, Wang, and Lu et al. showed that NR4A1 expression correlated with HeK4me3 (a histone modification marking active transcription) near tolerance-related genes.

In addition to activating dysfunctional programs, NR4A was also found to repress effector programs. Chen et al. noted that in chromatin selectively accessible in NR4A TKO CAR T cells, regions near certain chemokine and cytokine genes were enriched for binding motifs of transcription factors including bZIP and Rel/NFκB, and the activity of these transcription factors was associated with the increased effector functions of NR4A TKO CAR T cells in vivo. Meanwhile, Liu, Wang, and Lu et al. found that the top NR4A1-binding motifs were consensus sequences containing AP-1 – a transcription factor involved in effector function programming. They determined that mechanistically, NR4A1 was recruited to the binding sites of AP-1, inhibiting its function and ultimately repressing T cell effector programs. Both research teams found that PD-1 blockade reduced NR4A expression in CD8+ T cells by reducing the accessibility of NR4A binding motifs.

Understanding how exhaustion is regulated on the epigenetic and transcriptional level is critical to designing immunotherapies which depend on T cells’ ability to maintain effector functions. As observed here, knocking out NR4A in T cells prior to adoptive transfer enhances their effector function and prevents exhaustion – an effect similar to that of checkpoint blockade. These studies thus identify NR4A as a new “checkpoint”, and novel strategies of targeting NR4A in immunotherapy may be effective in preventing or controlling T cell dysfunction.

by Lauren Hitchings

References:

Chen J., López-Moyado I.F., Seo H., Lio C.J., Hempleman L.J., Sekiya T., Yoshimura A., Scott-Browne J.P., Rao A. NR4A transcription factors limit CAR T cell function in solid tumours. Nature. 2019 Feb 27.

Liu X., Wang Y., Lu H., Li J., Yan X., Xiao M., Hao J., Alekseev A., Khong H., Chen T., Huang R., Wu J., Zhao Q., Wu Q., Xu S., Wang X., Jin W., Yu S., Wang Y., Wei L., Wang A., Zhong B., Ni L., Liu X., Nurieva R., Ye L., Tian Q., Bian X.W., Dong C. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature. 2019 Feb 27.

In the Spotlight...

Order of administration of combination cytokine therapies can decouple toxicity from efficacy in syngeneic mouse tumor models

In an attempt to use scheduling to decouple therapeutic efficacy from toxicity, Rothschilds et al. treated tumor-bearing mice with a tumor-targeting antibody on day 0, IFNα on day 1, and an extended half-life version of IL2 (eIL2) on days 0, 1, 2, or 3. All regimens exhibited similar therapeutic efficacy, but only administration of eIL2 before IFNα resulted in significant weight loss, which could be attributed to eIL2-mediated increase in NK cell activation and sensitization to IFNα signaling as well as multiple systemic cytokine increases. NK cells played a critical role in weight loss, but not in efficacy.

SnapShot: Cancer Immunotherapy with Oncolytic Viruses

Gujar et al. briefly review and graphically illustrate the oncolytic virus approach to cancer immunotherapy, describing its mechanisms of action (oncolysis, transgene expression, tumor vasculature collapse, and stimulation of antitumor immunity via induction of cytokines and chemokines), outlining the resulting innate and adaptive immune responses, and describing barriers and potential solutions to improving use.

Peripheral T cell cytotoxicity predicts T cell function in the tumor microenvironment

Iwahori et al. analyzed tumors, normal lung tissue, and peripheral blood from patients with early-stage NSCLC for T cell markers, cytokine production, and cytotoxic capability (using a bispecific T-cell engager and a surrogate tumor cell line for the latter). Cytotoxicity of CD8+ T cells (mostly CD45RA+ effector memory) in peripheral blood correlated with cytotoxicity of tumor-infiltrating T cells. There was some overlap in TCR clonotypes between the three sample types. Cytotoxic capacity of peripheral blood T cells correlated with nivolumab efficacy in a small data set, suggesting the possibility of blood-based diagnostics as a predictive biomarker.

Phase I/II trial testing safety and immunogenicity of the multipeptide IMA950/poly-ICLC vaccine in newly diagnosed adult malignant astrocytoma patients

Migliorini and Dutoit et al. immunized 19 glioma patients with 9 HLA-A2 epitope-length peptides (identified from glioblastoma tumor samples using mass spectrometry and shown to be overexpressed in gliomas and immunogenic in vitro) and 2 HLA-DR peptides. The initial 6 patients received the peptides intradermally and poly-ICLC intramuscularly, and showed few T cell responses. Combining peptide and adjuvant improved immunogenicity, particularly evidenced by induction of multiple CD8+ and CD4+ T cell responses per patient. Clinical outcomes were unremarkable for this small cohort and there were no signs of immunoediting.

Autologous lymphocyte infusion supports tumor antigen vaccine-induced immunity in autologous stem cell transplant for multiple myeloma

To improve the immune contribution of autologous stem cell transplant (autoSCT) to long-term survival in multiple myeloma patients, Cohen and Lendvai et al. added autologous lymphocyte infusion of MAGE-A3-primed T cells shortly after autoSCT, followed by continued vaccination with full-length MAGE-A3 protein. Humoral and CD4+ T cell responses were detected in all patients, with only a small subset showing CD8+ responses. CD8+ T cells recognized processed antigen. Interestingly, in 4 of 8 patients biopsied following relapse, MAGE-A expression was lost compared to baseline, suggesting immune escape.

A Novel Form of 4-1BBL Prevents Cancer Development via Nonspecific Activation of CD4+ T and Natural Killer Cells

Barsoumian et al. find that prophylactic administration of a soluble, multimerizable fusion protein of 4-1BB ligand and streptavidin (SA-4-1BBL) alone prevented development of TC-1, LLC, and 3LL-huMUC1 tumors in mice in a dose- and timing-dependent manner. SA-4-1BBL expanded CD44+IFNγ+CD4+ T cells and IFNγ+ NK cells. Prophylactic anti-4-1BB agonistic antibody conferred neither effect. IFNγ blockade or depletion of CD4+ T cells or NK cells, but not CD8+ T cells and B cells, abrogated the protective effect of SA-4-1BBL. SA-4-1BBL treatment of mice with surgically resected TC-1 or 3LL-huMUC1 tumors prevented recurrence.

Contributed by Alex Najibi

REVIEW: Immune control by amino acid catabolism during tumorigenesis and therapy

Lemos et al. review in detail the evidence for and the processes by which catabolism of the amino acids tryptophan (Trp) and arginine (Arg) suppresses antitumor immunity and promotes tumor growth. Chronic inflammation within the TME leads to elevated Trp and Arg catabolism via IDO/TDO and ARG1, respectively, by tumor, stromal, and monocytic cells. Lack of sufficient Trp and Arg suppresses proliferation and function of effector T cells, but stabilizes Tregs. In particular, increased IDO expression correlates with poor survival in some cancer types, making understanding the underlying regulatory mechanisms critical to targeting these inhibitory pathways.

Targeting the tumor stroma with an oncolytic adenovirus secreting a fibroblast activation protein-targeted bispecific T-cell engager

In order to eliminate the immunosuppressive tumor stroma, which also prevents oncolytic adenoviruses (OAds) from entering the tumor, de Sostoa et al. designed a bispecific T-cell engager (BiTE) targeting the cancer-associated fibroblast (CAF) activation protein-α (FAP) and transduced it into an OAd. Cells infected with FBiTE-OAd secreted BiTE, which induced activation and proliferation of CD4+ and CD8+ T cells in the presence of FAP-expressing cells in vitro. When injected intratumorally, FBiTE-OAd increased accumulation and retention of T cells, killed FAP+ CAFs, reduced tumor growth, and increased survival in two mouse tumor models.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.