Weekly Digests
‹ Back to December

Like coffee for T cells, c-Jun mediates resistance to exhaustion

December 18, 2019

In an effort to understand and overcome exhaustion in adoptively transferred CAR T cells, Lynn et al. developed a cellular model for exhaustion and interrogated its programming. Their results, recently published in Nature, reveal a critical role for the transcription factor component c-Jun (part of the canonical AP-1 c-Fos–c-Jun heterodimer) in preventing exhaustion. Capitalizing on this information, the researchers also show that c-Jun overexpression can be used to enhance the efficacy of CAR T cell therapy in several models.

To first model T cell exhaustion, Lynn et al. incorporated a tonically signaling HA-28z CAR into T cells to induce hallmark features of exhaustion, including reduced expansion, increased expression of inhibitory molecules, increased differentiation towards effector phenotypes, reduced IFNγ production, and markedly reduced IL-2 production following stimulation compared to control CD19-28z CAR T cells. Differential gene expression and RNAseq data validated this exhausted state.

To probe the molecular mechanisms driving differential gene expression in tonically signaling HA-28z CAR T cells, the researchers performed ATACseq analysis, which revealed extensive differential chromatin accessibility driven primarily by the expression of the HA-28z CAR. Clustering the top 5000 differentially accessible regions showed evidence of a consistent epigenetic state across HA-28z-expressing cells, with a consistent pattern among CD4+, CD8+, naive, and memory subsets. This epigenetic state showed increased chromatin access to genes like CTLA4 and reduced chromatin access to memory-associated genes like IL7R, consistent with exhaustion.

Analyzing the transcriptional programs associated with the observed exhaustion-associated epigenetic state, Lynn et al. found that AP-1–bZIP and bZIP–IRF binding motifs were particularly accessible in exhausted CAR T cells, as were NF-κB, NFAT, and RUNX transcription factor motifs (in some clusters). Paired RNAseq analysis revealed increased bZIP and IRF transcription factor expression (most notably JUNB, FOSL1, BATF, BATF3, ATF3, ATF4, and IRF4), and protein analysis confirmed the increase in JunB, IRF4, and BATF3. Importantly, the researchers noted that BATF and IRF4 – transcription factors that infamously drive regulatory programs – were present at higher relative levels than c-Jun, which dimerizes with c-Fos to form the canonical AP-1 transcription factor that drives IL-2 expression. As AP-1, BATF, and IRF4 are known to compete for access to binding motifs, Lynn et al. hypothesized that an imbalance between these activating and regulatory transcription factors may underlie exhaustion programming.

To test this hypothesis, Lynn et al. engineered tonically signaling HA-28z CAR T cells to overexpress AP-1. This effectively restored cellular production of IL-2, and was dependent on c-Jun, but not c-Fos. HA-28z CAR T cells engineered to overexpress just c-Jun showed similar restoration of IL-2 and IFNγ production; to fully restore IL-2 production, c-Jun overexpression was required during both T cell expansion and antigen stimulation. JUN-HA-28z CAR T cells also showed increased functional capacity and a shift from effector and effector memory differentiation towards stem cell memory and central memory differentiation.

In non-tonically signaling CD19-specific CAR T cells, c-Jun overexpression did not affect IL-2 or IFNγ production, but did enhance long-term proliferative capacity and reduce expression of exhaustion markers in vitro. In vivo, c-Jun overexpression in transferred CAR T cells enhanced their capacity for expansion. In one model, however, this also accelerated the progression of graft-versus-host disease.

Based on their evidence that c-Jun overexpression prevents the induction of exhaustion programs, the researchers looked at ATACseq and RNAseq data to compare JUN-HA-28z CAR T cells to HA-28z CAR T cells. While c-Jun overexpression did not appear to change the epigenetic profile within cells, it did substantially alter the transcriptome, reducing expression of exhaustion-associated genes and increasing expression of memory-associated genes. Unsurprisingly, many of the genes affected by c-Jun overexpression were found to be enriched for AP-1 family binding sites.

Lynn et al. then hypothesized that c-Jun overexpression may prevent exhaustion directly, by activating AP-1 target genes, and/or indirectly, by disrupting the activity of immunoregulatory transcriptional complexes. To test which of these mechanisms might be at play, the researchers evaluated a panel of c-Jun mutants predicted to be deficient in transcriptional activation, DNA binding, or dimerization. Two mutants deficient in transcriptional activation restored IL-2 production as effectively as wild-type c-Jun, while a third demonstrated partial rescue of IL-2. Mutants deficient in DNA binding or dimerization, on the other hand, did not rescue cytokine production in exhausted HA-28z CAR T cells. Overexpression of c-Jun decreased levels of exhaustion-driving AP1–IRF and JunB–BATF transcriptional complexes, as well as total chromatin-bound JunB, BATF, and BATF3. Further, knocking out JunB, BATF, or especially IRF4 increased production of IL-2 and IFNγ. Together, these results support the notion that c-Jun overexpression staves off exhaustion programming primarily by mediating the displacement of immunoregulatory transcription factors.

Finally, to test antitumor efficacy, c-Jun-overexpressing CAR T cells were adoptively transferred into various murine tumor models. Compared to HA-28z CAR T cells, JUN-HA-28z CAR T cells showed enhanced antitumor activity in a Nalm6-G2D+ leukemia model. Importantly, overexpression of c-Jun in CD22-specific or CD19-specific CAR T cells enhanced antitumor efficacy in leukemia models with low target antigen expression. In a solid tumor setting, JUN-Her2-BBz CAR T cells prevented the growth of 143B osteosarcoma tumors and improved long-term survival of mice. The JUN-Her2-BBz CAR T cells showed greater expansion, greater accumulation in tumors, reduced expression of exhaustion markers, longer-term persistence, and enhanced cytokine production upon restimulation ex vivo compared to control Her2-BBz CAR T cells. A small cluster of JUN-Her2-BBz CAR T cells characterized by high IL7R expression persisted long-term in mice, consistent with the maintenance of a self-renewing memory-like population. Similar results were observed when treating 143B sarcoma with JUN-G2D-BBz CAR T cells.

Overall, Lynn et al. show that a functional deficiency in c-Jun mediates dysfunction in exhausted T cells, and that forcing overexpression of c-Jun can prevent exhaustion programming by outcompeting exhaustion-inducing immunoregulatory transcription factors. This strategy could be used to prevent exhaustion and enhance the efficacy of CAR T cells.

by Lauren Hitchings

References:

Lynn R.C., Weber E.W., Sotillo E., Gennert D., Xu P., Good Z., Anbunathan H., Lattin J., Jones R., Tieu V., Nagaraja S., Granja J., de Bourcy C.F.A., Majzner R., Satpathy A.T., Quake S.R., Monje M., Chang H.Y., Mackall C.L. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature. 2019 Dec.

In the Spotlight...

Recruitment of CD103+ dendritic cells via tumor-targeted chemokine delivery enhances efficacy of checkpoint inhibitor immunotherapy

Williford and Ishihara et al. showed enhanced CCL4 levels and stroma localization in tumors when mice received CCL4 fused to a collagen-binding domain (CBD). CCL4-CBD synergized with checkpoint inhibitors (CPI) to slow tumor growth in a spontaneously developing and in multiple implanted CPI-poorly responsive cancer mouse models, and to establish memory. IHC and flow cytometry of tumor infiltrates revealed that these effects correlated with CD103+ DC and CD8+ T cells (eliminated in Batf3-/- mice). CXCR3 blockade eliminated the therapeutic benefit, showing that CCL4-CBD recruited CD103+ DC and CD8+ T cells that make IFNγ and TNFα to mediate efficacy.

Contributed by Paula Hochman

Discovery of Small Molecules for the Reversal of T Cell Exhaustion

Marro et al. prepared exhausted lymphocytic choriomeningitis virus (LCMV)-specific splenic CD8+ T cells that were deficient in IFNγ production following re-stimulation in vitro with LCMV peptides (but rescuable by anti-PDL-1), and utilized these cells in an antigen-dependent assay to screen a drug-repurposing library of 12,000 small molecules for reversal of T cell exhaustion, identifying Ingenol mebutate (IngMb), a diterpene ester targeting protein kinase C (PKC). IngMb induced IFNγ-producing CD8+ T cells that expressed markers of CD8+ T cell memory lineage and effector differentiation, supporting a role for PKC in reversing T cell exhaustion.

Contributed by Samuel Goldman

Single-Cell Profiling Defines Transcriptomic Signatures Specific to Tumor-Reactive versus Virus-Responsive CD4+ T Cells

Using single-cell RNAseq, Magen and Nie et al. compared LCMV-specific CD4+ T cells in tumors (TIL) or draining lymph nodes (dLN) of mice with MC38 tumors expressing LCMV antigen and in spleens of mice with acute or chronic LCMV infection. These populations showed distinct, heterogeneous transcriptome patterns. TIL and dLN profiles were dominated by Th1-like and T follicular helper cells, respectively. A type I IFN-driven signature shown for exhausted PD-1+ mouse Th1-like TILs (and not for chronic LCMV exhausted T cells) was also identified for CD4+ T cells infiltrating human tumors and negatively correlated with melanoma patient response to checkpoint blockade.

Contributed by Paula Hochman

Myeloid cells orchestrate systemic immunosuppression, impairing the efficacy of immunotherapy against HPV+ cancers

To study immune suppression in advanced HPV+ cancers, Galliverti et al. used a nanoparticle-based E7 therapeutic vaccine in a human-mimicking cervical cancer GEMM with myeloid cell accumulation and poor DC and CTL function. The vaccine alone, or in combination with ICB, failed to increase CD8+ T cells in the TME. CD11b+ myeloid cells (monocytes; neutrophils could not be studied) in lymphoid organs were found to potently suppress CD8+ T cell proliferation and cytokine production, inhibit synergistic effects with ICB, and decrease APC function, suggesting that sustained depletion of myeloid cells might be beneficial in cancers with myeloid cell accumulation.

Contributed by Katherine Turner

Self-maintaining CD103+ cancer-specific T cells are highly energetic with rapid cytotoxic and effector responses

Hamid et al. compared functionality of TCR-paired CD103+ and CD103- cytotoxic CD8+ T cells (CTLs) specific to cancer testis antigens. CD103+ CTLs showed increased antigen sensitivity, cancer cell killing, and migration rates, dependent on expression of E-cadherin (the CD103 ligand) on target cells. CD103 expression was maintained through autocrine TGFβ1 signaling, and CD103+ CTLs had enhanced metabolic activity and more rapidly apoptosed after prolonged antigen exposure. In patient lung tumors, CD103+ CTLs clustered with E-cadherin-rich tumor areas and expressed inhibitory molecules when the CTLs were CD39high, but not CD39low.

Contributed by Alex Najibi

On-target restoration of a split T cell-engaging antibody for precision immunotherapy

Aiming to precisely target tumors, Banaszek and Bumm et al. designed a tri-specific antibody split into two halves (hemibodies) – one composed of a VH domain of anti-CD3 fused to an scFv targeting one antigen, and the other containing the VL domain of anti-CD3 fused to an scFv targeting a second antigen. The two hemibodies associated and reconstituted the CD3-binding site only upon binding both target antigens on the same cell. Unlike BiTEs, the hemibodies stimulated donor T cells to lyse double-positive, but not single-positive, leukemia and breast cancer cells in vitro and in NSG mice, leading to prolonged survival.

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.