Weekly Digests
‹ Back to October

BATF drives CAR T cells… to exhaustion

October 19, 2022

T cell exhaustion is a major focus of research in cancer immunotherapy, and particularly in CAR T cell therapy, but still the exact mechanisms that drive T cells to exhaustion are not fully understood. Recently, Zhang, Zhang, and Qiao et al. developed an in vitro CAR hypofunction model and identified the AP-1 family transcription factor BATF – which different groups have reported to have conflicting functions – as a major driver of CAR T cell exhaustion. The results of this investigation were recently published in Cancer Cell.

To develop their hypofunction CAR T cell model, Zhang, Zhang, and Qiao et al. cultured T cells expressing the M28Z CAR (anti-mesothelin [MSLN] scFv, CD28 costimulatory domain, and CD3ζ intracellular domain) with NCI-H226 or OVCAR3 tumor cells, which express the target antigen MSLN. The M28Z CAR T cells induced robust tumor cell lysis upon co-culture, though at a low effector:target cell (E:T) ratio, CAR T cell mediated-lysis plateaued around day 4, despite the maintained expression of both the M28Z CAR on T cells and MSLN on tumor cells.

The researchers then compared “hypofunction” M28Z CAR T cells (from 7-day co-culture at a low E:T) to activated M28Z CAR T cells (from 1-day co-culture at a high E:T ratio), control “fresh” M28Z CAR T cells (cultured alone for 2 days), and control “unstimulated” M28Z CAR T cells (cultured alone for 7 days). Hypofunction M28Z produced fewer cytokines, showed increased expression of exhaustion-related genes, (e.g., PDCD1, HAVCR2, CTLA, and TIGIT), and showed reduced expression of activation-related genes (e.g., IFNG, IL2, TNFRSF9, and GZMB) compared to activated M28Z or controls. Gene set enrichment analysis (GSEA) further revealed that exhaustion-related genes identified in patients with cancer were enriched in the upregulated genes of hypofunction M28Z. Together, these results are indicative of a profound exhaustion phenotype in hypofunction M28Z.

Next, the researchers set out to identify genes that might contribute to hypofunction/exhaustion. After validating the function of PDCD1 by showing that its knockout improved CAR T cells’ ability to eliminate tumors, the researchers evaluated several less studied genes, including 19 genes with expression patterns that resembled PDCD1, and 11 genes that were selected for their upregulation in relation to T cell function. Of the genes screened, two stood out for their role in mediating exhaustion: BATF and IRF4.

To confirm the roles of BATF and IRF4 in CAR T cells, Zhang, Zhang, and Qiao et al. generated knockout models of each and showed that elimination of either gene led to increased cytokine release and tumor cell lysis, and to increased proliferation upon multiple rounds of tumor challenge. These effects were more pronounced in the BATF knockouts than in the IRF4 knockouts. Similarly, RNAseq and gene ontology analysis showed upregulation of genes enriched in T cell activation-associated pathways in both knockout models, but reduced expression of genes enriched in exhaustion-associated pathways only in the BATF knockout CAR T cells.

Testing whether this enhanced functionality would carry through to in vivo models, the researchers evaluated BATF-knockout CAR T cells and IRF4-knockout CAR T cells against control M28Z CAR T cells in cell line-derived xenograft (CDX) mice bearing NCI-H226 tumors. While standard M28Z and IRF4-knockout CAR T cells both slowed tumor growth compared to control-treated mice, this effect was significantly stronger in mice treated with BATF-knockout CAR T cells. Further, only the BATF-knockout CAR T cell treatment increased tumor-infiltrating T cells, which expressed IFNG and MKI67 by in situ RNA hybridization. Similar effects were observed in a patient-derived xenograft (PDX) model. In this setting, standard and IRF4-knockout CAR T cells only slowed tumor growth, while BATF-knockout CAR T cells cleared tumors in 4 of 5 mice. Further, higher levels of human CD3+ T cells were maintained in this group, and cured mice were able to clear tumors upon rechallenge, indicating durable protection. Similar results were also observed in a second PDX model,

To further validate the role of BATF in human CAR T cells, BATF was overexpressed by lentiviral transduction. This reduced the cytolytic capacity of CAR T cells upon multiple rounds of tumor challenge, while BATF knockout enhanced cytolysis. These effects were most pronounced when the E:T ratio was low, and with each additional cell challenge. Similar results were observed across in vitro and in vivo studies of 5 different CAR T cell types with different targets and costimulatory domains, as well as in murine OT-1 cells.

Next, the researchers performed RNAseq on CAR T cells after 3 rounds of in vitro tumor challenges, followed by ChIPseq and motif analysis of regions bound by BATF in CAR T cells. Comparing human exhaustion-related genes with those bound by BATF, the researchers found significant overlap. BATF was also found to directly bind to and upregulate PDCD1, increasing PD-1 expression and downstream exhaustion in BATF-overexpressing cells. In mouse models with overexpression of PD-L1 in tumors, BATF-knockout T cells showed enhanced antitumor activity. Interestingly, though BATF-overexpressing cells showed upregulation of genes related to proliferation, BATF-knockout cells were more abundant under exhaustion-inducing conditions, suggesting that the role BATF plays in inducing exhaustion is dominant over its role in inducing activation.

BATF has also been reported to play a role in the differentiation of effector T cells, and here the researchers found that BATF bound to and upregulated a number of effector genes (e.g., RUNX3, KLRG1, and TBX21), but also bound to and repressed genes involved in memory formation. In line with this, BATF-overexpressing cells showed more of an effector phenotype, especially in fresh or activated populations, while BATF-knockout cells were enriched for a central memory phenotype and memory T cell markers, especially after several rounds of tumor challenge. In vivo, TILs and peripheral blood from BATF-knockout CAR T cell-treated mice were both enriched for central memory cells, while those from mice treated with BATF-overexpressing cells were more terminally differentiated. By day 42, human CD3+ T cells could be detected in the peripheral blood of 5 out of 5 mice treated with BATF-knockout cells, but only 1 of the 5 mice treated with control CAR T cells, and none of the 5 mice treated with BATF-overexpressing CAR T cells, suggestive of enhanced persistence with BATF knockout.

Overall, these results help to clarify the role of BATF in orchestrating T cell activation, exhaustion, and differentiation. They also show that elimination of BATF can be used to enhance the functionality of CAR T cells for use against tumors, which could potentially translate to improved results in clinical applications in the future.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, lead author Haoyi Wang answered our questions.

From left to right, co-first author Chenze Zhang, lead author Haoyi Wang, and co-first author Xingying Zhang.



What was the most surprising finding of this study for you?

The most surprising finding of this study is that genetically modifying BATF leads to different, sometimes opposite functional outcomes, depending on the specific experimental setup. BATF promotes T cell proliferation, however, it drives T cell exhaustion as well. Therefore, under a non-exhaustion condition (high CAR T cell to tumor cell ratio), overexpressing BATF leads to better CAR T cell proliferation and efficacy. However, under an exhaustion-inducing condition, overexpressing BATF deepens CAR T cell exhaustion, reducing cell proliferation and efficacy. This explains the discrepancy in previous studies, and highlights that the interpretation of a gene’s function could be highly context-dependent.

What is the outlook?

In this study, we established a primary human CAR T cell exhaustion model, which demonstrates a profound exhaustion phenotype. Using this model, we are currently performing larger-scale genetic screens and drug screens to identify genes and small molecules that could be used to rescue T cell exhaustion. On the other hand, we are combining BATF depletion with other modifications, such as knocking out TGFBR2 or ADORA2A to further improve the efficacy of CAR T cells against solid tumors.

What was the coolest thing you’ve learned (about) recently outside of work?

I have recently become a fan of audio books. Using a popular audio book reading app, any book can be directly played in my car audio system. Therefore, I have been reading many books during my everyday driving. This really gives me an opportunity to finish the books I always wanted to read, but had no time to before. The books I most recently enjoyed reading are “In search of memory”, “Journey to the Edge of Reason: the Life of Kurt Gödel”, “Shantaram”, and “Neapolitan Novels”.

References:

Zhang X, Zhang C, Qiao M, Cheng C, Tang N, Lu S, Sun W, Xu B, Cao Y, Wei X, Wang Y, Han W, Wang H. Depletion of BATF in CAR-T cells enhances antitumor activity by inducing resistance against exhaustion and formation of central memory cells. Cancer Cell. 2022 Oct 7.

In the Spotlight...

Immune targeting of three independent suppressive pathways (TIGIT, PD-L1, TGFβ) provides significant antitumor efficacy in immune checkpoint resistant models

Franks et al. reported increased immunosuppressive TIGIT, PD-1, and TGFβ expression in the MC38-CEA colon cancer model, and demonstrated that the combination of anti-TIGIT with bintrafusp alfa (a bifunctional PD-L1 and TGFβ inhibitor) led to enhanced antitumor activity, increased overall survival, provided immunologic memory, and protected from rechallenge in MC38-CEA (colon) and TC-1 (lung) tumor models. The combination treatment showed increased immune cell infiltration into the tumors, an increased immune-activated cytokine and transcriptomic profile, and increased antitumor activity mediated by CD4+ and CD8+ T cells, but not NK cells.

Contributed by Shishir Pant

Cancer-specific T helper shared and neo-epitopes uncovered by expression of the MHC class II master regulator CIITA

Hos et al. showed that transfection of tumor cells lacking MHC-II with full-length human MHC-II transactivator (CIITA) cDNA induced stable expression of cell surface MHC-II and appropriate subcellular localization and function of processing machinery (CD74 and H2-DM) for MHC-II peptide presentation. MS analysis of peptides eluted from purified MHC-II molecules identified shared cancer oncoviral, and neo-epitope peptides, which when synthesized and presented by DCs, were recognized similarly to endogenous peptides by CD4+ T cells in vitro. Vaccination with adjuvanted synthetic peptides induced CD4+ T cell responses in tumor-bearing mice.

Contributed by Paula Hochman

Integrated TCR repertoire analysis and single-cell transcriptomic profiling of tumor-infiltrating T cells in renal cell carcinoma identifies shared and tumor-restricted expanded clones with unique phenotypes

Xu et al. demonstrated heterogeneity in T cell distribution and density in RCC, and identified local antigen-experienced clonally expanded T cells and shared populations among PBMC and/or normal adjacent tissue (NAT). Tumor restricted T cell clones displayed a Tem phenotype, with the highest expression of antigen-activation, exhaustion, and proliferation markers of any T cell subset within the TME. A subset of the Tem clones comprised CD4+CD8+ double positive T cells with a similar antigen-experienced phenotype. The clonotypes shared among tumor, NAT, and PBMC displayed a Teff phenotype with higher cytotoxic markers and lower activation and exhaustion markers.

Contributed by Shishir Pant

Post-translational modifications reshape the antigenic landscape of the MHC I immunopeptidome in tumors

To extend the useful immunopeptidome, Kacen and Javitt et al. established a pipeline (PROMISE) to identify the common post-translational modifications from mass spec data of HLA class I-eluted peptides. Analysis of the position of modifications demonstrated effects on peptide:HLA interaction, supported by modeling and binding data, including elucidation of new binding motifs, particularly involving modified cysteines. Some modification occurred in presumptive TCR recognition regions. Analysis of immunopeptidome data from a set of breast cancer tissues revealed potentially therapeutically useful tumor-specific changes, such as phosphorylation and cysteinylation.

Contributed by Ed Fritsch

BATF epigenetically and transcriptionally controls the activation program of regulatory T cells in human tumors

Itahashi and Irie et al. used multiple sequencing methods (ATAC, RNAseq, and sc versions) to profile Treg differentiation in the TME of NSCLC tumors. Compared to CD8+ and conventional CD4+ TILs, tumor-infiltrating Tregs displayed a unique open chromatin profile in which BATF was identified as a critical epigenetic and transcriptional regulator of Treg remodeling, differentiation, and activation. Deleting BATF in Tregs in the TME significantly delayed tumor growth in animal models, while high Treg BATF expression was associated with poor relapse-free survival in NSCLC, including PD-1-non-responders, and several other tumor types (lung, kidney and melanoma).

Contributed by Katherine Turner

Murine Allogeneic CAR-T Cells Integrated Before or Early After Posttransplant Cyclophosphamide Exert Anti-Tumor Effects

In an effort to directly integrate CAR T cell therapy with allogeneic hematopoietic stem cell transplant (allo-HCT) and post-transplantation cyclophosphamide (PTCy; given 3 and 4 days after allo-HCT to prevent GvHD). Patterson and Khan et al. tested different sequences and timings of the combination treatment in a T cell-replete, MHC-haploidentical mouse model. When given on the same day as allo-HCT (day 0) or 5 days after (but not 9 or 14 days after), CD19 CAR T cells could effectively clear leukemia without exacerbating CRS or compromising the function of PTCy. CAR T cells given on day 0 showed the most clinical efficacy, which was associated with a superior activation profile.

Contributed by Lauren Hitchings

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.