Weekly Digests
‹ Back to July

T<FR> are the new suppressive kids on the block for tumors

July 14, 2021

Regulatory T cells (Treg) can differentiate into follicular regulatory T cells (TFR) that have important roles in secondary lymphoid organs and may have higher suppressive activity. However, their role in antitumor immunity has not been elucidated. Eschweiler et al. investigated the role of TFR in cancer, analyzing their function, characteristics, and effects on the efficacy of checkpoint inhibition using RNAseq of human cancer cohorts and mouse models. Their results were recently reported in Nature Immunology.

The researchers began by analyzing nine published single-cell RNAseq datasets to assess CD4+ TIL in six human cancer types. Between 5 and 55% of the evaluated CD4+ TIL were FOXP3-expressing Tregs, and of these, 5-30% also expressed BCL6 and/or CXCR5, indicative of follicular lineage TFR cells. The presence of TFR was confirmed in tumor samples from patients with non-small lung cancer (NSCLC), where they were mainly found in tertiary lymphoid structures (TLS). TFR also expressed CD25 and ICOS and expressed the highest levels of CTLA-4 and PD-1 of all TIL.

Murine TFR obtained by immunizing mice with ovalbumin (OVA) and adjuvant had similar transcriptomes as Tregs, such as genes encoding heightened suppressive capacity, and were associated with CD8+ T cell dysfunction and survival, suggesting these cells have a suppressive potential. Performing bulk RNAseq on enriched subsets of Tregs and TFR TILs from human NSCLC revealed a cluster that correlated with the TFR phenotype. This cluster also expressed BCL6 and FOXP3, and was enriched for genes involved in the cell cycle, translational and transcriptional activity, TCF-1 expression, and mTOR signaling. These data suggest TFR are active and proliferative in the tumor microenvironment (TME).

The researchers then hypothesized that tumor antigen recognition might trigger conversion from Tregs to TFR within the TME. To analyze this, they sorted CD4+ and CD8+ populations from the tumor tissue and tumor-infiltrated lymph nodes from two patients with head and neck squamous cell carcinoma and subjected these cells to single-cell RNAseq and TCR sequencing. Two distinct FOXP3-expressing CD4+ clusters were detected, which had distinct transcriptomic signatures indicative of TFR and Tregs. TFR expressed higher levels of transcripts linked to metabolism, cell activation, co-stimulation, TFR function, suppressive capacity, and cell cycle than Tregs. 

There were also shared clonotypes between Treg and TFR, but TFR were more clonally expanded. A trajectory analysis suggested that conversion from Treg to TFR happened in the tumor. In a separate large single-cell RNAseq dataset of CD4+ TIL, most clonally expanded clonotypes of TFR were shared with Tregs. The Tregs that shared clonotypes with TFR expressed 4-1BB, suggestive of recent TCR activation. Genes linked to cell activation, co-stimulation, and suppression were expressed at higher levels in the 4-1BB+ TCR-sharing Tregs, and in the clonally expanded TFR. Additionally, TFR expressed lower levels of CCR7 and S1PR1, suggesting these cells have tissue-resident characteristics.

Eschweiler et al. then used the B16F10 melanoma and the MC38 colorectal tumor model system to assess tumoral TFR function. Similar to human cells, intratumoral TFR had increased Ki67, TCF-1, and 4-1BB expression compared to Tregs. In vitro and in vivo experiments showed that these cells were more suppressive than Tregs, inhibiting proliferation and reducing the secretion of IFNγ, IL-2, and TNFα by CD8+ T cells. Transferring OT-I T cells alone or with Tregs or TFR in B16F10-OVA tumor-bearing RAG1-knockout mice showed that TFR inhibited OT-I tumor cell killing, with only a limited effect of Tregs, further confirming the highly suppressive function of TFR.

Single-cell RNAseq of barcoded Foxp3+CD4+ T cells from B16F10-OVA tumor-bearing mice, at day 11 and day 18 of tumor growth, revealed five clusters. Only one cluster was enriched on day 18, and this cluster was enriched for genes previously linked to TFR, suggesting these cells increase in the tumor over time. Constructing a developmental path with cell-trajectory analysis revealed that TFR cells develop from a cluster of cells with naive, recirculating Treg features. To confirm this further, Eschweiler et al. conducted a time-course experiment, which showed that intratumoral TFR, but not Tregs, increased with tumor progression.

Like their human counterparts, murine TFR expressed high levels of CTLA-4 and PD-1. Therefore, these cells might respond to checkpoint inhibition therapy. Indeed, anti-PD-1 treatment increased the number of TFR in MC38 and B16F10 tumors, potentially opposing the benefit of PD-1 blockade. Re-analysis of published single-cell RNAseq data from patients who had received PD-1 blockade therapy suggested that TFR were enriched for T cell activation and costimulation transcripts after therapy. To assess the specific inhibitory effects of TFR, apart from any Treg inhibition, the authors used a genetic knockdown system to selectively deplete TFR, and found that knockdown increased treatment efficacy.

To further validate the role of TFR in tumor development and its effects on anti-PD-1 treatment, the researchers used the B16F10 tumor model, which is known to not respond to anti-PD-1 therapy, and modified it to be TFR-deficient (Foxp3cre- Bcl6fl/fl). Without TFR, tumor growth was less robust, and anti-PD-1 therapy further increased tumor control, suggesting TFR indeed decrease the efficacy of anti-PD-1 therapy. CD8+ T cells in the lymph nodes of these TFR-deficient mice had higher expression of granzyme B, suggesting TFR may inhibit CD8+ T cell function in the lymph nodes. A competition assay showed that BCL-6 was required for the tissue persistence of Foxp3+ cells in the TME.

Given its inhibiting effects on anti-PD-1 therapy, the researchers hypothesized (1) that depletion of TFR before treatment might enhance efficacy and (2) that given the high expression levels of CTLA-4 on these cells, blockade of CTLA-4 might deplete TFR. Monotherapy of anti-CTLA-4 indeed depleted TFR preferentially over Tregs, and sequential treatment with anti-CTLA-4 followed by anti-PD-1 resulted in a reduction in tumor volume compared to either monotherapy, and elevated the frequency of CD8+ TIL and granzyme B+ CD8+ and CD4+ T cells in the B16F10-OVA model.

Finally, retrospective analysis of survival of patients with unresectable melanoma treated with either anti-CTLA-4 or anti-PD-1 monotherapy, simultaneous combination therapy, or sequential anti-CTLA-4 and anti-PD-1 therapy showed that patients in the sequential treatment group had the best outcome. In addition, a cohort of patients with melanoma showed that those who had poor survival and did not respond to anti-PD-1 treatment had more FOXP3+BCL-6+CD4+ TIL, indicative of TFR

Together, these data from Eschweiler et al. suggest a significant role of TFR in cancer, the presence of TFR in TLSs could potentially predict which patients to treat with sequential checkpoint inhibition therapy, and that TFR may be a new target for immunotherapy.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Eschweiler S, Clarke J, Ramírez-Suástegui C, Panwar B, Madrigal A, Chee SJ, Karydis I, Woo E, Alzetani A, Elsheikh S, Hanley CJ, Thomas GJ, Friedmann PS, Sanchez-Elsner T, Ay F, Ottensmeier CH, Vijayanand P. Intratumoral follicular regulatory T cells curtail anti-PD-1 treatment efficacy. Nat Immunol. 2021 Jun 24

In the Spotlight...

Resident Kupffer cells and neutrophils drive liver toxicity in cancer immunotherapy

Siwicki et al. used a CD40 agonist mouse lung tumor model to investigate TH1-driven IL-12 and IFNγ toxicity in non-tumor-bearing liver tissue. In tumor-bearing mice treated with agonist CD40, liver-resident Kupfer cells were activated by increased levels of immune cell-produced IFNγ, stimulating IL-12 production and cooperative augmentation of liver damage. Dendritic cells were required for tumor control, but were not responsible for toxicity. Mechanistically, IFNγ, IL-12, and macrophages induced a toxic neutrophil response that correlated with tissue damage. Similar pathways were observed in humans treated with immunotherapy (anti-PD-1 or anti-PD-1 plus anti-CTLA-4).

Contributed by Katherine Turner

Antitumor efficacy and reduced toxicity using an anti-CD137 Probody therapeutic

Immunostimulatory agonist antibodies to CD137 have antitumor activity in preclinical models, but concomitant serious liver inflammation has precluded clinical dosing to therapeutic levels. Etxeberria et al. report on Pb-Tx-1D8, (anti-CD137 Probody Therapeutic), a prodrug antibody that recognizes CD137 only after proteolytic cleavage (by proteases present in TMEs) of a peptide coupled to the antigen binding site. Pb-Tx-1D8 had antitumor activity comparable to conventional anti-CD137, but liver adverse effects (as measured by ALT, CD8+ T liver infiltration, and extravascular T cell clusters) were reduced. Synergy was observed with anti-PD-1.

Contributed by Margot O’Toole

Combination of radiation therapy, bempegaldesleukin, and checkpoint blockade eradicates advanced solid tumors and metastases in mice

Pieper et al. treated tumor-bearing mice with local radiation therapy (RT) and bempegaldesleukin (BEMPEG), a PEGylated, CD122-engaging IL-2 receptor agonist. Compared to the single therapies, this combination improved tumor control in B78 melanoma, MOC2 head and neck carcinoma, and 4T1 breast cancer models. RT of B78 tumors increased CD122 expression on splenic T cells and, with BEMPEG, CD8+ T cell tumor infiltration; depleting T cells eliminated antitumor efficacy. Adding immune checkpoint blockade (anti-CTLA-4 or anti-PD-L1) to the combination treatment further suppressed larger tumors and metastases.

Contributed by Alex Najibi

CRASH-IT switch enables reversible and dose-dependent control of TCR and CAR T-cell function

Sahillioglu et al. engineered a small molecule-regulated genetic safety switch (CRASH-IT), with three functional units, that induces proximity to antigen receptor (Zap70, SH2 domains), provides inhibitory signals (PD-1 tail), and regulates the strength of inhibitory signals (SMASh tag or FKBP12F36V domain). In the absence of asunaprevir, HCV protease cleaves the linker in the Zap70–PD-1-SMASh fusion protein, which stops Zap70–PD-1 fusion degradation, resulting in inhibition of T cell activation. CRASH-IT demonstrated dose-dependent recovery of T cell function, reversibility, and potential to be combined with endogenous/transduced TCR or CARs.

Contributed by Shishir Pant

Memory-like differentiation enhances NK cell responses to melanoma

Marin, Krasnick, and Becker-Hapak et al. showed that in patients with advanced melanoma (AM), mostly immature NK cells expressing low levels of perforin and granzyme B infiltrated uninvolved lymph nodes and metastases, and blood NK cells expressed low levels of NK activating receptors. IL-12/15/18 induction of memory-like (ML) NK cells from the blood of healthy donors and patients with AM boosted NK cell lysis of melanoma cells. Blockade of NKG2D and NKp46 activating receptors reduced melanoma cell lysis by autologous ML NK cells. In immunodeficient mice, a human melanoma cell line was better controlled by transferred human ML than control NK cells.

Contributed by Paula Hochman

Immunogenomics of colorectal cancer response to checkpoint blockade: analysis of the KEYNOTE 177 trial and validation cohorts

Bortolomeazzi, Keddar, and Montorsi et al. integrated multiomics, histopathologic, and immune-phenotypic data from 738 tumor regions from 29 CRC patients prior to anti-PD-1 therapy to determine the molecular factors associated with the response. TMB below 12 mutations/Mbp predicted resistance to therapy, however, higher TMB failed to predict response. In high TMB patients, response was associated with high clonality of immunogenic mutations, higher cytotoxic and proliferating PD1+CD8+ T cells, low WNT signaling, and higher levels of CD74+PD-L1+ macrophages, which co-located with PD-1+CD8+ T cells and, surprisingly, low B2M expression.

Contributed by Shishir Pant

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.