Weekly Digests
‹ Back to September

The inhibitory signaling receptor protocadherin-18 regulates tumor-infiltrating CD8+ T cell function

September 20, 2017

The activation of T cells from a memory to an effector state depends on the integration of the positive signal from the T cell receptor upon binding with a cognate antigen and the negative signals from any of several inhibitory signaling receptors (ISRs) binding their cognate ligands on the antigenic cell’s surface. ISRs, typified by receptors like PD-1 and CTLA-4, downregulate TCR-mediated signal transduction and raise the threshold of immune activation, preventing the effector phase. In this study, Alan Frey identified protocadherin-18 (pcdh18) as a novel ISR expressed in memory T cells. The only known ligand to pcdh18 is itself, which functions in trans.

In a previous study by the Frey lab, pcdh18 was found to be expressed on dysfunctional tumor infiltrating lymphocytes (TILs) from MCA38 colon tumor-bearing mice. These TILs lacked lytic function against pcdh18-expressing MCA38 cells immediately after isolation, however, their function could be restored after brief culture, suggesting that the interaction with tumor cells was related to their dysfunction. In this new work, Frey used shRNA to knock down pcdh18 on tumor cells, and TILs from the tumors were functional immediately upon isolation. Similarly, in mice engineered to lack pcdh18 expression on T cells (pcdh18-/-), tumor growth was significantly delayed compared to wild type mice, suggesting that pcdh18-/- T cells were more effective against pcdh18-expressing MCA38 tumors than pcdh18+ T cells. Together, this data confirms Frey’s hypothesis that pcdh18 on tumor cells (acting as ligand) initiates the inhibitory function of pcdh18 on T cells (acting as receptor).

To determine why tumor growth was only delayed, and not entirely halted in pcdh18-/- mice, Frey analyzed the gene expression of TILs isolated from these animals. While their gene expression profiles showed significant upregulation of immune function genes related to T cell activation, signaling, effector functions, and chemokine and cytokine receptors, they also showed increased expression of genes encoding other known negative regulators, including PD-1 and CTLA-4.

Since PD-1 is highly expressed on wild-type TILs, and further upregulated on TILs from pcdh18-/- mice, Frey tested the addition of anti-PD-1 therapy in both wild type and pcdh18-/- models. In wild type mice, anti-PD-1 delayed tumor growth, but only when administered at an early stage (day 5). In pcdh18-/- mice, the addition of anti-PD-1 further inhibited tumor growth, even if treatment was delayed (to day 10). TILs from the combination group also produced slightly more IFNγ than those treated with PD-1 blockade alone. Depletion of CD8+ T cells abrogated the effect of anti-PD-1 in both mouse models, indicating their essential role in mediating the delay of tumor growth.

While genetically knocking out pcdh18 helped prove its role as an ISR, Frey developed and tested a murine anti-pcdh18 serum in anticipation of the development of a clinical therapy. As monotherapies, anti-PD-1 was still more effective than anti-pcdh18, which only slightly delayed tumor growth. Combining the two antibody therapies, however, was much more effective than the use of either as a single agent checkpoint blockade, suggesting anti-pcdh18 could be used to improve the efficacy of anti-PD-1 therapies.

In a translational study, Frey explored the expression of pcdh18 in human T cells. Immunohistochemistry of a normal human lymph node showed regions where low proportions (~5%) of both CD4+ and CD8+ cells expressed pcdh18. Using freshly isolated peripheral blood mononuclear cells and fluorescence-activated cell sorting, high pcdh18 expression was observed in CD4+ and CD8+ effector memory T cell populations, and conversion of central memory CD8+ T cells to an effector memory phenotype demonstrated upregulation of pcdh18 protein. Finally, in vitro activation of human CD8+ effector memory T cells coincided with an increase in pcdh18 expression and loss in viability; anti-pcdh18 sera rescued viability and restored INFγ expression.

The expression of pcdh18 in human memory T cells further supports its role as an ISR with regulatory control over the effector phase, opening the door to its potential as a clinically-relevant target for immunotherapy, especially in conjunction with anti-PD1 therapy.

by Lauren Hitchings

References:

Frey A.B. The Inhibitory Signaling Receptor Protocadherin-18 Regulates Tumor-Infiltrating CD8+ T-cell Function. Cancer Immunol Res. 2017 Sep 5.

In the Spotlight...

Administration of low-dose combination anti-CTLA4, anti-CD137, and anti-OX40 into murine tumor or proximal to the tumor draining lymph node induces systemic tumor regression

Hebb et al. demonstrate that direct intratumoral injection of agonist OX-40, agonist CD137, and anti-CTLA-4 antibodies produce a profound abscopal effect at a fraction of the dose used systemically, curing most mice in two tumor models. Moreover, s.c. injection of the combination treatment in the proximity of (1) the tumor, (2) the tumor draining lymph node (TDLN), or (3) a non-TDLN with irradiated tumor cells produced nearly similar results.

De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors

Low doses of a fusion protein comprised of LIGHT (a TNF-related cytokine) and a vascular targeting peptide (VTP) reversed tumor vessel abnormalities in mice with pancreatic tumors and enabled T cell infiltration into the tumor via LIGHT-VTP-stimulated macrophages. Combination of LIGHT-VTP with a vaccine and dual checkpoint blockade (anti-PD-1 and anti-CTLA-4) significantly reduced tumor growth, induced immune memory response, and increased survival.

Multipeptide-coupled nanoparticles induce tolerance in 'humanised' HLA-transgenic mice and inhibit diabetogenic CD8+ T cell responses in type 1 diabetes

The other side of the coin to increasing T cell responses for cancer therapy is reducing T cell responses in autoimmunity, and the mechanisms leading to such reduction may be highly relevant to immunosuppression in cancer. Xu et al. show that i.v. treatment of NOD/HLA-A02:01 mice with polystyrene bead nanoparticles coupled to epitope peptides relevant to human diabetes prevented onset of diabetic symptoms and extended survival. Xu et al. link marginal zone macrophage phagocytosis, CCL2 production, and recruitment of Treg cells and CD8+CD103+ dendritic cells to the mechanism of action.

CD155T/TIGIT Signaling Regulates CD8+ T Cell Metabolism and Promotes Tumor Progression in Human Gastric Cancer

The functionally exhausted CD8+ T cells expressing the immune checkpoint receptor TIGIT in gastric cancer patients had impaired glucose uptake due to the interaction between tumor-expressed CD155 and TIGIT. This metabolic impairment was reversed in vitro by TIGIT blockade or the addition of glucose. The combined blockade of TIGIT and PD-L1 in mice suppressed tumor progression and increased survival.

A TCR-based Chimeric Antigen Receptor

Walseng et al. fused a soluble TCR construct to a CAR-signaling tail, resulting in the aptly named TCR-CAR. The TCR portion maintained the advantage of specifically targeting epitopes presented on MHC molecules, while the CAR signaling portion allowed for the transduction of cell types beyond just T cells. The functionality (cytokine release, degranulation, cell killing) of TCR-CARs was demonstrated in T and NK cells.

Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment

Yeku et al. delineated the mode of action of Muc-16-directed “armored” CAR T cells that constitutively secrete IL-12. In an aggressive murine disseminated ovarian cancer model, these cells maintained efficacy, demonstrated enhanced antitumor function, and improved overall survival. The armored CAR T cells evaded immune suppression by phenotypic skewing and Fas/FasL-dependent depletion of tumor-associated macrophages, and by resisting inhibition by PD-L1.

Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy

In a 21-patient, phase 1b clinical trial evaluating the safety of the combination of intratumoral injections of oncolytic virus talimogene laherparepvec with i.v. administered anti-PD-1 (beginning on week 6) for the treatment of advanced melanoma, the generally well-tolerated therapy resulted in 62% confirmed objective response rate and 33% complete response rate, likely due to the virus-induced increase in CD8+ T cells, PD-L1 expression, and IFNγ expression within the tumor.

NF-kappaB c-Rel Is Crucial for the Regulatory T Cell Immune Checkpoint in Cancer

Grinberg-Bleyer et al. found that the c-Rel subunit of canonical nuclear factor κB (NF-κB) plays a critical role in the identity and function of activated Tregs. Genetic knockdown of c-Rel decreased expression of Treg activation genes, and reduced melanoma growth in mice by impairing activated Treg-mediated immunosuppression of CD8+ T cell responses. Use of the FDA-approved drug pentoxifylline to knock down c-Rel showed antitumor efficacy in vivo, especially when combined with anti-PD-1.

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.