Weekly Digests
‹ Back to May

RIGging the game: LRIG1 induces CD8+ T cell quiescence by engaging VISTA

May 29, 2024

Immune checkpoint inhibitors have revolutionized the cancer immunotherapy field, and V domain immunoglobulin suppressor of T cell activation (VISTA) has garnered attention as a potentially relevant immune regulatory molecule. In preclinical models, VISTA blockade has promoted CD8+ T cell infiltration and inflammatory cytokine secretion in tumors, leading to antitumor efficacy. VISTA appears to function both in cis (between T cells) and in trans (between APCs and T cells), but the cognate receptor(s) are not known. In a recent publication in Science Immunology, Ta et al. identified LRIG1 as an engager of VISTA that controlled antitumor immunity by constraining T cell proliferation and effector function.

Ta et al. took a direct approach for receptor identification by using selective proteomic labeling proximity ligation assay using tyramide (SPPLAT) on wild-type murine splenic T cells or T cells knocked out for VISTA. Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) was identified as a binder of VISTA, and the result was validated in T cells and HEK293T cells using co-immunoprecipitation and SPPLAT. The binding affinity of the extracellular domains was elucidated by surface plasmon resonance (~1 nM), with minimal variation between neutral and acidic conditions (PSGL-1 had previously been identified as a receptor for VISTA, but only under acidic conditions). Split-luciferase-tagged VISTA and LRIG1 were used to probe the relevance of binding in cis and in trans. Luciferase activity was observed in T cell cultures and in a B16OVA model, suggesting binding partnership between T cells in cis. Furthermore, luciferase activity was observed when LRIG1-labeled T cells were exposed to VISTA-labeled DCs in vitro and tumors in vivo, yet was abrogated in absence of VISTA, suggesting that LRIG1 can also act in trans.

LRIG1 was not detected on naive T cells, yet was induced following activation and increased in response to TCR stimulation. Given that LRIG1 is activation-inducible, the authors sought to understand its impact on T cell activation and signaling. Co-expression of VISTA and LRIG1 in T cells, but neither alone, inhibited phosphorylation of downstream TCR signaling proteins upon stimulation with anti-CD3/CD28 antibodies. Conversely, knockout of LRIG1 (Lrig1-/-) or VISTA (Vsir-/-) enhanced phosphorylation of signaling molecules, suggesting that ligation of VISTA with LRIG1 constrained intracellular TCR signaling. Consequently, activated Lrig1-/- T cells exhibited up to 3-fold greater proliferation and IFNγ secretion compared to wild-type (WT) T cells, along with a significant reduction in apoptosis. Similar trends, including increased expression of the costimulatory molecule CD28, were observed when Lrig1-/- cells were stimulated in an antigen-specific manner by OVA peptide-loaded DCs.

To parse the effects of cis and trans VISTA engagement on T cell phenotype, the authors generated VISTA knockout (Vneg) and VISTA-overexpressing (VOE) DC2.4 cells and cultured them with T cells. In culture with Vneg DCs (where only T cell-derived VISTA was present), Lrig1-/- T cells proliferated more substantially than WT T cells, suggesting that cis engagement plays a role in VISTA/LRIG1-mediated immune suppression. In the presence of VOE DCs, WT T cells failed to expand, while Lrig1-/- T cells expanded vigorously, suggesting remediation of the inhibitory effects of VISTA signaling in trans. Vsir-/- T cells (hence unable to engage T cell LRIG1 in cis) failed to proliferate in culture with VOE DCs, confirming the contribution of trans signaling. To confirm the contributions of cis and trans signaling in the TME, where other putative binding partners of VISTA could be present, VOE and Vneg MC38-OVA tumor cells were generated, and 1:1 mixtures of WT and Lrig1-/- T cells were administered. WT T cells failed to expand in VOE tumors while Lrig1-/- T cell expansion was unaffected, suggesting that trans VISTA signaling plays a role in vivo and that knockout of LRIG1 conferred resistance to this mode of suppression.

Next, the authors sought to examine the therapeutic impact of LRIG1 deletion for cancer immunotherapy. To elucidate the effects of LRIG1 deficiency on TIL proliferation and phenotype in a standard tumor model, WT and Lrig1-/- T cells were adoptively transferred and profiled in B16-OVA tumors. Lrig1-/- T cells robustly expanded after 1 week, while WT T cells steeply declined, leading to a >10-fold difference in accumulation by 2 weeks. Gene set enrichment analysis (GSEA) of TILs indicated enrichment of pathways and genes related to cell cycle regulation in Lrig1-/- cells. Next, tumor growth was compared in WT and Lrig1-/- mice, where antitumor effects would be mediated by endogenous T cells. Lrig1-/- mice experienced significant tumor control compared to WT mice bearing B16-OVA tumors, with TILs upregulating Ki67, IFNγ, and CD28, and being more resistant to cell death. In the non-OVA transfected B16 melanoma model, treatment with a TLR agonist/peptide vaccine furthered tumor control, while addition of CTLA-4 blockade led to 80% rejection and 76% resistance to rechallenge. Lrig1-/- mice exhibited a greater frequency of TILs, including those specific for the tumor antigen TRP2, along with increased CD28 and Ki67 expression and superior IFNγ expression upon peptide restimulation ex vivo.

Single-cell RNA sequencing (scRNAseq) was conducted to further understand the nature of CD8+ TILs and their modulation by LRIG1 deficiency. CD8+ CTLs separated into 12 clusters; 6 clusters expressed Tcf7 (the gene for TCF1), indicating stemness, while the remaining 6 likely represented cells in terminally differentiated states. The C1 cluster, which expressed Tcf7, but lacked both effector genes and inhibitory receptors, was significantly enriched in WT mice compared to Lrig1-/-. This result was validated by flow cytometry and in an adoptive transfer model with Pmel-specific T cells, further indicating that the quiescent state could be induced in activated tumor-specific T cells, and was not merely a consequence of naive T cell infiltration. The reduction in quiescent Lrig1-/- T cells was accompanied by an increase in the frequency of Tpex cells, which exhibited superior responses to peptide stimulation.

To interrogate the relevance of LRIG1 in human cancers, the authors first showed that LRIG1 was induced upon T cell restimulation in patient samples, while VISTA expression was unchanged. They then investigated a melanoma scRNAseq dataset, where a T cell cluster resembling the quiescent C1 cluster in mice was identified. LRIG1 was broadly expressed across T cell clusters in patients that failed to respond to ICB, while expression was more restricted to exhausted cells in responding patients. CD8+ TILs of non-responding patients were enriched for LRIG1, suggesting a role for LRIG1 in the regulation of human T cell effector function.

Overall, these findings identify LRIG1 as a co-inhibitory T cell receptor that functions by binding VISTA. VISTA engaged LRIG1 both in trans and in cis to block TCR signaling pathways, which led to diminished proliferation, CD28 expression, and cytokine secretion. In vivo, LRIG1 knockout greatly enhanced TIL persistence and cytotoxicity, preventing acquisition of a quiescent phenotype and promoting tumor control. LRIG1 expression in human TILs was associated with response to checkpoint blockade, suggesting that blockade of the LRIG1/VISTA axis may be an attractive and practical option for cancer immunotherapy.

Write-up by Morgan Janes, image by Lauren Hitchings

Meet the researcher

This week, senior author Lily Wang answered our questions. 

Senior author Lily Wang


What was the most surprising finding of this study for you?
There are pretty clear phenotypes of the LRIG1 KO T cells when examined in vitro, including better proliferation, better survival, and better cytokine production. Although these were sort of “expected” based on the hypothesis, the most surprising data was from in vivo experiments in murine tumor models: LRIG1 KO mice transiently delayed tumor growth… the long-term tumor-free survival was achieved when in conjunction with a peptide vaccine therapy. This result suggests that we need to continue to explore new combinatorial therapeutic strategies to completely eradicate cancer and improve survival benefit.

What is the outlook?
Outside of T cells, LRIG1 has been studied in cancer cells and stem cells. But for T cell biology, LRIG1 is a newly discovered coinhibitory receptor and a new player in regulating antitumor T cell responses. We are only seeing the tip of the iceberg. Many outstanding questions remain to be addressed. For example: does LRIG1 have other ligands besides VISTA?, what signaling adaptors work with LRIG1 to change T cell function and fate?, is there a role of LRIG1 in other immune cell types?, how is LRIG1 expression is regulated?, and can LRIG1-targeted inhibitors be developed for cancer therapy?.
Like other inhibitory immune checkpoint receptors, the biology of LRIG1 may be relevant to not only cancer immunotherapy, but also for autoimmune and inflammatory disease. LRIG1 antagonists and agonists may be suitable for treating various immune dysfunctions. This study opens a door to an exciting new direction of research in tumor immunology and other diseases with immune dysfunctions.

What was the coolest thing you’ve learned (about) recently outside of work?
VISTA is also a Visible and Infrared Survey Telescope for Astronomy: “VISTA” has been gazing deep into the cosmos for decades. We have been studying the “VISTA” molecule for more than a decade (the initial discovery was made in 2011). I feel that studying VISTA is like a deep gaze into the mystery and wonder of the cosmos of biology.

References:

Ta HM, Roy D, Zhang K, Alban T, Juric I, Dong J, Parthasarathy PB, Patnaik S, Delaney E, Gilmour C, Zakeri A, Shukla N, Rupani A, Phoon YP, Liu C, Avril S, Gastman B, Chan T, Wang LL. LRIG1 engages ligand VISTA and impairs tumor-specific CD8+ T cell responses. Sci Immunol. 2024 May 17.

In the Spotlight...

PD-1 blockade plus COX inhibitors in dMMR metastatic colorectal cancer: Clinical, genomic, and immunologic analyses from the PCOX trial

Wu et al. report that in a phase 2 clinical trial, treatment with a COX inhibitor increased the efficacy of PD-1 blockade in patients with DNA mismatch repair deficient (dMMR) metastatic colorectal cancer. The combination therapy was safe and well tolerated. Out of 30 patients, the ORR was 73.3%, meeting its predefined target; the disease control rate was 93.3%. Whole-exome and RNA sequencing demonstrated that the antigen processing and presentation pathways were associated with treatment response. High TAPBP expression was associated with improved PFS and OS in patients with dMMR CRC treated with ICIs.

Contributed by Shishir Pant

A RIPK1-specific PROTAC degrader achieves potent antitumor activity by enhancing immunogenic cell death

Mannion et al. developed a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated acute RIPK1 deletion primed TNFR1 and TLR3/4 pathways to overactivate NF-κB, MAPK, and IFN signaling, and simultaneously promoted RIPK3 activation and necroptosis induction. RIPK1 depletion sensitized cancer cells to radiation therapy-induced TNF and interferons, promoted immunogenic cell death, drove long-lasting antitumor immunity, and enhanced the immunostimulatory effects of radio- and immunotherapy.

Contributed by Shishir Pant

The CD8+ T cell tolerance checkpoint triggers a distinct differentiation state defined by protein translation defects

Using a mouse model of peripheral tolerance, Van Der Byl, Nussing, and Peters et al. showed that soon after in vivo exposure to self-antigen, self-reactive CD8+ T cells expressed phenotypic, transcriptomic, and epigenetic profiles that minimally overlapped with CD8+ naïve T, TEFF and TEX cells, and resembled aberrantly primed CD8+ T cells from tumor-reactive TDLNs. Self-reactive CD8+ T cells prematurely exited the cell cycle, and their differentiation toward tolerance diverged early from that of CD8+ TEFF cells. Breaking CD8+ T cell tolerance required synergistic robust TCR and inflammatory signals that induced MYC-dependent protein translation.

Contributed by Paula Hochman

A lactate-SREBP2 signaling axis drives tolerogenic dendritic cell maturation and promotes cancer progression

Plebanek et al. identified a cDC-derived, CD63+ mature regulatory DC (mregDC) population in mouse TDLNs that potently inhibited cDC cross-presentation and induced Treg formation. mregDCs exhibited unique transcriptional and metabolic signatures associated with the mevalonate (MVA) pathway, including elevated FAO and cellular lipids. Inhibition/KO of the MVA regulator SREBP2 promoted tumor control, improved the TDLN cDC1/mregDC ratio, and increased MHC-I expression by DCs. The presence of lactate in the TME enhanced mregDC abundance in a SREBP2-dependent manner, leading to diminished tumor control.

Contributed by Morgan Janes

Anti-4-1BB×PD-L1 bispecific antibody reinvigorates tumor-specific exhausted CD8+ T cells and enhances the efficacy of anti-PD-1 blockade

Jeon, You, and Park et al. detected the highest 4-1BB expression on terminally exhausted tumor-specific CD8+ TIL in human melanoma and hepatocellular carcinoma. These cells did not respond to pembrolizumab treatment, but ABL503, an anti-4-1BB×PD-L1 bispecific antibody, increased their in vitro proliferation, which was further enhanced in combination with anti-PD-1. While either monotherapy induced some tumor growth delay in a 4-1BB-, PD-1-, and PD-L1-humanized murine tumor model, combination treatment of anti-PD-1 and ABL503 further delayed tumor growth, with increased TIL density and activation of CD8+ TIL.

Contributed by Maartje Wouters

PVRIG is expressed on stem-like T cells in dendritic cell-rich niches in tumors and its blockade may induce immune infiltration in non-inflamed tumors

Alteber and Cojocaru et al. found that the inhibitory receptor PVRIG was mostly detected on stem-like memory T cells, and its ligand PVRL2 was highly expressed on all tumoral DC subsets. LAMP3+PVRL2+ mregDCs were found to interact with CD28+PVRIG+CD8+ T cells in tumoral immune aggregates. Triple therapy with blockade of PVRIG, PD-1, and TIGIT in patients with solid tumors resulted in immune activation, including increased CD8+ T cell proliferation, increased TCR clonality, and expansion of pre-existing clones. Combination therapies with PVRIG blockade induced these effects in patients with “immune desert” tumors.

Contributed by Maartje Wouters

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.