Weekly Digests
‹ Back to March

TIGIT and PD-1 join forces to inhibit CD226 costimulation

March 16, 2022

Combination immune checkpoint blockade (ICB) therapies have been successful in the clinic, but the exact mechanisms of their synergy are not completely understood. For example, co-inhibition of PD-1 and TIGIT can be superior to either alone, although the distinct contributions of each to antitumor immunity remain unclear. Recently reported in Immunity, Banta and Xu et al. provided a mechanistic view into the observed synergy between these pathways.

Previously, it was found that antibody blockade of CD226, a costimulatory T cell marker regulated by TIGIT, reduced the efficacy of dual anti-PD-L1 and anti-TIGIT treatment, indicating a relationship between PD-1 and CD226. Confirming this interplay, the authors found that early after implantation, CT26 colon carcinoma-bearing Cd226-/- mice, but not wild-type mice, were incapable of responding to either anti-PD-1 or anti-TIGIT monotherapy. In addition, CD8+ T cells from Cd226-/- mice showed inferior killing of antigen-expressing target cells ex vivo. The loss of CD226 ligands PVR/PVRL2 from target cells also reduced killing by wild-type CD8+ T cells, emphasizing a role of CD226 costimulation in CD8+ T cell functionality.

With the importance of CD226 in ICB response established in a mouse model, the researchers questioned whether CD226 may also be important in patient ICB efficacy. Analyzing bulk RNAseq data from three clinical trials of atezolizumab (anti-PD-L1) in non-small cell lung carcinoma (NSCLC), they found that CD226 expression correlated positively with overall and progression-free survival. The expression of other costimulatory genes, particularly CD28, PDCD1, and TIGIT, among others, did not display this correlation, suggesting a unique role of CD226 in the patient immunotherapy response.

Next, the team investigated the phenotypes of CD226- and CD28-expressing CD8+ T cells within NSCLC tumors using CyTOF. As expected, CD226 and CD28 were primarily expressed on T cells. Dividing these cells into four populations based on their CD226 and CD28 co-expression, differences in phenotypic marker expression were observed. Although the double-positive CD28+CD226+ population was the rarest, it expressed high CD27, associated with antigen-specific expansion and memory, and low TIM-3 and CD103, compared to the CD28- populations. The two CD28+ populations displayed a similar profile.

To analyze T cell phenotypes more rigorously, the authors used scRNAseq, investigating CD8+ T cells from NSCLC tumors and adjacent healthy tissue. Eight distinct CD8+ T cell clusters were uncovered, matching previously characterized phenotypes (e.g., effector, resident memory). In both tumors and healthy tissue, CD226 and CD28 were variably expressed among these distinct clusters. Interestingly, CD226 and CD28 were most strongly co-expressed in proliferative (MKI67+) and cytotoxic memory (KLRB1+) CD8+ T cell states, likely to be involved in immunotherapy response.

Next, the authors probed more precisely how PD-1, TIGIT, and CD226 interact on T cells. They established a co-culture system between CD226+ Jurkat T cells, which could be further transduced with PD-1 or TIGIT, and antigen-expressing Raji lymphoma cells expressing PD-L1, PVR (TIGIT ligand), or both. CD226 phosphorylation was used as a readout of CD226 activity. Interestingly, the co-expression of either PD-1 or TIGIT on CD226+ Jurkat cells decreased CD226 phosphorylation, indicating that each molecule can inhibit CD226, and this inhibition was maximized when both were expressed. CD226 phosphorylation could be restored by antibody blockade of either PD-1 or TIGIT, but most effectively with dual blockade, altogether demonstrating that PD-1 and TIGIT both inhibit CD226 activity in an additive manner.

Investigating which portion of these receptors mediated CD226 inhibition, the authors found that the intracellular domain (ICD) of PD-1 was required to block CD226 phosphorylation, as a mutant PD-1 lacking the ICD did not induce this effect. However, this was not the case for TIGIT, which could still inhibit CD226 activity without its ICD. A FRET-based assay found that the PD-1 ICD recruited phosphatases Shp2 and Shp1, among other T cell signaling proteins, while the TIGIT ICD did not, potentially indicating that the recruitment of these phosphatases (or associated molecules) was responsible for the dispensability of the TIGIT ICD in CD226 inhibition.

Because TIGIT’s ICD was not required for CD226 inhibition, the authors considered how the extracellular domain of TIGIT might be responsible. They hypothesized that TIGIT could simply outcompete CD226 on the cell surface for its ligand PVR. Testing this theory, they co-cultured CD226+TIGIT+ Jurkat cells with Raji cells expressing PVR at either a high or low density. Theoretically, if PVR ligand availability was the limiting factor for CD226 activation, CD226 phosphorylation should be reduced more strongly in the low-PVR case. Indeed, the researchers observed reduced CD226 phosphorylation at the lower PVR concentration, suggesting that the TIGIT extracellular domain outcompetes CD226 for its ligand.

Finally, the authors used confocal microscopy to visualize the Jurkat-Raji cell interface. CD226 localized to the interface, but less so when TIGIT was co-expressed (which was also enriched at the interface). CD226 polarization could be restored by TIGIT antibody blockade, and as previously observed, only the TIGIT extracellular domain was required for CD226 interference.

In summary, the authors found that both PD-1 and TIGIT restrict CD226 costimulation on CD8+ T cells, but through different mechanisms: PD-1 by recruiting inhibitory phosphatases to its intracellular domain, and TIGIT by extracellular competition for CD226 ligands. Coupled with phenotypic analysis of CD8+ T cells coordinately expressing both CD226 and CD28, these findings shine light on a novel mechanistic underpinning for this clinically encouraging combination ICB therapy. Additional exploration of these mechanisms may support the further development of effective, non-redundant combination immunotherapy strategies for cancer treatment.

Write-up by Alex Najibi, image by Lauren Hitchings

Meet the researcher

This week, senior author Ira Mellman answered our questions.

From left to right, Karl Banta, Ira Mellman, and Eugene Chiang in front of the South San Francisco Bay.


What prompted you to tackle this research question?
The rationale really goes back to the early days of PD-1/PD-L1. The field began with the understandable assumption that checkpoint inhibitors acted by reversing T cell exhaustion in the tumor. Expecting this was the case, we were surprised in our early mechanistic and clinical biomarker studies of atezolizumab that this appeared not to be the case, rather that PD-L1/PD-1 blockade had a marked effect on T cell function much earlier in the pathway, namely at or around the so-called Tscm compartment, which was activated as a consequence of dendritic cell stimulation. TIGIT, along with PD-1, appeared to be present on these early T cells, or at least T cells poised for activation and proliferation. So, TIGIT’s mechanism also became of interest. In addition, given pre-clinical and now clinical data attesting to the positive benefit of combining PD-L1/PD-1 blockade with anti-TIGIT, we became interested in understanding how the two pathways interacted and why combinations were generally more effective than PD-L1/PD-1 inhibition alone. Further work on TIGIT is indicating also that there remains a lot more to learn about how this molecule works, and the significance of signaling via CD226 in various cell types.

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

It was unexpected to find that there was such a close mechanistic relationship between the PD-L1/PD-1 and TIGIT pathways. For a variety of reasons, this appears to “make sense”, especially given human genetics indicating that T cell immunity is not wholly dependent on CD28 costimulation, implicating CD226 as playing a more important role than we previously thought.

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

Learning how important Draymond Green is to the success of the San Francisco Warriors. So much else learned of late about the behavior of our fellow human beings has been profoundly depressing.

References:

Banta K.L., Xu X., Chitre A.S., Au-Yeung A., Takahashi C., O'Gorman W.E., Wu T.D., Mittman S., Cubas R., Comps-Agrar L., Fulzele A., Bennett E.J., Grogan J.L., Hui E., Chiang E.Y., Mellman I. Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8+ T cell responses. Immunity. 2022 Mar 8.

In the Spotlight...

Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial

Treatment-naive RCC patients (n= 29) were randomized 2:1 to receive combination nivolumab plus ipilimumab treatment with or without oral CBM588, a live bifidogenic bacterial product. At data cutoff, the median follow-up time was 12.2 months, 12 patients were still on treatment, and 24 patients were alive. Median PFS was significantly prolonged in the CBM588-treated group (12.7 months versus 2.5 months, hazard ratio 0.15, P<0.001). Disease control was reached in 15 (79%) CBM588-treated patients versus 4 (40%) in the other arm. Exploratory microbiome analyses indicated Bifidobacterium increase in treatment-responsive CMB588-treated patients.

Contributed by Margot O’Toole

A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies proline metabolism as a means to enhance CAR-T therapy

Ye et al. developed catalytically dead-guide RNA based genome-wide gain-of function (GOF) CRISPR activation screen, and identified PRODH2 as a lead target to boost primary CD8+ T cell effector functions. Genomic knock-in or lentiviral overexpression of PRODH2 in CAR T cells reprogrammed T cell metabolism, activated the proline metabolic pathway, enhanced mitochondria biogenesis and oxidative metabolism, increased T cell effector function, and enhanced CAR T cell therapy in vitro and in vivo. PRODH2 GOF CAR T cells showed a memory-like phenotype after long-term antigen stimulation.

Contributed by Shishir Pant

The interaction of CD4+ helper T cells with dendritic cells shapes the tumor microenvironment and immune checkpoint blockade response

Cohen, Giladi, and Barboy et al. used scRNAseq and sequencing of physically interacting cells (PIC-seq) to systematically characterize the cell landscape in the TME to determine if a functional interacting T–myeloid cell signature was present. CD4+PD-1+CXCL13+ T cells (T-helper tumor [Tht] cells), and not CD8+ T cells, preferentially formed interactions with LAMP+ mature regulatory DCs, and were clonally expanded in human NSCLC lesions, but not in normal tissue. Tht cells were tumor-specific (primed by tumor antigen-presenting DCs in tumor-draining LNs), conserved across several tumor types, and required for an antitumor response to anti-PD-1.

Contributed by Katherine Turner

A carcinoembryonic antigen-specific cell therapy selectively targets tumor cells with HLA loss of heterozygosity in vitro and in vivo

To overcome on-target, off-tumor toxicity caused by CAR T cell therapy targeting carcinoembryonic antigen (CEA) in colon cancer, Sandberg and Wang et al. developed a modular dual-signal system (Tmod) consisting of a third-generation CEA CAR construct combined with an A*02-specific scFv blocker module, providing an OFF-switch for HLA-A*02-expressing cells. A*02 expression on Tmod was reduced using shRNA to prevent cis inhibition. In vitro experiments showed that Tmod were tumor-specific, had low off-tumor activity, and could sequentially switch between ON and OFF states. The tumor-specific activity was confirmed in mouse xenograft models.

Contributed by Maartje Wouters

Tumor-specific T cells support chemokine-driven spatial organization of intratumoral immune microaggregates needed for long survival

Abdulrahman et al. demonstrated that patients with HPV16+ oropharyngeal squamous cell carcinoma (OPSCC) with a tumor-specific T cell response (IR+) showed enrichment of immune signaling pathways, increased immune cell infiltration, and longer OS compared to HPV16+IR and HPV patients. HPV16+IR+ tumors showed enrichment of CD103+CD8+ T cells, DCs, and CD14+ inflammatory myeloid cells. Spatial analysis revealed DC/T cell-enriched microaggregates, potentially organized by the differentially expressed genes LTB and CXCL12. HPV16+IR+ tumors showed preferential expansion of CD8+ T cell clusters expressing CCL4, associated with improved survival.

Contributed by Shishir Pant

Inhibition of renalase drives tumour rejection by promoting T cell activation

Guo et al. showed that T cells mediated rejection of non-immunogenic murine melanomas in mice treated with antibody to, or lacking expression of flavoprotein renalase (RNLS), known to protect cells from injury. scRNAseq showed tumors from these mice had higher PD-1, CD40, and exhaustion marker levels; greater densities of inflamed macrophages, neutrophils, DCs, NK cells, and T cells; and fewer Foxp3+CD4+ cells than controls. Anti-RNLS induced memory and was enhanced by anti-PD-1 or anti-CTLA-4 co-therapy. High RNLS expression in pre-treatment samples from patients treated with PD-1 inhibitors was associated with decreased survival.

Contributed by Paula Hochman

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.