CD226 has role in tumor-specific CD8+ T cell differentiation
Since human TILs in NSCLC differentially express CD226 and CD28 in various CD8+ T cell clusters, combination treatment may be required to optimally activate the entire tumor-reactive TIL repertoire13. To evaluate the role of CD226 on tumor-specific CD8+ T cell subsets in the mouse tumor model, we segregated gp70+ CD8+ T cells based on CD226 expression. Anti-TIGIT alone or in combination with anti-PD-L1 increased the frequency of CD226+gp70+CD8+ T cells in both dLN and tumor, even with FTY720 treatment (Fig. 2a). Following combination blockade, CD226+gp70+CD8+ T cells were significantly more proliferative (Ki67+), but only in dLN (Fig. 2b, c, Extended Data Fig. 3a). CD226–gp70+CD8+ T cell proliferation was not affected by any treatment. Few CD226+gp70+CD8+ T cells in dLN were naïve as compared with the CD226– fraction (Fig. 2b, Extended Data Fig. 3b); combination treatment, but neither monotherapy, increased the frequency of CD226+gp70+CD8+ T cells with a Teff or Tem phenotype whereas no effects were observed in the CD226– population (Fig. 2b, Extended Data Fig. 3c).
To further elucidate the effects of checkpoint blockade on activation and differentiation, we measured various markers of T cell states. Slamf6 and TCF1 co-expression are considered markers of Tscm or Tpex cells7, 16. In dLN, the frequency of these cells in the CD226+ fraction was not affected by any treatment, but anti-TIGIT alone or in combination with anti-PD-L1 significantly reduced frequencies in the CD226– subset (Fig. 2b; Extended Data Fig. 3d, p = 0.0014). By contrast, in tumor, anti-TIGIT and combination treatment increased frequencies of Slamf6+TCF1+ cells in both CD226+ and CD226− subsets (Fig. 2c; Extended Data Fig. 3d, p = 0.0014).
As T cells differentiate from the Tscm or Tpex state, they express immune checkpoints such as Tim3. Combination treatment as well as anti-TIGIT alone increased the frequencies of both CD226+ and CD226– TCF1+Tim3+ gp70+CD8+ T cells in tumor whereas effects in the dLN were limited to the CD226+ subset; FTY720 largely abolished these effects (Fig. 2b,c; see Extended Data Fig. 3e for statistics). As T cells further differentiate, they lose expression of TCF1 although transcription of the Tcf7 gene appears to precede the loss of the TCF1 protein itself (compare to Fig. 3b). In the dLN, a significant increase in the frequency of TCF1– tumor specific CD8+ T cells is seen in the CD226+ fraction with anti-TIGIT or combination treatment; no effect was detected in CD226– cells (Fig. 2b; Extended Data Fig. 3f).
Tox is a key transcriptional regulator of exhaustion programming and differentiation towards terminal exhaustion4, 5. Treatment with either anti-TIGIT alone or anti-TIGIT plus anti-PD-L1 markedly decreased Tox expression in CD226+ but not CD226– gp70+CD8+ T cells in dLN, while decreased Tox expression was seen in both CD226+ and CD226– fractions in tumor; FTY720 appeared to diminish the combination effect on Tox expression in some cases (Fig. 2b,c; see Extended Data Fig. 3g for statistics).
Similar effects were seen in the EO771 model, with combination treatment increasing the frequency of CD8+ T cells in tumors, promoting CD226 expression on tumor CD8+ T cells, and increasing the TCF1+Tim3+ phenotype while reducing Tox+ frequencies (Extended Data Fig. 3h-m).
To assess the effector state of TILs responding to checkpoint blockade, we measured production of the proinflammatory effector cytokines IFN-g and TNF-a. Single-agent anti-TIGIT and combination treatment increased dual production of proinflammatory cytokines IFN-g and TNF-a in the CD226+ fraction of intratumoral CD8+ T cells relative to the CD226– fraction, with FTY720 eliminating this effect, suggesting that T cells derived from the periphery might possess superior effector function (Fig. 2d, e); assessment of cytokine production by tumor-specific TILs was not possible due to downregulation of TCR upon in vitro stimulation.
As anti-TIGIT plus anti-PD-L1 appeared to have more pronounced effects on CD8+ T cells expressing CD226, particularly in dLN, we concurrently treated mice receiving the combination with CD226-blocking mAb. As we could not segregate gp70-specific CD8+ T cells on the basis of CD226 expression in the presence of the blocking mAb, we examined total gp70+ cells and could not discern effects on Slamf6+TCF1+ cells (Fig. 2f, Extended Data Fig. 3n). However, anti-CD226 mAb impaired the ability of combination treatment to increase the frequency of TCF1+Tim3+ tumor-specific CD8+ T cells in dLN and tumor (Fig. 2f, Extended Data Fig. 3o). CD226 blockade also showed a trend towards reducing the ability of combination treatment to drive differentiation to a Teff/Tem phenotype (Fig. 2f, Extended Data Fig. 3p). Anti-CD226 mAb prevented the reduction in Tox-expressing cells in dLN and to a greater extent in tumor (Fig. 2f; Extended Data Fig. 3q, p = 0.025 and 0.009 respectively).
Taken together, addition of anti-TIGIT to PD-1/PD-L1 blockade initiated distinct differentiation pathways of Tscm or Tpex cells in dLN in a CD226-dependent fashion. These cells were further expanded in the tumor and were guided to develop into qualitatively better polyfunctional effectors. Similarly, upregulation of Tox characteristic of Tpex and Tex differentiation was prevented, again in a CD226-dependent manner.
TIGIT and PD-L1 co-blockade promotes and expands different CD8 + T cell states in dLN, blood, and tumor
We further examined how co-blockade affects the generation, phenotype, and trajectory of tumor-specific T cells using a multi-omics single-cell approach, performing single-cell RNA sequencing (scRNA-seq) and TCR sequencing (scTCR-seq) on T cells from tumor, dLN, and blood. These assays were supplemented by antibody-derived tag sequencing (ADT-seq) with tetramers against gp70 and cellular indexing of transcriptomes and epitopes (CITE-seq) using a panel of 18 cell surface proteins.
Gene expression profiles of a large dataset of 245,675 T cells yielded 24 distinct clusters (Extended Data Fig. 4a), with contributions across treatment groups (Extended Data Fig. 4b), but with some clusters appearing selectively localized to dLN, blood, or tumor (Extended Data Fig. 4c). Effector status, as indicated by granzyme B expression, was confined primarily to CD8+ T cells that showed clonal expansion and high ADT counts, a measure of the number of gp70 tetramers bound (Extended Data Fig. 4d–g). CITE-seq provided a complementary characterization of T cell differentiation, effector, and memory states based on surface marker expression (Extended Data Fig. 4h).
We obtained greater resolution of CD8+ T cell phenotypes by re-analyzing the T cells with high CD8a expression. These 155,496 CD8+ T cells comprise one of the largest datasets used for this type of analysis, enabling higher resolution clustering and unprecedented insight into the responses of CD8+ T cells to checkpoint inhibition. 20 distinct CD8+ clusters were identified (Fig. 3a,b; Extended Data Fig. 5; see Supplementary Table 1 for genes defining each cluster), with contributions consistent across individual mice (Extended Data Fig. 6a). As before, clusters belonged to specific tissues, and had contributions across experimental groups (Extended Data Fig. 6b). Clonal expansion and ADT counts were differentially distributed amongst clusters, with increases seen in non-Ccr7 clusters (Extended Data Fig. 6c).
The clusters exhibited various phenotypes (Fig. 3a):
(a) four Ccr7 clusters ("Ccr7.1-4") characterized by Ccr7, a marker expressed by naïve, Tscm and central memory (Tcm) cells but low in cytotoxic CD8+ Teff and Tem cells28, as well as genes associated with Tscm cells such as Sell, Lef1, and Tcf718, and also high expression of ribosomal proteins;
(b) a distinct cluster ("Early") characterized by expression of Cd69 and other markers of early T cell activation;
(c) a distinct "Slamf6" cluster marked by high Slamf6 and Tcf7 expression representative of a Tscm population;
(d) three Ifit clusters ("Ifit.1-3") with hallmarks of interferon response genes indicating activated T cells;
(e) two Ccl5 clusters ("Ccl5.1-2") marked by this chemokine that can exert chemotactic effects on T cells and is associated with CD8+ T cell infiltration into tumors29;
(f) two Cytotox clusters ("Cytotox.1-2") exhibiting hallmarks of cytotoxic gene expression as well as genes associated with exhaustion such as Tox and checkpoint inhibitory checkpoint receptors;
(g) three Cyt/Mit clusters ("Cyt/Mit.1-3") that represent proliferating cytotoxic cells as they express genes associated with cytotoxicity and mitosis;
(h) two Mitotic clusters ("Mitotic.1-2") expressing genes associated with mitosis but not genes associated with effector function; and
(i) two clusters representing dying cells ("Dying.1-2").
The Ccl5 clusters shared expression of a number of genes associated with the Cytotox or Cyt/Mit clusters, but did not have properties of exhaustion. CITE-seq analysis using various surface-expressed proteins corroborated this categorization by gene expression (Extended Data Fig. 6d–f). Both scRNAseq and CITE-seq analysis showed that CD226 expression was most characteristic of Ccl5.1 T cells. CD28 showed some overlapping expression with CD226 but also marked a few distinct clusters consistent with our previous findings for human NSCLC TILs13 (Extended Data Fig. 6d–f). The Ccl5.1 cluster is of particular interest in that it was the only major non-naïve cell state found in the blood.
Comparison of our clusters with reference gene signatures from published datasets23, 30, 31, 32 showed general concordance albeit with more granularity due to the larger sample set used here (Fig. 3c). Of particular relevance, our Ccl5, Ifit.3, and Cytotox clusters shared strong similarities with the “better effectors” described by others in response to a combination of anti-PD-1 therapy with IL-2 agonists31. However, our Ccl5.1 cluster also corresponded with the "Stem-like cluster" in that study and with the "Transitory Tex cluster" by Huang and colleagues23.
Our multi-omics dataset allowed us to convert spliced and unspliced mRNA counts to estimate RNA velocity measurements and infer differentiation trajectories. Although the directionality of cell traffic often cannot be assigned confidently from velocity-based trajectories33, visualization results from Li and colleagues using photoactivation have established the in vivo migration of T cells into and out of tumors34. By assigning our clusters to the Li et al. gene expression signatures (Extended Data Fig. 7a), we can ascertain directionality in our analysis. Using control-treated tumor-bearing mice as a reference, RNA velocity patterns differed in dLN and tumor (Fig. 3d). In dLN, a major trajectory originated from Early and Ccr7 clusters and yielded Slamf6 cells, which then differentiated into Ifit or Ccl5 cells. In tumors, differentiation progressed from Ccl5 cells through Cytotox cells to Cyt/Mit cells. From there, a second differentiation pathway generated Mitotic cells. RNA velocity patterns were similar across treatment groups, indicating that differentiation pathways were not fundamentally affected by the various treatments (Extended Data Fig. 7b,c).
Combination treatment expanded tumor-specific CD8 + T cells marked by Ccl5 that transit from dLN to tumor via blood
We then applied our scTCR-seq data to segregate T cells by the expansion of their parent clone, revealing striking differences across treatment groups, especially when using ADT-seq counts to distinguish gp70+ from gp70− cells (Extended Data Fig. 6c). As shown in Fig. 4, cells in dLN were predominantly singletons (having only one cell expressing a given TCR clonotype) across each cluster, but showed evidence of clonal expansion in the Slamf6 and Ccl5.1 clusters following combination treatment. In contrast, cells in tumor were almost exclusively expanded clones. Although clones were specific to individual mice, these results were not attributable to any single mouse (Extended Data Fig. 8).
At day 7, gp70+ CD8+ T cells were detected in the blood of mice treated with anti-TIGIT or combination treatment and were comprised of Ccl5.1 cells (Fig. 4, bars facing right). The absolute cell numbers were low, likely reflecting the transient residence of mobilized T cells in the blood. Their appearance was blocked by FTY720 treatment, indicating that expanded Ccl5.1 cells likely originated in dLN. This inference was supported by the accumulation of clonally expanded gp70+ Ccl5.1 cells in dLN.
Some gp70− CD8+ T cells were found in the Ccl5.1 cluster, but they were mostly in the immature Ccr7 clusters (Fig. 4, bars facing left). Since these cells were apparent at day 0 and in all treatment groups, they were not elicited by combination TIGIT/PD-L1 blockade. The gp70− cells in the Ccl5.1 cluster, however, were significantly enhanced by the combination, and could include both bystanders and T cell clonotypes that were specific to tumor antigens other than gp70.
Tumors, unlike the dLN or blood, contained relatively large numbers of both clonally expanded gp70− and gp70+ TILs in all treatment groups. However, in mice treated with both anti-PD-L1 and anti-TIGIT, this increase was most pronounced for gp70+ T cells, which were found in the Ifit, Ccl5.2, Cytotox, and Cyt/Mit clusters (Fig. 4). The increase in gp70+ clones in the Ccl5.2 cluster was both most pronounced and selectively decreased by FTY720 treatment, strongly suggesting that these cells derived from the blood-borne Ccl5.1 population. Interestingly, in FTY720 treated mice, gp70+ clones expanded in the other clusters, indicating that these may pre-exist in tumor and expand and differentiate intratumorally in response to combined PD-L1/TIGIT blockade.
Thus, in response to combination treatment, tumor antigen-specific (and possibly also non-specific) clonotypes expand in the dLN, exit as Ccl5.1 cells into the blood, and continued to expand after arrival in the tumor.
Co-blockade of PD-L1 and TIGIT focuses the TCR clonal diversity of tumor antigen-specific CD8 + T cells
We next compared the degree of clonal expansion in dLN, blood, and tumor at day 7 post-treatment, characterizing each clone by its majority cluster at each site (Fig. 5a, Extended Data Fig. 9a). Inhibiting both PD-L1 and TIGIT elicited strikingly coordinated clonal dynamics. Although only a few clones exhibited large expansions, they did so in each of the three tissue compartments (Fig. 5a). In dLN, expansion occured mostly in Ifit.3, Ccl5.1 and Cytotox.1 cells, while in the tumor Ccl5.2, Cytotox.1 or Cytotox.2 cells were preferentially expanded. Combination treatment also resulted in expansion in the blood (illustrated by the diameter of the circles shown in each plot, Fig. 5a; Extended Fig. 9a). Here, the expanded clonotypes were contained almost exclusively in the Ccl5.1 population (Fig. 4; Extended Data Fig. 9b,c), and these were shared with the corresponding clusters in dLN or the tumor (illustrated by the color of the circles in each plot, Fig. 5a; Extended Fig. 9a). Expansion due to single agent treatment occurred (to a greater extent following anti-TIGIT alone) but expansion was mostly limited to dLN or tumor.
In the presence of FTY720, many clones exhibited dual expansion in dLN and tumor with relatively limited expansion in blood, suggesting that these dual-expanded clones arose independently in dLN and tumor.
The most highly expanded clones following combination treatment were gp70+, indicated by a high ADT count (blue/purple circles, Fig. 5b); little or no expansion occurred after anti-TIGIT or anti-PD-L1 alone. Most of the dual-expanded clones in the single-agent treatment groups had low or undetectable gp70 ADT counts, suggesting that they were either “bystander” non-tumor reactive T clones35, 36 or specific for other tumor-associated antigens. In the presence of FTY720, high gp70 ADT counts were also detectable in dual-expanded clones, as expected if these cells represented pre-existing clones already present in dLN and tumor prior to treatment.
Since the scatterplots (Fig. 5a,b) depict only the primary cluster type for each clone, we evaluated the composition of the 30 most expanded clones for each treatment group in tumor, and matched them to dLN and blood to study the distribution of individual clones across T cell clusters (Fig. 5c). The largest clones in tumor had measurable counterparts in dLN but only following combination treatment. In dLN, these clones consisted predominantly of the Ccl5.1, Cytotox.1 and Cytotox.2 populations. The same expanded TCR clones were also found in the blood, again contained almost exclusively in the Ccl5.1 population. FTY720 treatment prevented the appearance of this population.
The picture was quite different following single-agent treatments. CD8+ T cells in the tumor following anti-PD-L1 had largely the same composition as the control group, comprised primarily of Cytotox.2 and Cyt/Mit clusters. Expansion of the Cytotox.2 cluster was more pronounced than with other treatments, suggesting that anti-PD-L1 drives T cell differentiation towards this specific state in tumor. Anti-TIGIT, in contrast, promoted a shift in the tumor towards the Ccl5.2 cluster. With single-agent treatment, none of the largest clones in tumor had appreciable counterparts in dLN or blood.
When we examined clonal expansion separately in each tissue compartment, each treatment had distinct effects on T cell differentiation (Extended Data Fig. 9b, c). In dLN, anti-TIGIT and combination treatment, but not anti-PD-L1 alone, caused expansion of Ccl5.1 T cells and, to a lesser extent, Mitotic clusters. FTY720 treatment shifted the intralymphatic composition to almost exclusively Ccl5.1, suggesting that these cells accumulated in dLN since their egress into blood was inhibited. Combination treatment, with or without FTY720, resulted in reduced proportions of the Slamf6 cluster in dLNs, especially in the most expanded clones, reflecting the possibility that the Slamf6 (putative Tscm) cluster is the source from which Ccl5.1 T cells are mobilized.
Anti-PD-L1 and anti-TIGIT differentially reshape differentiation and trajectories of CD8 + T cells in dLN and tumor
We next probed the lineage relationships across CD8+ T cell clusters following various treatments. Although we previously evaluated cellular trajectories using RNA velocity (Fig. 3d), it is apparent that individual clones exhibit complex expansion behaviors. scTCR-seq unambiguously identifies lineages of T cells, which provides a complementary approach to infer kinetics and differentiation based on the co-occurrence of phenotypes in individual clonotypes within and across tissue compartments.
We analyzed co-occurrences of cell phenotypes by tabulating numbers of intraclonal pairs over all clonotypes, plotting only pairs between different clusters (Fig. 6a–c). As with RNA velocity, we could use signatures derived from empirical observations34 to interpret such co-occurrences as directional steps in differentiation.
In dLN (Fig. 6a), control mice exhibited a predominant differentiation of Slamf6 to the Cytotox.1 phenotype, with little connection to other populations as illustrated by the absence of additional intercluster links. With single-agent treatment, increased differentiation from Slamf6 to the Ccl5.1 phenotype was observed, but with anti-TIGIT further increased differentiation of Ccl5.1 cells into Cytotox.1 and Mitotic.1 cells. Combination treatment produced an even more complex pattern of differentiation, with Ccl5.1 cells also differentiating to Ifit.3 cells, and those co-occurrences being shared across cytotoxic and mitotic clusters. FTY720 treatment resulted in most Slamf6 cells differentiating to Ccl5.1, but then a sharp reduction in Ccl5.1 cells differentiating to other clusters, as indicated by the absence of intercluster links. Intraclonal pairs in dLN were comprised of primarily gp70+ specificities across treatment groups, and some gp70– with control or single-agent treatment. Thus, although Slamf6 (Tscm) cells differentiated to cell states other than Ccl5.1 in dLN, only the Ccl5.1 population entered the blood, seeding tumors with new CD8+ T cells.
Co-occurrence profiles were different in tumor compared to dLN (Fig. 6b). Intraclonal pairs in control tumors showed an origin from the Cytotox.2 phenotype to the Cyt/Mit.1 and Cyt/Mit.2 phenotypes. Anti-PD-L1 had a similar pattern, but with additional co-occurrence of Cytotox.2 with the Ifit.3 and Cytotox.1 clusters. In sharp contrast, anti-TIGIT exhibited an expansion of clones with Ccl5.2 cells that differentiated to Cyt/Mit.2, Cyt/Mit.1, and Cytotox.2 cells; these clones were largely gp70− (blue lines), consistent with the largest clonotypes in that group being gp70− (Fig. 5c). Combination treatment resembled anti-TIGIT monotherapy in terms of Ccl5.2 expansion, but those Ccl5.2 cells differentiated primarily to Cytotox.1 cells. FTY720 treatment produced a complex pattern of co-occurrences among Ccl5.2, Cytotox.1, Cytotox.2, Cyt/Mit.1, and Cyt/Mit.2 clusters, revealing the extent of differentiation within tumor. In contrast with anti-TIGIT treatment, the vast majority of intraclonal pairs in tumor with combination treatment were gp70+.
We then tabulated intraclonal pairs from across tissues to determine migration relationships, plotting only co-occurrences between different tissues, but otherwise showing co-occurrences between both same and different clusters (Fig. 6c). In contrast to single-agent therapy, the anti-PD-L1/TIGIT combination facilitated migration of Ccl5.1 cells from dLN to Ccl5.2 cells in tumor, presumably through blood Ccl5.1 cells, but with co-occurrences from dLN to blood less apparent because of its relatively low degree of clonal expansion in both compartments (Fig. 4). Co-occurrences were also seen from Ccl5.1 cells in dLN to Cytotox.1 and Cytotox.2 clusters in tumor, but these are presumably attributable to intratumor differentiation (Fig. 6b). The co-occurrences between Ccl5.1 in dLN and Ccl5.2 in tumor were also observed in the presence of FTY720, with an absence of blood involvement, indicating that combination treatment may act on preexisting TILs in tumor that had progenitors remaining in the dLN.
To visualize these differentiation and migration patterns in the context of gene expression, we projected the co-occurrence data onto our previously computed UMAPs. From these plots (Fig. 6d,e), it is apparent that Slamf6 cells (putative Tscm) in dLN serve as progenitors for Cytotox.1 cells in control and anti-PD-L1 treated mice and for Ccl5.1 cells in other treated mice. These Ccl5.1 cells then migrate into blood, with more frequent migration occurring with anti-TIGIT and combination-treated mice in gp70+ clones (Fig. 6d) than gp70− clones (Fig. 6e). In these groups, and especially with combination treatment, the migration links revealed a convergence of multiple clusters from dLN onto Ccl5.1 cells in blood, and then a divergence from these cells into multiple clusters in tumor. With anti-TIGIT, and to a greater extent with combination therapy, gp70+ Ccl5.1 cells in blood then migrated into tumor where they appeared to give rise to the Ccl5.2 phenotype. Ccl5.2 cells differentiated into Cytotox.2 cells, which then differentiated into other cytotoxic and mitotic (precursor exhausted) phenotypes. Differentiation from gp70+ Cytotox.2 cells to other phenotypes was greater for anti-PD-L1 and FTY720 treatment, compared with anti-TIGIT and combination treatment. These results suggest that anti-TIGIT and especially combination treatment promote an immune response characterized by an influx of tumor-specific Ccl5.1 T cells, whereas anti-PD-L1 and FTY720 treatment exhibit primarily the differentiation of Cytotox.2 T cells already existing in the tumor.
Gene signatures derived from reference mouse CD8 + T cell clusters show association with response to tiragolumab plus atezolizumab in cancer patients
To explore whether these observations inform the clinical setting, we analyzed scRNA-seq data of peripheral blood T cells from a phase 1b study of NSCLC patients treated with the combination of tiragolumab plus atezolizumab (T + A) (GO30103)37. We mapped human CD8+ T cells onto the nearest mouse reference CD8+ T cell cluster (Extended Data Fig. 10a,b). Patients with a clinical response, evaluated as either complete response (CR) or partial response (PR), compared with non-responders (stable disease, SD, or progressive disease, PD), had an increased frequency of CD8+ T cells mapping to the Ccl5.1 and Ccl5.2 clusters and a decreased frequency mapping to Ccr7.3 and Ccr7.4 clusters (Extended Data Fig. 10c). This finding is consistent with Ccl5 clusters in our mouse models predominating with effective treatment.
To address whether gene signatures derived from the mouse CD8+ T cell clusters associated with improved overall survival (OS), we analyzed bulk RNA-seq data from baseline tumor samples from patients in CITYSCAPE10. The top 20 differentially expressed signature genes for each mouse CD8+ T cell cluster were used to derive orthologous human gene signature “scores” in CITYSCAPE samples (Supplementary Table 2) which compared patients treated with T + A or placebo plus atezolizumab (P + A). Ccr7.3, Slamf6, Ifit.1, Ifit.2, Ifit.3, Ccl5.2 and Cytotox.2 gene signature scores were significantly higher in CR and PR responders as compared with SD and PD non-responders (Fig. 7a). While all CD8+ T cell cluster signatures trended with favorable OS hazard ratio (HR) in patients treated with T + A compared to P + A (Extended Data Fig. 10d), high expression of Ccr7.3, Slamf6 and Ccl5.1 gene scores associated with significantly improved HR for OS (HR = 0.44 (95% CI: 0.22–0.91; p = 0.028), 0.46 (95% CI: 0.22–0.95; p = 0.036), and 0.45 (95% CI: 0.22–0.90; p = 0.025), respectively), as did low expression of Cytotox.1 and Cyt/Mit.2 (OS HR = 0.46 (95% CI: 0.23–0.90; p = 0.023) and HR = 0.48 (95% CI: 0.23–0.98; p = 0.045), respectively). Dichotomization of patients on the basis of high or low cluster gene signature score and by treatment showed that high expression of the Ccl5.2 gene signature trended with increased OS with T + A but not P + A (Extended Data Fig. 10e).
Gene signatures predominantly associated with response to T + A were characterized by high expression of chemokines or chemokine receptors. We focused on CXCR3, CXCR6, and CCL5, genes that were among the most highly expressed in each of the clusters (Supplementary Table 1). High expression of each of these individual genes was associated with response in patients treated with T + A (Fig. 7b), and high expression of CCL5 or CXCR3 was individually associated with favorable OS HR in T + A compared to P + A, outperforming CD8A (OS HR = 0.32 (95% CI: 0.14–0.73; p = 0.006), 0.41 (95% CI: 0.18–0.94; p = 0.035) and 0.43 (95% CI: 0.20–0.91; p = 0.027), respectively) (Fig. 7c). CXCR3, CXCR6, and CCL5 were associated with improved OS for T + A, again outperforming CD8A (Fig. 7d).
We then generated a composite gene signature score comprised of the average expression of CCL5, CXCR3, and CXCR6. This gene signature score was significantly higher (p = 0.012) in responder CITYSCAPE patients as compared with non-responders, (Fig. 7e). A high gene signature score was associated with favorable OS HR in patients treated with T + A (HR = 0.43, p = 0.035) compared with P + A, while a low signature score did not associate significantly with OS benefit (HR = 0.70, p = 0.277) (Fig. 7f). Segregation of patients on the basis of high or low gene signature scores showed that those treated with T + A who had high gene score expression had improved OS compared to patients with a low gene signature (Fig. 7g). The composite gene signature score was also associated with improved progression-free survival (PFS) and OS in the phase 3 OAK study (NCR02008227) of atezolizumab monotherapy in patients with locally advanced or metastatic, previously treated NSCLC38 (Extended Data Fig. 10f).
Thus, our analysis of patients treated with T + A largely recapitulates the findings of anti-TIGIT plus anti-PD-L1 in our mouse tumor studies, providing translational evidence that the events observed in dLN of tumor-bearing mice may also be detected in human blood and tumors. Furthermore, our study suggests that CD8+ T cell quality, as represented by cells newly arrived from dLN, rather than the mere presence of CD8+ T cells in the tumors supplied by the periphery at steady state 39, may be more strongly predictive of response and clinical benefit.