Weekly Digests
‹ Back to July

Rethinking the meaning of T cell exhaustion in non-small-cell lung cancer

July 25, 2018

Blockade of the PD-1/PD-L1 axis effectively reverses T cell exhaustion in some instances of cancer, but understanding what defines “exhaustion” and which cells have the potential for reinvigoration will be important in predicting responses, improving efficacy, and broadening the applications of PD-1 checkpoint blockade. To better understand T cell exhaustion in the context of cancer, Thommen et al. evaluated intratumoral CD8+ T cell populations from patients with non-small-cell lung cancer (NSCLC) and categorized tumor-infiltrating lymphocytes (TILs) as having high PD-1 expression (PD-1T), intermediate PD-1 expression resembling that of healthy donor PBMCs (PD-1N), or no PD-1 expression (PD-1-), and compared their transcriptional, metabolic, and functional signatures.

First working to define characteristics of the the PD-1T subset, Thommen et al. noted that PD-1T TILs showed upregulation of additional exhaustion markers, including Tim-3 and Lag-3. Ex vivo assessment of cytokine secretion following CD3/CD28 stimulation showed that PD-1T TILs failed to produce substantial amounts of classical effector cytokines including IL-2, TNFα, or IFNγ. The PD-1T subset was also characterized by high clonality, with the top 30 clones accounting for nearly 90% of the TCR repertoire, suggesting that this phenotype might be driven by TCR signaling. Further, these dominant clonotypes were not frequent among PD-1N or PD-1- cells. Interestingly, following in vitro expansion, PD-1T TILs reduced PD-1 levels, regained effector function, and were found to have an increased capacity for tumor recognition, with tumor reactivity largely restricted to the PD-1T subset.

Using RNA-seq, Thommen et al. uncovered that while PD-1N and PD-1- populations had nearly overlapping gene expression profiles, with only 8 genes differentially expressed, cells within the PD-1T subset had a vastly different transcriptional signature from PD-1N TILs, with 577 genes significantly up- or downregulated between them. Among the genes most upregulated in PD-1T TILs were those related to cell cycle regulation and proliferation (deviating from what has been observed in chronic infection models), and cell cycle analysis showed that PD-1T TILs were able to progress through the cell cycle. While PD-1T TILs were highly clonal and expressed high levels of inhibitory receptors and reduced levels of activating receptors, they showed little evidence of senescence.

Investigating whether high PD-1 expression by TILs could be linked to metabolic changes, the researchers found that cells in the PD-1T subset showed signs of increased glucose and lipid uptake and high lipid content, which could possibly be accounted for by observed expression of the scavenger receptor CD36. PD-1T TILs had, overall, a slightly larger mitochondrial mass, however, the mitochondrial membrane potential was found to be significantly reduced, indicating dysfunctionality. Morphological changes including fewer and shorter cristae were also observed.

When PD-1T TILs were expanded in vitro, derivative TILs continued to express high levels of Tim-3 and Lag-3, which could indicate an underlying, inherited state of dysfunction. Having observed that monocytes and tumor cells in NSCLC both express high levels of IL-10 and that the number of IL-10-producing cells correlated with the number of PD-1T TILs, the researchers tried exposing IL-2-expanded TILs to IL-10 and found that PD-1T-derived TILs rapidly re-expressed high levels of PD-1. Expansion in the presence IL-10 reduced IFNγ production most severely in PD-1T TILs. Together, these results indicate that the PD-1T subset might be inherently susceptible to certain modes of immunosuppression.

Early cytokine secretion analysis had shown that PD-1T TILs failed to produce high levels of classical effector cytokines, but interestingly, transcriptome analysis revealed that they did constitutively secrete high levels of the effector chemokine CXCL13, which controls recruitment and organization of B cells within lymphoid follicles. Through in vitro testing, the researchers found that CXCL13 secretion by PD-1T TILs was able to to attract CXCR5 receptor-expressing cells including B cells, CD4+ follicular helper T cells (Tfh), and a subset of exhausted CD8+ T cells, with the migration of CD4+ T cells being the strongest. In vivo, PD-1T TILs localized in intra- and peritumoral tertiary lymphoid structures alongside Tfh and B cell infiltrates, indicating that PD-1T may play an active role in recruitment of these immune cell types to the tumor.

Finally, Thommen et al. evaluated the presence of PD-1T TILs in pretreatment biopsies from twenty-one NSCLC patients who were being treated with PD-1 blockade therapy and found that the presence of PD-1T TILs strongly predicted response to therapy and correlated with durable responses and longer overall survival.

These studies clarify the qualities of “exhausted” CD8+ T cells in cancer and bring to light the strong correlation between intratumoral PD-1T TIL levels and clinical activity, including overall survival. The presence of PD-1T TILs may be important as a biomarker for efficacy, and understanding their qualities may serve as a tool to better understand how PD-1 expression and immune cell recruitment contribute to antitumor effects.

by Lauren Hitchings

References:

Thommen D.S., Koelzer V.H., Herzig P., Roller A., Trefny M., Dimeloe S., Kiialainen A., Hanhart J., Schill C., Hess C., Savic Prince S., Wiese M., Lardinois D., Ho P.C., Klein C., Karanikas V., Mertz K.D., Schumacher T.N., Zippelius A. A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med. 2018 Jun 11.

In the Spotlight...

IL-23 secreted by myeloid cells drives castration-resistant prostate cancer

Calcinotto et al. demonstrated that in human and mouse castration-resistant prostate cancers, polymorphonuclear myeloid-derived suppressor cells are enriched in the tumor microenvironment, where they confer resistance to androgen deprivation therapy (ADT) via secretion of IL-23. IL-23 regulated the pSTAT3-RORγ signaling axis to drive the transcription of the androgen receptor (AR) and its target genes, leading to survival of prostate cancer cells despite androgen deprivation. In mice, anti-IL-23 treatment reversed resistance to ADT and enhanced the efficacy of AR antagonist enzalutamide to decrease tumor volume.

A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments

Barry et al. show that in mice with melanoma, intratumoral NK cell expression (but not T cell expression) of Flt3l controls the levels of intratumoral CD103+BDCA-3+ cDC1s, a subset of DCs uniquely capable of intratumoral stimulation of cytotoxic T cells. NK cells appeared to control the frequency of cDC1s in the tumor via frequent and stable interactions between the two cell types. In patients with melanoma, NK cell levels correlated with both FLT3L expression and intratumoral cDC1 levels. Levels of NK cells and intratumoral cDC1s correlated with increased survival and predicted response to anti-PD-1 therapy.

Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment

Using single-cell RNA-seq, Azizi et al. computationally analyzed 45,000 immune cells from 8 breast cancer carcinomas, along with cells from matched normal breast tissue, blood, and lymph nodes. Despite similarities between tissue-resident and tumor-resident immune cells, there was increased heterogeneity in cell states and expansion of diverse immune populations within the tumor. A continuous spectrum of T cell activation states was observed, likely due to both TCR diversity and exposure to different microenvironments within the tumor. Interestingly, the data did not align with the canonical M1/M2 macrophage polarization model.

Ilixadencel - an Allogeneic Cell-Based Anticancer Immune Primer for Intratumoral Administration

Karlsson-Parra et al. reviewed preclinical and clinical testing of the cell-based off-the-shelf immune primer, Ilixadencel, a healthy donor-derived, ex vivo activated, intratumorally administered dendritic cell (DC) product that secretes proinflammatory cytokines and chemokines. Ilixadencel induces the recruitment and activation of endogenous immune cells, including NK cells, which promote the cross-presentation of tumor antigens by co-recruited dendritic cells. Results from early clinical trials have shown good safety profiles, evidence of tumor-specific immune responses, and antitumor activity.

Distinct human circulating NKp30+FcεRIγ+CD8+ T cell population exhibiting high natural killer-like antitumor potential

From the PBMCs of healthy volunteers, Correia et al. uncovered a unique subset of CD8+ T cells that express the activating natural killer receptor NKp30, display a diverse TCR repertoire, and exhibit a naive phenotype with few exhaustion markers. Treatment of NKp30-CD8+ T cells with IL-15 led to surface expression of functional NKp30, dependent on upregulation of the FcεRIγ adaptor, and reprogramming to an innate-like gene expression profile. IL-15 further differentiated this subset into effector memory NKp30hiCD8+ T cells with an NK-like phenotype; these cells efficiently killed tumor cells in vitro and controlled tumor growth in a xenograft mouse model of melanoma.

Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells associate with clinical outcomes in NHL

Rossi et al. utilized a T cell polyfunctionality strength index (PSI) to evaluate preinfusion CD19-targeted CAR T cells used to treat non-Hodgkin lymphoma. Although only 20% to 25% of cytokine-producing product T cells were polyfunctional, PSI correlated with objective response and grade 3+ cytokine release syndrome. The association was stronger when combined with markers of in vivo CAR T cell expansion or Day 0 IL-15 levels, particularly for CD4+ T cell subsets. Polyfunctional T cells expressing IL-17A were associated with grade 3+ neurotoxicity and antitumor efficacy.

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.