Weekly Digests
‹ Back to June

Age is not just a number: age-related dysfunctional T cell subset restricts antitumor responses

June 5, 2024

Aging is known to induce immune dysfunction in cancer, but the mechanisms behind it remain poorly understood. In a recent Nature Immunology publication, Chen et al. assessed tumor-infiltrating T cells and the tumor microenvironment (TME) of young and aged mice to determine how age-related dysfunction relates to chronic stimulation-induced T cell exhaustion.

To evaluate the effects of aging on the antitumor immune response, OVA-expressing B16 melanoma (B16-OVA) tumors were implanted in C57BL/6J mice of various ages. In mice aged 68-70 weeks (aged mice) tumor growth was faster than in 10–12-week-old mice (young mice). Further, the aged mice had fewer tumor-infiltrating CD8+ T cells and OVA-specific CD8+ T cells.

scRNAseq of the tumor-infiltrating CD8+ T cells revealed several populations of Pdcd1- and Tox-expressing exhausted cells, progenitor-like exhausted Tcf7- and Slamf6-expressing cells (Tprog), and terminally exhausted cells (Tterm), characterized by Havcr2 and Entpd1 expression. In the aged TME, exhausted T cells were depleted, while less differentiated subsets were increased, including naive-like (Tnaiv), memory-like (Tmem-like) and transitory (Ttran) CD8+ T cells. Further, there was a cluster only expressed in the aged TME characterized as PD-1+Tox+IL-7R+CD8+ T cells, which did not express Tcf7, Slamf6, or Havcr2. The researchers named this subset tumor-infiltrating age-associated dysfunctional T cells (Ttad). In the aged TME, there was increased Ttad cell formation, including within the OVA-specific subset, and a decrease in Tterm cells. The aged tumoral CD8+ T cells produced less IFNγ, TNF, IL-2, and granzyme B, and had lower proliferative capacity.

To determine whether these age-related differences could be explained by defects in baseline T cells in healthy aged mice, the researchers transferred OT-1 cells from young mice (OT-1y>y) and OT-1 cells from aged mice (OT-1a>y) into young tumor-bearing mice, and profiled the tumor-infiltrating cells. The aged OT-1a>y had a higher ratio of Tterm to Tprog cells than the young OT-1y>y and the ratio of Tterm to Tprog cells skewed further over time during tumor progression. Before transfer, at baseline, many of the characteristics of the aged population were already present. Additionally, the aged OT-1 CD8+ T cells had reduced cytotoxicity in vitro, and OT-1a>y did not induce tumor control in young mice, in contrast to the young cells.

To assess how these defects are encoded in the aged T cells, chromatin accessibility changes were profiled between young and aged CD8+ T cells at baseline and after transfer in the TME. DNA damage/repair and replicative senescence signatures were differentially regulated in aged OT-1a>y Tterm cells, suggesting these cells might be more susceptible to genotoxic stress. Exposing the cells to UV irradiation confirmed that the aged cells upregulated the DNA damage markers γH2AX and p21 more rapidly and to higher levels. In vivo, the aged population had reduced proliferation in young mice, while young OT-1 that were transferred into aged mice showed no difference in proliferation to those transferred into young mice, even though the endogenous T cells in the TME had lower proliferation capacity.

The researchers did not detect the Ttad population in tumors of young mice transferred with aged OT-1. However, when young OT-1 were transferred into aged mice, there was Ttad formation and a loss of Tterm cells. Therefore, the formation of Ttad was not an intrinsic CD8+ T cell characteristic, but required signals from the aged TME. To further assess this, young OT-1 were transferred into young (OT-1y>y) and aged (OT-1y>a) mice with B16-OVA tumors. The transfer of the young OT-1 did not induce tumor control in the aged mice, and fewer transferred cells ended up in the tumor.

Chen et al. isolated intratumoral OVA-tetramer+CD8+ (Tet+CD8+) T cells and OT-1 populations from tumors in young and aged mice and performed coculture experiments. This showed that young and aged Tprog cells had low cytotoxicity, young Tterm had higher cytotoxicity than aged Tterm cells, and Ttad cells had almost no cytotoxic activity. When Tet+CD8+ cell populations were transferred into young mice, those receiving young Tprog cells had better tumor control, while there was no benefit from Ttad cells.

To determine whether Ttad cells could also be detected in human cancers, published pan-cancer scRNAseq datasets were assessed, and a similar cluster of PD-1+IL-7R+CD8+ T cells not expressing SLAMF6 or Tim3 was found to be enriched for the transcriptional signature of the mouse Ttad. This cluster was more abundantly present in older than in younger patients.

Profiling the immune populations in aged murine tumors, the researchers found that only CD8+ T cell, NK cell, and DC levels were lower than in young tumors. The aged TME had lower expression levels of antigen presentation and chemokine genes. Aged DCs downregulated antigen presentation molecules B2m and Tap1, and upregulated Apoe, which inhibits antigen presentation. There were fewer interactions between NK cells, cDC1, and CD8+ T cells in the aged TME. Aging tumors and tumor-draining lymph nodes (TDLNs) had fewer cDC1s, which expressed lower levels of CD40, CD86, MHC-I/II, and FLT3.

Finally, the effects of an OVA-mRNA neoantigen vaccine, optimized using lipid nanoparticles, were assessed in young and aged mice. While the vaccine induced tumor control and increased CD8+ T cell tumor and TDLN infiltration in young mice, there were no effects in aged mice. Since CD40 agonists can increase myeloid cells and their function in tumors, the researchers alternatively evaluated whether activating the cDC1 population in the aged TME could improve CD8+ T cell antitumor responses. Treatment with an agonist anti-CD40 antibody improved tumor control in both young and aged mice, and it increased cDC1s in the TDLN, and CD8+ T cells in the tumor and TDLN. Treatment induced upregulation of CD40, CD86, MHC-I/II, and FLT3 on aged cDC1s and decreased the Tprog, Tterm, and Ttad subsets, while increasing memory- and effector-like subsets.

These data suggest that aging-related unfavorable antitumor responses are linked to a newly-identified T cell subset, which appears due to reduced interactions with cDC1s in the TME. These findings have important implications. Therapies that enhance the interactions between cDC1s and CD8+ T cells may improve therapeutic efficacy in older patients.

Write-up by Maartje Wouters, image by Lauren Hitchings.

References:

Chen ACY, Jaiswal S, Martinez D, Yerinde C, Ji K, Miranda V, Fung ME, Weiss SA, Zschummel M, Taguchi K, Garris CS, Mempel TR, Hacohen N, Sen DR. The aged tumor microenvironment limits T cell control of cancer. Nat Immunol. 2024 Jun;25.

In the Spotlight...

Engineered CD47 protects T cells for enhanced antitumour immunity

CAR T cells in combination with anti-CD47 failed to promote tumor clearance due to systemic macrophage-mediated depletion of CAR T cells, which was so pronounced that anti-CD47 could function as a CAR T cell safety switch. A CD47 variant (47E) was designed to evade antibody blockade while maintaining function as a “don’t eat me” signal. 47E T cells resisted depletion and induced myeloid-activating/attracting genes in intratumoral T cells, which promoted phagocytic macrophage infiltration. 47E T cells + anti-CD47 promoted significant tumor control and survival across multiple solid and liquid models incorporating CAR or TCR T cells.

Contributed by Morgan Janes

Mapping variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens

Naturally occurring single-nucleotide variants (SNVs) in T cells can affect their functionality, to the point of clinical pathology, and so Walsh and Shah et al. hypothesized that precise base editing of essential T cell genes could uncover variants with superior T cell activity. High-throughput adenine base editor screens in human T cells generated a library of SNVs targeting relevant loci in 102 genes critical to T cell function. Several variants (particularly in PIK3CD) improved T cell proliferation, activation, and cytokine production. Editing these SNVs into TCR T cells or CD19 CAR T cells boosted antigen-specific cytokine production and tumor cell killing.

Contributed by Alex Najibi

Dose escalation study of a personalized peptide-based neoantigen vaccine (EVX-01) in patients with metastatic melanoma

Mørk et al. report the feasibility, safety, efficacy, and immunogenicity of the dose-escalation phase of a personalized neoantigen vaccine, EVX-01-CAF09b, in addition to anti-PD-1 therapy in patients with metastatic melanoma. The combination was safe, well tolerated, and capable of eliciting T cell responses, without vaccine-related serious adverse events. Vaccine neoantigen-specific CD4+ and CD8+ T cell responses were elicited at all dose levels, and objective tumor responses were observed in 66% of patients. The number of immunogenic peptides and PIONEER epitope quality scores were predictive of clinical responses and longer PFS.

Contributed by Shishir Pant

Preclinical efficacy and pharmacokinetics of an RNA-encoded T cell-engaging bispecific antibody targeting human claudin 6

Stadler et al. showed that BNT142, a lipid nanoparticle RNA formulation encoding a bispecific Ab that binds CD3 monovalently and (tumor-expressed) oncofetal antigen claudin 6 bivalently was translated, self-assembled, and secreted as active Ab RiboMab02.1 in cell cultures, mice, and cynomolgus monkeys. BNT142 mediated claudin 6+cell-dependent activation of T cell functions in vitro, and in mice, induced target cell-dependent T cell tumor infiltration, claudin 6+ cell depletion, and regression of claudin 6+ human tumor s.c. xenografts. BNT142 i.v. injection of mice and cynomolgus monkeys led to high, persistent RiboMab02.1 serum levels, and was well tolerated.

Contributed by Paula Hochman

Dual role of the peptide-loading complex as proofreader and limiter of MHC-I presentation

Brunnberga et al. used fluorescently engineered, enhanced-affinity, soluble TCRs and antibody–nanobody complexes to quantitatively detect and demonstrate the localization of pMHC-I complexes and different MHC-I allomorphs. Deletion of some components of the PLC editing modules (TSN, CRT, or ERp57) reduced the amount of MHC-I surface molecules, changed the MHC-I surface composition, and favored an increased presentation of suboptimally loaded peptides. Deficiencies in the editing module led to elevated presentation of abundant, high-affinity peptides, suggesting that the PLC restricts the presentation of highly expressed peptides.

Contributed by Shishir Pant

Improvement of tumor neoantigen detection by high field asymmetric waveform ion mobility mass spectrometry

To improve the sensitivity of mass spectrometry (MS)-based peptidomic profiling for the detection of neoantigens, Meng, Tekeuchi, and Ward et al. developed FAIMS – a gas-phase separation technique that can be implemented as an attachment to existing MS technology. In the T3 sarcoma model, FAIMs improved detection of the functionally validated major (mLama4 on MHC-I and mItgb1 on MHC-II) and minor (mAlg8 on MHC-I) neoantigens. FAIMs also effectively identified mLama4 from individual 100mg tumor samples, without the need for tumor cell expansion. Similar results were observed in 1956 and F244 tumors.

Contributed by Lauren Hitchings

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.