Weekly Digests
‹ Back to September

Age is more than just a number in cancer immunotherapy

September 11, 2024

Age-related changes to the immune system contribute to increased cancer progression and resistance to immunotherapy in older patients. Investigating this, Georgiev and Han et al. recently found that tumors grew more rapidly in aged mice due to impaired cytotoxic CD8+ T cell activation and effector differentiation within tumors, and a loss of tumor antigen-specific CD8+ T cells. These findings, along with further investigation into how to overcome these age-related deficiencies, were recently published in Cancer Immunology Research.

To study age-related changes to antitumor immunity, Georgiev and Han et al. evaluated MC38, EO771, and B16 tumors in young adult (2-4 months) and aged (20-24 months) mice. MC38 and EO771 tumors, both of which are immunogenic, grew more rapidly in aged mice, while B16 tumors, which are less immunogenic, grew similarly in young and aged mice. Expression of OVA in MC38, EO771, and B16 tumors enhanced the magnitude of the difference in tumor growth between aged vs. young mice across all models, including in the B16 model, where OVA expression induced a distinction. Depletion of CD8+ T cells accelerated tumor growth in young, but not aged mice, suggesting existing defects in antitumor CD8+ T cells in the aged mouse models.

Investigating the T cell compartment in young versus aged mice bearing OVA-expressing tumors, the researchers found that in aged mice, CD3+ T cells and CD8+ T cells were reduced in tdLNs, but not tumors. Within tumors, a smaller proportion of intratumoral CD8+ T cells in aged mice expressed both CD44 (antigen experience) and PD-1 (activation/exhaustion), and within CD44+PD-1+CD8+ T cells, fewer cells expressed markers associated with exhaustion. TILs in aged mice also expressed reduced markers of cytotoxic effector differentiation (T-bet), cytotoxicity (granzyme B), proliferation (MKi67), and effector memory, and increased markers of progenitor exhaustion (Tim3-TCF+) and central memory, consistent with impaired effector functions and differentiation. This defect was likely not related to impaired priming by APCs, as aged mice actually showed evidence of increased APCs with enhanced antigen presentation and costimulatory functions in tdLNs, though DCs expressed higher levels of PD-L1 and Tregs were increased in these tissues.

Next, the researchers performed scRNAseq and clustering analysis on tumor-infiltrating CD8+ T cells from MC38 tumors. While CD8+ T cells from aged mice fell more into progenitor exhausted and naive clusters, those from young mice fell more into transitory exhausted, progenitor exhausted/IFN sensing, and terminally exhausted clusters. Comparing quiescent and stimulated cells, the researchers noted more differentially expressed genes by age in quiescent cells. Further, trajectory analysis showed that while CD8+ T cells followed a similar differentiation trajectories in young and aged mice, those from young mice were distributed across the range of the trajectory, while those from aged mice were crowded towards the naive end of the pathway, suggesting fewer cells in activated states.

Investigating whether reduced activation was related to reduced antigen-specific CD8+ T cells in aged mice, the researchers found that while SIINFEKL-reactive CD8+ T cells could be readily detected in OVA-expressing tumors on day 12 in young mice, they were essentially absent from those in aged mice at the same time point. In tdLNs, SIINFEKL-reactive CD8+ T cells were detectable in aged mice, but were decreased compared to in young mice. Looking at CD8+ T cells specific for endogenous antigens in B16-OVA (MuLV p15E, gp100, and TRP-2), a similar decline was observed with age.

After determining that these effects could not be attributed to differences in T cell survival or TCRβ expression levels, Georgiev and Han et al. evaluated changes in TCR diversity over time by measuring TCR Vβ chain usage. In young mice, the pattern of Vβ expression was relatively consistent between animals in tissues, but varied between animals in tumors. In aged mice, the central memory pool became dominated by single Vβ chains in spleens and lymph nodes, and variation in naive cells increased between animals in spleens, lymph nodes, and tumors, suggesting reduced TCR diversity in aged mice.

Looking at how the frequencies of CD8+ T cells recognizing SIINFEKL versus TRP-2 varied over time in B16-OVA-bearing mice, the team found that the proportions and absolute numbers of intratumoral antigen-specific CD8+ T cells progressively reduced in tumors with age. This occurred earlier and was more pronounced for SIINFEKL-reactive CD8+ T cells than for TRP-2-reactive T cells. Interestingly, while granzyme B expression and proportions of TIM3+PD-1+ cells remained consistent in SIINFEKL-reactive CD8+ T cells, those features declined with age in TRP-2–reactive CD8+ T cells, suggesting that T cells with different antigen specificities may show distinct kinetics in their age-related declines.

Exploring opportunities to overcome age-related CD8+ T cell deficiencies, the researchers transferred OT-I CD8+ T cells from young mice into young or aged mice prior to B16-OVA tumor implantation. In young mice, this transfer slowed tumor growth, resulting in 25% long-term survival. In aged mice, the treatment initially slowed tumor growth more dramatically than in young mice, but ultimately all tumors escaped immune control, resulting in only a slight extension of survival. OT-I cells were reduced in the tumors of aged mice compared to young mice, but were increased in tdLNs, where a higher proportion of cells expressed MKi67, PD-1, and T-bet.

Next, the researchers evaluated the addition of anti-PD-1 therapy, which on its own, was more effective in young mice than in aged mice, which were unresponsive to treatment. The combination of OT-I and anti-PD-1 therapies was still more potent in young mice than in aged, though it did show some improved antitumor efficacy over OT-I cells alone in aged mice, leading to significantly longer survival and some long-term survivors.

Finally, testing whether age-related changes in CD8+ T cells impact antitumor immunity and responses to immunotherapy in patients, the researchers analyzed data from ~10,000 patients across multiple cancers from TCGA and categorized the patients by a CD8+ T cell infiltration score and age within each cancer type subset. This identified a pattern in which higher diagnosis age was associated with increased lymphocytes, consistent CD8+ T cells, but reduced CD8+ effector T cells. Among younger patients, the progression-free interval (PFI) was found to be significantly longer in individuals with a high CD8+ T cell score, while in older patients, PFIs were not stratified by T cell scores.

Together these results suggest that age-related changes in the immune system, including reduced TCR diversity, loss of tumor antigen-specific T cells, and defects in effector function and differentiation, likely contributing to increased cancer progression and resistance to immunotherapy in older patients. These age-related defects could be overcome with a combination of immunotherapies in mice. These results also support the use of aged mice for modeling cancer research in older patients, who make up the largest portions of most treatment populations.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, co-lead author Alison E. Ringel answered our questions.

Co-lead author Alison E. Ringel

What was the most surprising finding of this study for you?
We were surprised to uncover the extent to which antigen-specific killer T cells, which are important for recognizing new threats like tumors, decline with advanced age! This is a mechanism that may contribute to the rise in cancer incidence in later stages of life.

What is the outlook?
On one hand, our work suggests that aging may shift the features of tumors that are recognized by T cells in older individuals. Going forward, we need to understand more about how this affects tumor control by the immune system and if this influences how patients respond to immunotherapy. From a research-focused standpoint, we need to make sure that we accurately account for this decline in tumor-specific T cells when using mice as a preclinical model for human aging. This is important as more and more research focuses on the intersection of aging and antitumor immunity.

What was the coolest thing you’ve learned (about) recently outside of work?
My family recently traveled to Colorado for vacation. We visited an old gold mine with my two kids, where we learned that there’s still a lot of gold left in the mountains in Colorado, but it costs as much money to get it out as it’s worth!

References:

Georgiev P, Han S, Huang AY, Nguyen TH, Drijvers JM, Creasey H, Pereira JA, Yao CH, Park JS, Conway TS, Fung ME, Liang D, Peluso M, Joshi S, Rowe JH, Miller BC, Freeman GJ, Sharpe AH, Haigis MC, Ringel AE. Age-associated contraction of tumor-specific T cells impairs antitumor immunity. Cancer Immunol Res. 2024 Aug 24. 

In the Spotlight...

CMG901, a Claudin18.2-specific antibody-drug conjugate, for the treatment of solid tumors

Xu, Liu, and Wang et al. summarized preclinical data supporting the ongoing phase 3 clinical investigation of CMG901 in patients with advanced gastric/gastroesophageal junction tumors. CMG901, a Claudin 18.2-targeted Ab (CM311) conjugated to the microtubule-targeting agent MMAE, was shown to specifically bind to and be internalized by Claudin 18.2+ tumor cells. Mechanisms of killing included direct cytotoxicity, ADCC, and CDC, as well as bystander killing of Claudin 18.2-negative tumor cells. In patient-derived xenograft (PDX) models, CMG901 had significant antitumor efficacy, while toxicity studies in primates and rats revealed reversible hematological changes.

Contributed by Katherine Turner

NaCl enhances CD8+ T cell effector functions in cancer immunotherapy

Scirgolea and Sottile et al. showed that CD8+ T cells use NaCl to promote potent T cell effector functions. NaCl supplementation induced effector differentiation, IFNγ production, and cytotoxicity while maintaining stem-like plasticity. NaCl-mediated reprogramming depended on increased glutamine consumption from the microenvironment, leading to transcriptional, metabolic, and epigenetic remodeling, recapitulating PD-1 blockade-mediated T cell reinvigoration. A short pulse with excess NaCl in vitro reprogramed MART-1-specific CD8+ T cell function to exert more potent effector functions and tumor control in NSG mice.

Contributed by Shishir Pant

Sodium chloride in the tumor microenvironment enhances T cell metabolic fitness and cytotoxicity

Soll, Chu, and Sun et al. demonstrated increased intratumoral Na+ ion concentrations and enrichment of an NaCl gene signature in solid tumors from patients with breast cancer. High NaCl concentration augmented nutrient uptake and metabolic reprogramming to potentiate activation and cytotoxic effector functions of human CD8+ T cells. Mechanistically, NaCl upregulated Na+/K+-ATPase activity, membrane hyperpolarization, calcium influx, and downstream TCR signaling, leading to enhanced cytotoxicity. Stimulation of CAR T cells or TCR-engineered T cells under high NaCl conditions increased tumor cell killing in vitro and in vivo in the PancOVA model.

Contributed by Shishir Pant

Activation and antitumor immunity of CD8+ T cells are supported by the glucose transporter GLUT10 and disrupted by lactic acid

Liu and Wang et al. found the glucose transporter GLUT10 to be rapidly upregulated and trafficked to the cell surface during CD8+ T cell activation, and increased GLUT10 abundance on tumor-infiltrating CD8+ effector T cells. GLUT10 deletion in T cells inhibited the activation and proliferation, and reduced the antitumor efficacy of CD8+ T cells in vivo. Tumor cell-derived lactic acid bound directly to GLUT10, interfering with CD8+ T cell glucose uptake and effector functions. PG10.3, a peptide derived from GLUT10, overcame lactate inhibition. PG10.3 combined with a GLUT1 inhibitor or anti-PD-1 further enhanced antitumor efficacy in patient-derived tumor xenograft models.

Contributed by Ute Burkhardt

Human/mouse CD137 agonist, JNU-0921, effectively shrinks tumors through enhancing the cytotoxicity of CD8+ T cells in cis and in trans

Liu, Chen, Li, and Yang et al. developed and tested JNU-0921 – a small-molecule CD137 agonist with cross-species reactivity that directly bound to the extracellular domain of both human and mouse CD137, inducing CD137 oligomerization, leading to the recruitment of TRAF2 and TRAF3 and the activation NF-κB signaling. In cytotoxic CD8+ T cells, JNU-0921 enhanced effector and memory functions and reduced exhaustion, while in conventional CD4+ T cells, it supported Th1 polarization and enhanced helper functions. In Tregs, it attenuated inhibitory functions. In vivo, JNU-0921 induced tumor regression.

Contributed by Lauren Hitchings

TNFR1 signaling promotes pancreatic tumor growth by limiting dendritic cell number and function

Using a Kras-driven PDAC mouse model, Alam et al. showed that anti-TNFR1 Ab therapy or Tnfr1 KO increased TME numbers of DCs, IFNγ/TNFα-producing TILs, and T cell-dependent DC activation, and inhibited tumor growth. Anti-PD-1 treatment had a minor impact on s.c. PDAC growth in WT mice, but increased anti-TNFR1-induced IFNγ/TNFα production by TILs. anti-TNFR1 + anti-PD-1 treatment increased numbers/activation of DCs infiltrating Kras-driven spontaneous tumors. Anti-TNFR1 alone had a greater impact than Flt3L + anti-CD40 agonistic Ab, potentially due to inhibition of TNF/TNFR1-mediated DC apoptosis, shown in vitro. DC numbers inversely correlated with TNFR1 expression in human PDAC.

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.