Weekly Digests
‹ Back to June

A Fountain of Youth for T cell Function

June 16, 2021

With advancing age comes a decrease in immune function, which can also dampen responses to immunotherapy. Investigating this, Nakajima et al. compared T cell subsets, based on their expression of CD44 and CD62L, in young versus aged mice. Among CD8+ T cells, CD44lowCD62Lhigh (P1) cells are defined as naive, CD44highCD62Lhigh (P2) cells are defined as central memory, and CD44highCD62Llow (P3) cells are defined as effector memory. Interestingly, CD44lowCD62Llow (P4) cells, which are rare and poorly defined, expanded after tumor inoculation in young, but not old mice, prompting further investigation into this subset. The findings were recently published in PNAS.

Using PD-1-knockout and wild-type mouse models, Nakajima et al. showed that aged mice failed to mount strong antitumor immunity and failed to generate P4 cells upon tumor inoculation. Looking more closely into this cell subset, they found that P4 cells had the potential for antitumor activity, as adoptive transfer of P4 cells controlled tumors as well as adoptive transfer of P3 cells. By day 6 after transfer, all of the transferred P4 cells had differentiated into P3 cells, suggesting that P4 cells may mediate antitumor immunity as precursors to effector/memory cells. Ex vivo stimulation studies showed that P4 cells differentiated from P1, but not P2 cells.

Looking more closely at gene expression in the P4 subset, the researchers found that genes related to activation were higher in P4 cells than in P1 cells, and genes associated with differentiation were lower in P4 cells than in P3 cells, further supporting the notion that P4 cells are pre-effector-like cells in an early activation state. Next, gene ontology enrichment analysis revealed that genes for enzymes associated with 1 carbon (1C) metabolism (a set of biochemical reactions that provide single carbon methyl units for multiple biosynthetic pathways) were uniquely upregulated in P4 cells. Genes for 1C metabolism were enriched upon tumor inoculation in young, but not aged mice, in line with previous data on the generation of the P4 subset. This was further accompanied by an increase in mitochondrial function. Consistent with this, T cells from aged tumor-bearing mice showed overall lower rates of oxygen consumption compared to their younger counterparts.

To better understand why P1 cells fail to differentiate into P4 cells in aged mice, Nakajima et al. found that TCR signaling was attenuated in P1 cells from older mice, as evidenced by limited ZAP-70 phosphorylation. After showing that TCR expression levels and avidity were unchanged, the researchers turned their attention towards CD45RB (the major CD45 isoform in naive and memory T cells in mice), which, when present at a high density on the cell surface, can weaken TCR signaling in naive T cells. In bulk CD8+ T cells from aged mice, CD45RB expression was higher, and this effect was greater when looking specifically at the P1 and P4 subsets. When CD45RB was blocked using a CD45 inhibitor, ZAP-70 phosphorylation increased in both aged and young mice, suggesting that CD45RB suppresses CD8+ T cell activation and differentiation from P1 to P4 by interfering with TCR signal transduction.

Interestingly, when P1 cells from aged mice were stimulated ex vivo using  anti-CD3/CD28 mAbs, they differentiated into P4 and P3 cells as effectively as P1 cells from young mice. This suggested that while nominal antigenic stimulation was insufficient to generate P4 cells, strong TCR stimulation could overcome this deficit.

In an effort to restore differentiation from P1 to P4 cells in aged mice, the researchers introduced xenogeneic cells (Daudi cells) to induce strong antigenic stimulation. Following injection of Daudi cells, the researchers observed increased P4 cells, increased 1C metabolism, and reduced CD45RB in P1 cells in both young and aged PD-1-knockout mice. When aged PD-1-knockout mice treated with Daudi cells were inoculated with MC38 tumors, they showed decreased tumor growth and increased survival compared to untreated mice. In wild-type mice, age-related unresponsiveness to PD-1 blockade was also recovered with Daudi cell injection, or with injection of splenocytes from Balb/c mice, which also served as effective alloantigens.

Looking more closely, the researchers found that Daudi cells increased phosphorylation of ZAP-70 in T cells, and increased the proliferation of tumor antigen-reactive CD8+ T cells. Within tumors, most CD8+ T cells were already differentiated into P3 cells, suggesting that strong antigen stimulation enhances the differentiation of antigen-specific T cells from naive to effector, thus increasing effector T cell infiltration into tumors.

Investigating the potential of using Daudi cells in a therapeutic setting, the researchers tested whether administration of Daudi cells could restore antitumor immunity in mice with established tumors. In this setting, Daudi cells increased P4 cells, TILs, and ZAP-70 phosphorylation. These results suggest that strong immune stimulation by xenogeneic or allogeneic cell transplant could potentially recover TCR signal transduction and P4 generation to rescue compromised antitumor immunity and responses to PD-1 checkpoint blockade in aged mice.

Overall, this research highlights the importance of CD44lowCD62Llow (P4) CD8+ T cells as an intermediate cell type between naive (P1) and effector/memory (P3) CD8+ T cells. The P4 subset expresses genes associated with 1C metabolism and mediates enhanced mitochondrial function that is required for sufficient T cell activation. In aged mice, accumulation of CD45RB in P1 cells attenuates TCR signaling and limits the formation of the P4 subset. Nakajima et al. showed that this limitation can be overcome by CD45 inhibition or strong antigenic stimulation through the introduction of xenogeneic or allogeneic cells. This work provides important insights that could lead to the development of new targets to enhance antitumor immunity and restore responses to checkpoint blockade in older patients, who are often less likely to reap the benefits of immunotherapy.

By Lauren Hitchings

Meet the researcher

This week, first author Yuka Nakajima answered our questions.

What prompted you to tackle this research question?

Despite clinical studies showing a remarkable efficacy of cancer immunotherapy by PD-1 blockade, many cancer patients cannot respond to the therapy. Further, while previous work has given us information about the linkage between aging and impaired immune responses, little is known about how aging affects the efficacy of PD-1 blockade therapy. Actually, aged PD-1 knockout mice showed strong resistance to tumor growth. Therefore, we tried to better understand the mechanism using the mouse model for PD-1 blockade therapy to overcome the resistance.

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

The most remarkable finding was the observation that the reduction of CD44lowCD62Llow cell frequency could be correlated with resistance to PD-1 blockade therapy in aged mice. In mouse CD8+ T cells, the CD44lowCD62Llow subset had been known as a remaining minor subset, an exception to the naive, effector, and memory subsets, and the importance was not known. Normally, it is difficult to detect the CD44lowCD62Llow cells because of the transient induction from naive cells in response to TCR stimulation. Luckily, we could use PD-1 KO mice that can strongly induce TCR-dependent T cell activation. Therefore, we could easily detect CD44lowCD62Llow cells in those mice. This finding helped us better understand how aging modulates antitumor immunity through CD8+ T cell differentiation.

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

Recently, I found a very beautiful cherry blossom area, when I took a walk for a change of pace during telework. Although the area was very close to my house, I didn’t know it at all. Due to the COVID-19 pandemic, we have to change our lifestyle to adapt to the new normal. Under the circonstances, I realized, “in the middle of difficulty lies opportunity”!

References:

Nakajima Y., Chamoto K., Oura T., Honjo T. Critical role of the CD44lowCD62Llow CD8+ T cell subset in restoring antitumor immunity in aged mice. Proc Natl Acad Sci U S A. 2021 Jun 8.

In the Spotlight...

Tim-4+ Cavity-Resident Macrophages Impair Anti-Tumor CD8+ T cell Immunity

Chow et al. showed that pleural and peritoneal cavity-resident Tim-4+ (a phosphatidylserine (PS) receptor) macrophages suppress the efficacy of immunotherapies in these anatomic spaces. Metastatic involvement of serous body cavities is associated with poor clinical outcomes after ICB therapy, and Tim-4 expressed on cavity-resident macrophages is inversely correlated with CD39+CD8+ T cells. Tim-4 abrogation enhanced anti-PD-1 therapy efficacy in mice. PShigh CD8+ T cells showed enhanced expression of proliferation- and cytotoxicity-associated genes and Tim-4+ macrophages functionally sequestered and impaired proliferation of PShigh CD8+ T cells.

Contributed by Shishir Pant

Targeted delivery of CRISPR-Cas9 and transgenes enables complex immune cell engineering

To facilitate genetic editing of T cells without electroporation, Hamilton and Tsuchida et al. used virus-like particles to deliver Cas9 ribonucleoprotein complexes (Cas9-VLPs). Cas9-VLPs targeting B2M effectively knocked out B2M expression on murine T cell lines and primary human T cells, and could also co-deliver a CAR transgene, enabling one-step production of CAR T cells with additional gene editing. The transduction efficiency and tropism depended on the VLP surface glycoprotein, and Cas9-VLPs engineered with an HIV-1 envelope glycoprotein could preferentially transduce CD4+ T cells in the presence of CD8+ T cells.

Contributed by Alex Najibi

In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models

Oba and Makino et al. generated iPSC-derived cDC2s. In syngeneic orthotopic mouse models of immunologically cold melanoma and breast cancer, intratumoral iPSC-DC injection + tumor irradiation (RT) synergized to delay growth of directly treated and, in a bilateral model, distal tumors. RT induced tumor-injected iPSC-DCs to migrate to tumor-draining lymph nodes, express CD40, and aggregate with other DCs and CD8+ T cells. RT + iPSC-DC tumor injection increased tumor-specific Slamf6+PD-1int exhausted progenitor CD8+ T cells, increased PD-L1 on TAMs and DCs in tumors, induced tumor-specific immune memory, and enhanced the efficacy of PD-L1 blockade.

Contributed by Paula Hochman

REVIEW: Cancer-associated fibroblasts: Key players in shaping the tumor immune microenvironment

Desbois and Wang summarized the roles of cancer-associated fibroblasts (CAFs) in the TME and their impact on immunotherapy efficacy. Using FAP, SMA, and CD29 markers, four functionally distinct CAF populations were identified in breast and ovarian cancer, including immunosuppressive CAF-S1(FAPhiCD29med/hiSMAhi) and contractile CAF-S4 (FAP-CD29hiSMAhi). In tumors with efficient immune cell infiltration, CAFs have antitumor roles (CD8+ T cell recruitment and cell trafficking) and pro-tumor roles (immunosuppressive cell recruitment and polarization, and Treg activation and retention).

Contributed by Katherine Turner

CSF-1/CSF-1R Signaling Inhibitor Pexidartinib (PLX3397) Reprograms Tumor-Associated Macrophages and Stimulates T-Cell Infiltration in the Sarcoma Microenvironment

Fujiwara et al. demonstrated that PLX3397, a potent small molecule CSF-1R inhibitor, reprograms tumor-associated macrophages (TAMs) to exert its antitumor effects. Osteosarcoma and fibrosarcoma cells secreted CSF-1 in conditioned medium, and, in an in vitro TAM model, CSF-1R blockade reduced M2 polarization, proliferation, and chemotaxis in bone marrow-derived macrophages. Systemic administration of PLX3397 in a mouse osteosarcoma model suppressed primary tumor growth and lung metastasis, improved metastasis-free survival, reduced TAMs and Foxp3+ Tregs, and increased the CD8+ T cell infiltration in the TME.

Contributed by Shishir Pant

Long-Term Antitumor CD8+ T Cell Immunity Induced by Endogenously Engineered Extracellular Vesicles

Ferrantelli et al. described a vaccine platform consisting of DNA vectors expressing a tumor antigen (either HPV16 E7 or E6 in this report) linked to a protein (Nefmut) that anchors to the inner membrane of naturally occurring extracellular vesicles (EVs). EVs from cells transfected with a Nefmut/tumor antigen vector contained both tumor antigen and Nefmut. Compared to non-Nefmut-containing vectors, i.m. DNA immunization with Nefmut-containing vectors induced enhanced tumor-specific polyfunctional CD8+ T cell responses and slowed the growth of established tumors – curing them in 7 of 12 cases. Cured mice were resistant to tumor rechallenge.

Contributed by Margot O’Toole

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.