Weekly Digests
‹ Back to July

CXCR3 on Tregs controls their accumulation, activation

July 5, 2023

Regulatory T cells (Tregs) often present a challenge to antitumor immunity, but exactly how they suppress immune responses is not entirely understood. Investigating the accumulation and activation of Tregs in tumors, Moreno Ayala et al. found that CXCR3+ Tregs interacted preferentially with CXCL9+ DC1s in tumors, and that genetic knockout of CXCR3 specifically in Tregs enhanced DC1 cross-presentation of tumor antigens, CD8+ T cell priming, and antitumor immunity. Their results were recently published in Immunity.

To begin, Moreno Ayala et al. investigated T cell phenotypes in mice bearing MC38 colon carcinoma, EL4 thymic lymphoma, or 9464D neuroblastoma tumors. While the frequencies of Tregs and CD8+ T cells varied across tumors, there was consistently a positive correlation between the two populations. The researchers also noted that CXCR3 expression was increased on CD8+ T cells, effector CD4+ T cells, and Tregs in tumors and draining lymph nodes relative to healthy tissues and peripheral lymph nodes, respectively.

An evaluation of dynamics showed that peak CXCR3 expression on Tregs correlated with peak CXCR3+CD8+ T cell infiltration into tumors, at which point Tregs began accumulating in dLNs. At the peak of tumor infiltration, antigen-experienced (CD44+) CXCR3+ Tregs showed increased markers of activation and effector function (CD69, CD103, KLRG1, PD-1, and CTLA-4) compared to CD44+CXCR3- Tregs. Together these results suggest that CXCR3 expression supports enhanced accumulation and activation of Tregs in tumors.

To study the effects of CXCR3 on Tregs specifically, Moreno Ayala et al. generated genetic mouse models that take advantage of X chromosome inactivation in female mice. In these models, the mice contained heterogeneous populations of CXCR3+ and CXCR3KO Tregs, with the option to deplete one Treg population or another (depending on the model) with the administration of diphtheria toxin. In mice depleted of CXCR3+ Tregs (leaving only CXCR3KO Tregs), growth of MC38, EL4, and 9464D tumors was delayed compared to when CXCR3+ Tregs were left intact or CXCR3KO cells were depleted. This effect was not observed when a similar proportion of Tregs was depleted irrespective of CXCR3, suggesting that CXCR3+ Tregs specifically play a role in suppressing antitumor effects.

Investigating this role further, the researchers found that in the setting of CXCR3KO Tregs, CD8+ T cells, including a population of tumor antigen-specific CD8+ T cells, were increased in tumors across MC38, EL4, and 9464D models. CXCR3 deficiency in Tregs did not affect the activity of CD8+ T cells, as measured by IFNγ and TNFɑ production upon ex vivo restimulation, nor did it appear to affect PD-1 expression, suggesting that CXCR3+ Tregs limit CD8+ T cell accumulation, but not activation. Further, the addition of anti-PD-1 could be used to enhance antitumor efficacy. Looking at the accumulation of Tregs in tumors, the researchers noted a modest reduction in Treg frequency associated with CXCR3 deficiency. Similar effects were also observed in a genetically engineered mouse model of sarcomagenesis (non-transplantable), in which depletion of CXCR3+ T cells after the formation of sarcomas led to increased intratumoral CD8+ T cells and reduced Tregs.

Looking at the quality of CXCR3KO Tregs, the researchers noted reduced expression of PD-1 and CTLA-4 throughout MC38 tumor progression, indicative of a less activated phenotype. In a direct competitive setting in mice with both CXCR3+ and CXCR3KO Tregs intact, CXCR3KO Tregs made up a smaller proportion of Tregs in tumors and dLNs and expressed less PD-1 and CTLA-4 compared to CXCR3+ cells, suggesting again that CXCR3 contributes to preferential Treg accumulation and activation in tumor tissues. Going a step further, the researchers tested the accumulation of CXCR3+ Tregs directly by adoptively transferring ex vivo-activated and -differentiated Th1-like Tbet+ Tregs with or without CXCR3 expression. After transfer to MC38 tumor-bearing mice, CXCR3KO Tregs were enriched in dLNs, but reduced in tumors compared to WT Tregs 24 hours after transfer, suggesting that CXCR3 expression itself was critical for Treg accumulation in tumors.

Next, the researchers hypothesized that CXCR3 in Tregs may function by promoting Treg colocalization with and regulation of DCs, which are known to produce the CXCR3 ligands CXCL9 and CXCL10 in C57BL/6 mice. To study this, the researchers developed a fluorescently tagged mouse model that allowed them to visualize and quantify interactions between Tregs and CD11c+ DCs as well as between CD8+ T cells and DCs. This showed that CXCR3-deficient Tregs were less likely to interact with DCs than CXCR3-competent Tregs, while CD8+ T cells increased and were more likely to interact with DCs when Tregs were CXCR3-deficient. In a competitive setting with both CXCR3+ and CXCR3KO cells intact, these results for Treg–DC interactions were similar, but the effect on CD8–DC interactions was abrogated.

Looking at CXCL9 and CXCL10 production, the researchers found that CXCL9 was produced almost exclusively by DCs, and that the number of CXCL9+CD11c+ cells positively correlated with the number of wild-type Tregs. This positive correlation was weaker when CXCR3 was knocked out in Tregs, and was not observed with CXCL10, which was not exclusive to DCs. CXCL9 was reduced in the absence of CD8+ T cells or IFNγ, supporting a hypothesis that IFNγ production by CD8+ T cells may contribute to DC upregulation of CXCL9, which in turn recruits CXCR3+ Tregs. This hypothesis was further supported by adoptive transfer experiments, which showed that the enhanced accumulation of CXCR3+ Tregs in tumors was lost in DC1-deficient mice.

Finally, the researchers hypothesized that CXCR3+ Treg cells may regulate the capacity of DC1s to stimulate antitumor CD8+ T cell responses, and tested their hypothesis using MHC-1-deficient MC38 tumors expressing OVA, which allowed the researchers to measure CD8+ T cell responses to DC cross-presentation. This revealed a significant increase in OVA-specific CD8+ T cells within tumors and dLNs in the context of CXCR3KO Tregs, but not CXCR3+ Tregs. This effect was dependent on the presence of DC1s. Enhanced cross-presentation of tumor antigens by DC1s was further confirmed using antibodies that bind to SIINFEKL-loaded H-2Kb molecules.

Altogether, these results describe a mechanism by which IFNγ from CD8+ T cells enhances CXCL9 expression by DC1s, which recruits CXCR3+ Tregs. These CXCR3+ Tregs in turn increase CXCL9+ DC1s, but limit DC1 cross-presentation of tumor antigens, impeding priming of antitumor CD8+ T cells and limiting their control over cancer progression. CXCR3 knockout in Tregs enhanced DC1 cross-presentation, CD8+ T cells, and antitumor efficacy, suggesting that CXCR3 could serve as a potential target for cancer immunotherapy.

Write-up and image by Lauren Hitchings

References:

Moreno Ayala MA, Campbell TF, Zhang C, Dahan N, Bockman A, Prakash V, Feng L, Sher T, DuPage M. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8+ T cell antitumor immunity. Immunity. 2023 Jun 27.

In the Spotlight...

Perforin-2 is a pore-forming effector of endocytic escape in cross-presenting dendritic cells

Using a translation-inhibition flow cytometry assay for CRISPR-Cas9 screening of 281 genes highly expressed in cDC1s, but not cDC2s, Rodriquez-Silvestre et al. identified perforin-2 (Mpeg1) as an APC-restricted pore-forming protein that regulates antigen transit from endosomes to cytosol. In vitro, endocytic escape was impaired in Mpeg1-/- DCs and enabled in non-immune cells by ectopic perforin-2 expression. Cell-associated antigen priming of CD8+ T cells was impaired in Mpeg1-/- mice. Perforin-2 was shown to reside mostly in lysosomes at steady state, transit through endosomes, and undergo proteolytic processing in low-pH antigen-containing compartments.

Contributed by Paula Hochman

B-cell-specific checkpoint molecules that regulate anti-tumour immunity

Sequencing B cells in B16-F10 tumors and lymph nodes, Bod et al. identified a cluster that expanded in the draining lymph nodes and expressed Havcr1 (encoding TIM-1) and other checkpoint genes. Havcr1 knockout in B cells increased frequencies of cytotoxic T cells in tumors and restricted tumor progression in multiple models. Anti-TIM-1 mAbs also slowed tumor growth. Havcr1-/- B cells upregulated antigen presentation and IFN-responsive genes, including IFNAR, and induced IFNγ expression while reducing Foxp3 in CD4+ T cells. The Havcr1 B cell signature was also found in human tumor datasets and was reduced in ICB-responding patients.

Contributed by Alex Najibi

c-Kit signaling potentiates CAR T cell efficacy in solid tumors by CD28- and IL-2-independent co-stimulation

Xiong et al. investigated the impact of c-Kit signaling on CAR T cell therapy for solid tumors. CAR T cells expressing a mutated c-KIT (KITv) that constitutively activates cKIT via phosphorylated STAT exhibited enhanced antigen-dependent growth, CD28/IL-2 independent co-stimulation, and increased IFNγ production with enhanced cytotoxicity compared to controls. In multiple solid tumor models, KITvCAR T cells improved survival, functioned well in TGFβ-high and antigen-low models, and were susceptible to TKI therapy. KITvCAR T cells were additive with CD28 costimulation and exhibited comparable or better in vivo efficacy than second-generation CARs.

Contributed by Katherine Turner

A platform trial of neoadjuvant and adjuvant antitumor vaccination alone or in combination with PD-1 antagonist and CD137 agonist antibodies in patients with resectable pancreatic adenocarcinoma

Based on analysis of resected tumors after neoadjuvant vaccination with GVAX (allogeneic GM-CSF-secreting whole-cell pancreatic cancer vaccine) + low-dose cyclophosphamide (cy-GVAX; arm A) or cy-GVAX + nivolumab (arm B), Heumann et al. added a CD137 agonist, urelumab (arm C). The post-treatment density of CD8+CD137+ TILs within tertiary lymphoid aggregates in the TME was significantly higher in Arm C than in arms A or B. Arm C showed a numerical improvement in disease-free and overall survival compared to arms A and B; no differences were detected between arms A and B.

Contributed by Maartje Wouters

Cancer cell plasticity and MHC-II-mediated immune tolerance promote breast cancer metastasis to lymph nodes

Lei and Pereira et al. performed a comprehensive analysis of breast cancer progression to lymph node metastases (LNM) at a single-cell resolution, and identified heterogeneous and spatially organized epithelial and mesenchymal phenotypes, with a subset of cancer cells expressing higher levels of the MHC-II gene in both human and mouse tumors. Invasion of MHC-II+ cancer cells into the lymph node expanded Tregs and induced immune tolerance. Overexpression of Ciita, an MHC-II transactivator, in 4T1 mouse tumor cells increased Treg expansion and LNM, whereas MHC-II knockout inhibited Treg expansion and LNM.

Contributed by Shishir Pant

PD-1 maintains CD8 T cell tolerance towards cutaneous neoantigens

Damo et al. used NINJA mice to show that de novo skin-specific expression of T cell antigen in the epidermis and CPI treatment led to elimination of antigen-expressing cells and localized cutaneous disease in the mouse skin, recapitulating features of CPI-dependent lichenoid IRAEs in humans. Skin-specific antigen expression caused local infiltration of antigen-specific CD8+ T cells with an effector gene-expression profile into the dermis. PD-1 prevented epidermal infiltration of CD8+ T cells and maintained skin tolerance, which was reversed in PD-1 knockout mice. Tolerance was associated with increased interactions between antigen-specific CD8+ T cells and myeloid cells.

Contributed by Shishir Pant

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.