Weekly Digests
‹ Back to May

Why does PD-1 blockade need CXCR3 signaling? It’s not what you think

May 22, 2019

Motivated to extend the benefit of PD-1 blockade to more patients, Chow et al. explored the role that chemokines play in resistance to anti-PD-1 and found that the chemokine receptor CXCR3 and its ligand CXCL9 were crucial for the response to anti-PD-1 therapy in mouse tumor models. The results were recently published in Immunity.

The researchers began by showing that anti-PD-1 significantly reduced the growth of MC38 colon adenocarcinoma or D4M.3A.3 UV3 melanoma and improved survival in wild-type (WT) mice compared with Cxcr3-/- mice, demonstrating that CXCR3 plays a role in PD-1 blockade. Given that CXCR3 plays a role in the recruitment of T cells into the tumor in adoptive cell transfer models, the researchers hypothesized that the subpar efficacy of anti-PD-1 in Cxcr3-/- mice may be due to reduced intratumoral CD8+ T cell infiltration. However, pre-treatment MC38 tumor analysis showed no difference in the percentage, absolute number, cytokine production, or phenotype of intratumoral CD8+ T cells between WT and Cxcr3-/- mice. Thus, in this model, CXCR3 was not required for the early recruitment and function of CD8+ T cells.

A different scenario emerged post-treatment. Anti-PD-1 significantly increased the number of intratumoral CD8+ T cells, as well as the percentage of IFNγ+TNFα+ and Ki-67+granzymeB+ CD8+ T cells in WT, but not in Cxcr3-/- mice. To understand the mechanism behind these different outcomes, Chow et al. blocked the egress of T cells from lymphoid organs with FTY720 before anti-PD-1 treatment and showed that although peripheral T cell numbers were dramatically reduced, neither the intratumoral CD8+ T cell expansion nor the antitumor effect in WT mice were affected. Monitoring of DNA replication demonstrated that anti-PD-1 functioned primarily by expanding the pre-existing intratumoral CD8+ T cells, and not by recruiting more CD8+ T cells into the tumor and that this intratumoral T cell expansion was dependent on CXCR3. The researchers confirmed the requirement for CXCR3 signaling in PD-1 blockade response using chimeric mouse models, in which anti-PD-1 preferentially expanded and increased the effector function of WT neoantigen-specific intratumoral CD8+ T cells compared with their Cxcr3-/- counterparts.

The team further analyzed the role of CXCR3 using CXCR3-GFP reporter mice bearing MC38 tumors. They found that CXCR3-PD-1hiCD8+ T cells were more exhausted and produced lower levels of IFNγ and TNFα than their CXCR3-expressing counterparts. In addition, the percentage of CXCR3+CD8+ T cells decreased as the tumor grew over time, suggesting that the expression of CXCR3 on CD8+ T cells is inversely correlated with tumor growth.

The researchers then analyzed the expression of CXCR3 ligands – CXCL9 and CXCL10 – in WT mice. Anti-PD-1 increased the levels of both ligands in MC38 tumors, and the anti-PD-1 antitumor effect was abrogated when both ligands were blocked with antibodies. To tease out the specific role of each ligand, the team utilized separate Cxcl9- and Cxcl10-knockout mice, and found that CXCL9, but not CXCL10, was crucial for the anti-PD-1-mediated CD8+ T cell antitumor response.

Utilizing CXCL9-CXCL10 dual reporter mice, Chow et al. observed that myeloid cells were the major source of the two ligands within the MC38 tumors, with CD103+ DCs producing CXCL9 and CD11b+ DCs producing CXCL10. PD-1 blockade increased the production of CXCL9 by CD103+ DCs. Diphtheria-toxin-induced depletion of classical, but not monocyte-derived or plasmacytoid DCs, demonstrated that CD103+ DC-derived CXCL9 was functionally important for anti-PD-1-mediated antitumor response and improved survival. Based on these results, the authors hypothesized that PD-1 blockade-mediated increase in CXCL9 expression allowed CD103+ DCs to attract CXCR3+CD8+ T cells that were already in the tumor, thus facilitating DC-T cell interactions and enhancing T cell antitumor responses.

The researchers then screened solid murine tumors and found that the anti-PD-1-responsive tumors had a much higher baseline expression of CXCL9 and CXCL10 in CD11c+MHCII+ DCs than tumors resistant to PD-1 blockade, suggesting that baseline intratumoral expression of CXCR3 ligands could predict response to PD-1 blockade. Combination of anti-PD-1 and epigenetic modulators (DZNeP + 5-AZA-dC), which upregulated DC expression of CXCL9 and CXCL10, delayed tumor growth and extended survival in mice bearing anti-PD-1-resistant AT-3 breast tumors. These results suggested that tumors resistant to PD-1 blockade could become more responsive to anti-PD-1 when it is combined with epigenetic modulation of CXCR3 ligand expression; clinical trials examining the efficacy of this combination are ongoing.

Turning to human data, Chow et al. analyzed plasma samples from melanoma patients treated with anti-PD-1 alone or with anti-CTLA-4, and found that levels of CXCR3 ligands, measured a few months post-treatment, positively correlated with response. This data suggest that CXCR3 ligands could serve as early biomarkers of immunotherapy response.

by Anna Scherer

Meet the researcher

This week, Melvyn T. Chow, first author on this paper, took the time to answer our questions.

Lead author Andrew Luster (left) and first author Melvyn T. Chow (right).


1. What prompted you to tackle this research question?
A successful and efficacious immune checkpoint blockade therapy requires the productive interaction of different immune cells to generate an optimal antitumor response. Chemokines likely play key roles in the recruitment of immune cells into tumors as well as their cell-cell interactions within tumors. Therefore, we were interested in understanding the role of the CXCR3 chemokine system in the recruitment and cellular interactions of T cells within the tumor microenvironment, ultimately with the goal of exploring the potential for specific enhancement of effector T cell infiltration and function.

2. What was the most surprising finding of this study for you?
The most surprising finding of this study was the important role the CXCR3 chemokine system plays for the activation of CD8+ T cells within tumors and for the success of PD-1 immune checkpoint blockade therapy.

3. What was the coolest thing you’ve learned (about) recently outside of the lab?
I was traveling in Iceland early March with my wife and one of the coolest things I learned on this trip is that in Iceland, hot water is pumped through tubes under many of the sidewalks to keep the streets free of ice and snow without plowing or shoveling. Furthermore, almost 100 percent of the electricity consumed in Iceland comes from renewable energy, which I find remarkable.

References:

Chow M.T., Ozga A.J., Servis R.L., Frederick D.T., Lo J.A., Fisher D.E., Freeman G.J., Boland G.M., Luster A.D. Intratumoral Activity of the CXCR3 Chemokine System Is Required for the Efficacy of Anti-PD-1 Therapy. Immunity. 2019 May 6.

In the Spotlight...

Targeting TMEM176B Enhances Antitumor Immunity and Augments the Efficacy of Immune Checkpoint Blockers by Unleashing Inflammasome Activation

Segovia et al. show that the transmembrane protein 176B (TMEM176B) suppresses antitumor immunity by reducing the accumulation of cytosolic Ca2+ within DCs and macrophages, which prevents activation of the NLRP3 inflammasome and the release of activated (cleaved) caspase-1. Thus, IL-1β and pro-IL-18 cannot be processed into their active, pro-inflammatory forms. Tmem176b-/- mice showed increased CD8+ T cell infiltration into tumors and improved responses to checkpoint blockades; BayK8644 was found to inhibit TMEM176B and recapitulate these effects. An inflammasome-activated signature was identified in patients responding to anti-PD-1.

Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response

Baeuerle et al. engineered TCR fusion constructs (TRuCs) comprised of an antibody-based binding domain fused to the extracellular N-terminus of either CD3α,β,γ,δ or ε.TRuCs were integrated into intact TCR constructs, reprogramming them to recognize tumor surface antigens; target recognition engaged the full signaling capacity of the TCR complex. Compared to second generation CAR T cells, TRuC T cells were as capable of recognizing and killing target antigen-expressing cells in vitro, but produced lower levels of cytokines. In vivo, TRuCs more effectively controlled the growth of both solid and liquid tumors and extended survival.

Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy

Perez-Ruiz et al. show that administration of clinically available TNF inhibitors before or during dual anti-CTLA-4 and anti-PD-1 therapy prevents the emergence or exacerbation of immune-related adverse events (colitis) in mice, while maintaining or slightly enhancing CD8+ T cell infiltration and antitumor efficacy. TNF inhibition was associated with expansion of PD-1- T cells and attenuation of activation-induced cell death. The TNF pathway was active in patients who developed ICB-induced colitis, indicating that prophylactic TNF inhibition may improve the clinical safety of ICB and allow for more potent dosing.

A library of Neo Open Reading Frame peptides (NOPs) as a sustainable resource of common neoantigens in up to 50% of cancer patients

Frameshift mutations can produce long neoantigens that may be highly immunogenic. Based on analysis of TCGA data, Koster and Plasterk describe that many different upstream frameshift mutations terminate with the same common region (Neo Open Reading Frame peptides; NOPs). Each individual NOP across the genome is predictable and could arise in multiple patients, and thus the total set of NOPs could serve as a pre-prepared, off-the-shelf vaccine library from which neoantigens could be immediately selected once frame-shift mutations are identified. A library of roughly 1,200 peptides would be relevant to about 30% of patients.

Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy

Chauhan, Chen, and Tong et al. developed an angiotensin receptor blocker (TMA-ARB) nanoparticle comprising valsartan (an ARB) conjugated with a polymer that breaks down in the slightly acidic TME. TMA-ARB reduced hypotension and increased levels of intratumoral ARB compared with free ARB, reprogrammed cancer-associated fibroblasts from immunosuppressive to immunosupportive by reducing hypoxia and TGFβ production, and increased the activation of CD8+ T cells. Combination of TMA-ARB with anti-CTLA-4 and anti-PD-1 delayed tumor growth and prolonged survival in primary and metastatic murine models of breast cancer.

Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response

Mandal and Samstein et al. investigated the variable response to anti-PD-1 in patients with mismatch repair-deficient (MMR-d) tumors. Quantification of microsatellite instability (MSI) in both MSH2 knockout murine tumor cell lines and patients with MMR-d tumors revealed a range of MSI intensity (and consequent mutation load). Responses to anti-PD-1 were associated with high MSI scores, and in particular, an accumulation of insertion-deletion (indel) mutations. An analysis of murine tumor genomes supported evolution through immunoediting. Genome-wide characterization of MSI intensity may help predict responses to anti-PD-1 in MMR-d patients.

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.