Weekly Digests
‹ Back to January

Communication is key in PD-1 blockade

January 23, 2019

The efficacy of immunotherapy is dependent on a variety of factors and cells in the tumor microenvironment, and understanding these complex interactions is critical to understanding and improving the efficacy of treatment. To study some of these interactions, Garris and Arlauckas et al. used real-time intravital imaging, RNA sequencing, and a series of pathway manipulations to study crosstalk within the tumor microenvironment, and the role that it plays in the the efficacy of anti-PD-1 immunotherapy.

To begin, Garris and Arlauckas et al. used intravital imaging to track IFNγ- and IL-12-producing cells in mice bearing MC38 colon carcinoma during treatment with anti-PD-1 immunotherapy. As expected, anti-PD-1 increased the number of IFNγ-producing CD8+ T cells. It also increased the frequency of IL-12-producing cells, which were likely dendritic cells (DCs) based on their branched morphology. After treatment with anti-PD-1, these DC-like cells were observed deeper in the tumor and were found to be more motile. RNA sequencing confirmed that these cells were in fact a subset of DCs, related to the well-described cDC1 subset of DCs, though interestingly, they did not express CD103.

Selective depletion of DCs abrogated the antitumor effect of PD-1 blockade, as did neutralization of IL-12, confirming that cytokine support by dendritic cells was required for therapeutic efficacy. After ruling out direct activation of DCs by anti-PD-1, Garris and Arlauckas et al. used intravital imaging to track IL-12 and found that when CD8+ T cells were depleted, or when IFNγ was blocked, production of IL-12 was reduced in the TME, which was mediated by both reduced IL-12 production by DCs and a reduced number of IL-12-producing DCs. Together this suggested that dendritic cells are activated by sensing of IFNγ, which is produced by T cells in direct response to anti-PD-1.

While PD-1 blockade induced production of IFNγ and IL-12 and proliferation of CD8+ T cells within the TME, similar responses were not observed in draining lymph nodes, suggesting that pre-existing tumor-infiltrating T cells were the primary drivers of antitumor response in this model. To determine how IL-12 might impact this effector population, the researchers intratumorally administered IL-12 to MC38-bearing mice and found that IL-12 increased the frequency of infiltrating IFNγ+ cells and led to robust tumor regression. CD8+ T cells isolated from MC38 tumors also increased IFNγ production upon exposure to IL-12 ex vivo, indicating that IL-12 plays a direct role in activating T cells and maximizing IFNγ production.

To better understand the effect of IL-12 in humans, Garris and Arlauckas et al. examined skin lesion biopsies from 19 melanoma patients before and after administration of an IL-12-expressing DNA plasmid and found that intratumoral expression of IL-12 enhanced the expression of a cytolytic gene signature, which was further associated with therapeutic responses. To confirm that IL-12 was acting directly on infiltrating T cells in the human setting, the researchers isolated CD8+ T cells from 6 solid tumor samples samples (various tumor types) and found that exposure to IL-12 ex vivo increased IFNγ production in 5 of 6 samples, confirming that crosstalk observed in mice is likely consistent with human cancer.

Because IL-12 appears to be a critical component in inducing a strong IFNγ response, Garris and Arlauckas et al. explored whether agonizing IL-12-producing cells could be used to enhance responses to anti-PD-1 immunotherapy. To this end, the researchers tested the use of either agonistic CD40 monoclonal antibodies or a small molecule inhibitor of cellular inhibitor of apoptosis protein (cIAP) to trigger the non-canonical NF-κB pathway, which was found to be enriched in IL-12-producing DCs. Both drugs directly enhanced the production of IL-12 (shown using intravital imaging for CD40 agonist and in vitro for the small molecule inhibitor of cIAP), increased the frequency of IL-12-producing DCs in vivo, and showed incomplete antitumor efficacy in MC38-bearing mice. Combination treatment using CD40 agonist and anti-PD-1 outperformed either monotherapy and led to complete, durable antitumor responses in most MC38 tumor-bearing mice. Similar results were observed in the less immunogenic B16F10 melanoma model, with combination treatment leading to tumor rejection in 50% of mice, dependent on IL-12.

Overall, Garris and Arlauckas et al. show that anti-PD-1 directly activates T cell production of IFNγ, which goes on to induce IL-12 production by a subset of dendritic cells. This increase in IL-12 further licensed and maximized effector T cell responses and was essential for the efficacy of anti-PD-1 immunotherapy, challenging the simple model that direct activation of resident antitumor T cells by anti-PD-1 is sufficient for antitumor effects. In preclinical studies, agonizing the NF-κB pathway enhanced IL-12 production and improved the efficacy of PD-1 checkpoint blockade. Data from patient samples suggest that the crosstalk observed in mice is likely maintained in humans, suggesting that the mechanisms observed here may be relevant in ongoing and future clinical trials.

by Lauren Hitchings

References:

Garris C.S., Arlauckas S.P., Kohler R.H., Trefny M.P., Garren S., Piot C., Engblom C., Pfirschke C., Siwicki M., Gungabeesoon J., Freeman G.J., Warren S.E., Ong S., Browning E., Twitty C.G., Pierce R.H., Le M.H., Algazi A.P., Daud A.I., Pai S.I., Zippelius A., Weissleder R., Pittet M.J. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity. 2018 Dec 18.

In the Spotlight...

The route of administration dictates the immunogenicity of peptide-based cancer vaccines in mice

Sultan, Kumai, and Nagato et al. showed that intravenous administration of a peptide vaccine resulted in a substantially more efficient expansion of antigen-specific cytotoxic T lymphocytes (CTLs) compared with subcutaneous administration. 24 hours after i.v. but not s.c. injection, antigen-presenting cells in the spleen and multiple lymph nodes strongly and broadly presented antigen. The CTL response was highly polyclonal and the magnitude of the CTL response positively correlated with tumor growth rate reduction. Palmitoylated, amphiphilic antigens were more immunogenic than long peptide formulations.

Neoantigen characteristics in the context of the complete predicted MHC class I self-immunopeptidome

Having computationally identified the set of all peptides from the canonical human reference proteome that could be presented by nearly 3000 MHC class I variants (the “self-immunopeptidome”), Brown and Holt found that only about 30% of self-peptides were predicted to be presented. Self-immunopeptidome size and mutational load combined to approximate neoantigen load, which correlated with actual predicted neoantigen load and longer progression-free survival. Evidence supporting immunoediting of untreated TCGA tumors was observed and mutations in MHC-binding anchor positions appeared to confer enhanced immunogenicity.

A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma

In an ongoing phase I trial, 57 patients with relapsed/refractory multiple myeloma were treated with LCAR-B38M, a high-avidity CAR T cell therapy targeting two epitopes of the B cell maturation antigen. All patients experienced adverse events, with 90% reporting cytokine release syndrome (mostly grades 1 and 2); 65% had grade ≥ 3 adverse events. At data cutoff, overall response rate was 88% (68% CR, 5% very good PR, 14% PR), which was independent of BCMA expression levels. Median progression-free survival was 15 months; median overall survival was not reached. 20% of the initial responders progressed by the data cutoff date.

HLA-DO modulates the diversity of the MHC-II self-peptidome

To understand how the non-classical MHC II molecule HLA-DO (H2-O in mice) affects antigen presentation, Nanaware and Jurewicz et al. used MS to examine the ligandome of human lymphoblastoid cells. DO knockout reduced the total repertoire, shifted the abundance, and increased the measured affinity of observed epitopes, all consistent with an increase in HLA-DM editing (DO is an inhibitor of DM). In mice, knockout of H2-O resulted in reduced tolerance to some unmutated self-epitopes. This suggests that the modulation of DO expression, which occurs in APCs, is critical to both self-tolerance and response to invading microbes.

Tumor-derived IFN triggers chronic pathway agonism and sensitivity to ADAR loss

Liu et al. analyzed IFN-stimulated gene (ISG) signatures in TCGA tumors and patient-derived tumor xenografts, and found that some tumor cells constitutively produced IFN via a STING-dependent pathway. The chronic ISG state rendered tumor cells sensitive to loss of ADAR (an IFN-inducible enzyme that converts adenosine to inosine and prevents sensing of double-stranded RNA [dsRNA]) and upregulated dsRNA sensors PKR and MAVS. In the presence of unedited dsRNA, activated PKR led to enhanced IFN production, while MAVS amplified the ISGs. In the context of chronic ISG, ADAR loss reduced tumor growth in vitro and in vivo.

Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade

Tumors with loss of ADAR1 (an enzyme that converts adenosine to inosine and prevents sensing of double-stranded RNA [dsRNA]) expressed elevated levels of IFNβ and IFNγ, which in turn upregulated the transcription of dsRNA species; dsRNA remained unedited and was detected by dsRNA sensors PKR (leading to tumor growth arrest) and MDA5 (resulting in increased tumor inflammation and immune cell infiltration). Lack of ADAR1 sensitized tumors to irradiation or anti-PD-1 therapy, and overcame adaptive resistance to immunotherapy even when tumor cells could not be recognized by CD8+ T cells due to B2m loss.

Ex vivo conditioning with IL-12 protects tumor-infiltrating CD8+ T cells from negative regulation by local IFN-γ

Lin et al. studied the mechanism of improved antitumor efficacy of IL-12-pretreated, adoptively transferred (ADT) CD8+ T cells. IL-12 exposure before ADT of Pmel-transgenic CD8+ T cells enhanced persistence and IFNγ production within the B16F10 TME and dampened PD-1 and IFNγR2 expression. In human melanoma CD8+ TILs, IL-12 treatment in vitro reduced PD-1 and enhanced IFNγ levels. Pmel IFNγR1-/- CD8+ T cells, lacking IFNγ signaling, showed improved expansion, reduced apoptosis, and enhanced survival, suggesting that IL-12 suppression of the IFNγ receptor reduces responsiveness to IFNγ and its downstream negative regulatory effects.

A Potent Tumor-Reactive p53-Specific Single-Chain TCR without On- or Off-Target Autoimmunity In Vivo

Echchannaoui et al. analyzed the safety prolife of a high-affinity, single-chain mouse TCR specific for the HLA-A2.1-restricted, non-mutated human p53(264-272). The p53scTCR was created by covalently linking the variable TCRα and TCRβ domains and co-expressing them with a truncated constant TCRα domain to prevent TCR mispairing. Adoptive transfer of p53scTCR T cells to HLA-A2+ mice that lacked murine p53 or expressed wild-type human p53 did not induce autoimmune toxicities. In humanized syngeneic and xenograft mouse models, treatment led to tumor elimination and formation of an antigen-specific memory response.

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.