Weekly Digests
‹ Back to September

RNA to the rescue: localized cytokine delivery shapes antitumor responses

September 22, 2021

Treatment of tumors with cytokines to boost antitumor immune responses is limited in the clinic due to systemic adverse events. Intratumoral delivery of cytokines may overcome these issues and improve therapeutic efficacy. Hotz and Wagenaar et al. thoroughly investigated the efficacy of local delivery of mRNA encoding a mixture of cytokines alone or combined with checkpoint blockade in murine models to take advantage of these powerful immune stimulants. Their work was published in Science Translational Medicine.

Based on in vivo screens, the researchers systematically established a cocktail of saline-formulated mRNAs encoding four murine cytokines: IL-12 single chain, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-15 sushi, and IFN-α4. Local administration of these mRNAs resulted in cytokine expression in the tumor microenvironment (TME), with limited expression in other organs, suggesting localized translation. In the TME, both CD45+ and CD45- cells were able to take up and translate the mRNA.

Repeated intratumoral injection resulted in tumor growth control, with complete regression (CR) in the majority of B16F10 and CT26 tumors, resulting in long-term survival. The cytokine expression also resulted in the induction of IFNγ and IP-10. To investigate the mechanistic contribution of each cytokine in the mixture, the researchers performed experiments with bone marrow cells and splenocytes that were exposed to the four cytokines and mixtures in which one of the cytokines was left out. When IL-15 sushi or IFN-α4 were missing, CD8+ T cells, NK cells, and DCs expressed lower levels of activation markers, and when GM-CSF or IL-12sc were missing there was less induction of IFNγ. Therapeutic responses were reduced most when IL-12sc was omitted, but all four were necessary for maximal effects. This was also shown in vivo, where all three-component mixtures of mRNAs resulted in varying levels of CT26 tumor control, with removal of IL-12sc mRNA having the most pronounced effect. In the B16F10 model, some of the CRs developed localized vitiligo, suggesting induction of T cell responses toward shared tumoral and melanocyte antigens. In addition, mice with CR were resistant to tumor rechallenge, suggesting the development of immunologic memory. This was further confirmed in immune cell depletion experiments, which showed that the antitumor activity was dependent on the presence of NK cells and CD8+ and CD4+ T cells.

Post-treatment, there was a higher infiltration of CD4+ and CD8+ T cells in the TME, and these TIL expressed higher levels of granzyme B, TNFα, and IFNγ. RNAseq of treated tumors showed upregulation of proinflammatory signatures, including the IFNγ and TNFα signaling pathways. In addition, cytokine mRNA treatment resulted in PD-L1 upregulation on immune and non-immune cells in the TME, while levels of PD-1 on CD8+ T cells remained unaffected. Further confirming the essential role of the IFNγ pathway in the therapeutic effects, IFNγ knockout mice with B16F10 tumors did not respond to therapy.

Hotz and Wagenaar et al. then investigated how the cytokines may affect T cell priming by using a B16F10-ovalbumin (OVA) model together with OVA-specific OT-I CD8+ T cells. Treatment with the mRNA mixture resulted in higher OT-I proliferation in the tumor-draining lymph nodes. Similar results were found in a CT26 model expressing the antigen gp70, where treatment resulted in the expansion of peripheral and tumoral gp70-specific T cells. Rechallenge experiments with CT26 tumors lacking gp70 revealed that immune memory directed against dominant and subdominant antigens was formed.

Treatment of subcutaneous B16F10 tumors also suppressed the growth of lung metastases, resulting in reduced lung tumor burden. In these metastatic lesions, NK cells, but not CD8+ T cells, upregulated CD69 and KLRG1. In a B16F10 model with subcutaneous tumors on both flanks, treatment in one flank was effective for both tumors, resulting in longer overall survival, suggesting local treatment results in systemic antitumor responses.

The researchers then investigated whether the combination of the mRNA treatment protocol with checkpoint blockade could further enhance efficacy. Although anti-CTLA-4 alone showed efficacy in this model, adding an anti-CTLA-4 antibody to a single intratumoral injection of the cytokine mRNA mixture in the CT26 and B16F10 models resulted in improved antitumor activity and survival. When mice with larger tumors were treated, there were more complete responses after combination treatment than after single treatment with mRNA. When PD-1 blockade was combined with the mRNA therapy in the B16F10 and MC38 models with tumors at both flanks or the B16F10 metastasis model, survival increased; in the metastasis model, 40% of mice experienced CR.

Given that many tumors are resistant to checkpoint inhibition, therapy efficacy was also assessed in a model in which β2M was knocked out of MC38 cells by CRISPR to model antigen presentation loss. These mice did not respond to anti-PD-1 single treatment. Injection of the cytokine mRNA mixture prolonged survival, but no additional effect in efficacy was observed when it was combined with checkpoint blockade. To model heterogeneous tumors, mice were inoculated with the MC38 β2M knockout cells on one flank, and MC38 WT tumor cells on the other flank. Anti-PD-1 alone was ineffective in this model. The knockout tumors were injected with the mRNA mixture, which temporarily improved outcomes, but the combination with anti-PD-1 further improved survival, suggesting abscopal effects with the combination.

Finally, the researchers prepared human mRNAs for the four cytokines to inject into a human A375 melanoma xenograft in SCID mice. All four cytokines were expressed in the 24 hours after injection, after which levels lowered to baseline at 72 hours post-injection. In vitro experiments using PBMCs from patients with advanced-stage cancer revealed that treatment with supernatants from individual cytokine mRNA-transfected cells induced the release of high levels of IFNγ. When combined with anti-CD3 stimulation, treatment resulted in increased CD8+ T cell proliferation in 3 out of 4 samples and CD4+ T cell proliferation in 2 out of 4 samples.

Overall, local intratumoral injection of a saline-formulated mRNA mixture encoding 4 distinct cytokines could improve the immune environment in tumors – an effect that was further enhanced by combining this strategy with checkpoint blockade. Clinical studies with this mRNA mixture are currently underway.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Hotz C., Wagenaar T.R., Gieseke F., Bangari D.S., Callahan M., Cao H., Diekmann J., Diken M., Grunwitz C., Hebert A., Hsu K., Bernardo M., Karikó K., Kreiter S., Kuhn A.N., Levit M., Malkova N., Masciari S., Pollard J., Qu H., Ryan S., Selmi A., Schlereth J., Singh K., Sun F., Tillmann B., Tolstykh T., Weber W., Wicke L., Witzel S., Yu Q., Zhang Y.A., Zheng G., Lager J., Nabel G.J., Sahin U., Wiederschain D. Local delivery of mRNA-encoding cytokines promotes antitumor immunity and tumor eradication across multiple preclinical tumor models. Sci Transl Med. 2021 Sep 8.

In the Spotlight...

Antigen and checkpoint receptor engagement recalibrates T cell receptor signal strength

Elliot et al. used a reporter mouse model, Tg4 Nr4a3-Tocky, to monitor the temporal dynamics of TCR signaling, track T cell activation in vivo, and identify quantitative and qualitative changes in T cells receiving different strengths of TCR signaling. Several key transcriptional programs were identified, and expression of co-inhibitory molecules, particularly PD-1, rapidly recalibrated the TCR activation threshold of T cells. Anti-PD-1 increased the probability of T cell reactivation and resulted in stronger TCR signaling. A signature for genes upregulated in response to strong TCR signaling could be used to stratify responses to anti-PD-1 in patients with melanoma.

Contributed by Lauren Hitchings

Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in the tumor draining lymph nodes

Using an autochthonous lung adenocarcinoma model, Schenkel and Herbst et al. showed that as tumors progressed, the numbers and functionality of tumor antigen-specific TCF-1+CD8+ T cells declined in lungs, but not dLNs. Cycling SlamF6+ and non-cycling SlamF6- TCF-1+CD8+ TILs with shared clonotypes, but distinct transcriptional profiles were identified. The former, emigrants from dLNs, had higher inhibitory receptor expression than the latter, which dominated in late stage disease. In dLNs, tumor-specific dividing CD8+ T cells and cDC1 migrants decreased as tumors progressed; this was reversed by Flt3L+CD40 agonism, which reduced tumor burden.

Contributed by Paula Hochman

Antigen Dominance Hierarchies Shape TCF1+ Progenitor CD8 T Cell Phenotypes in Tumors

Burger et al. expressed pairs of neoantigens in KP mouse lung adenocarcinomas to demonstrate that during tumor development, concurrently expressed neoantigens compete and establish an antigen dominance hierarchy, resulting in suppressed T cell expansion, differentiation, and effector function against the subdominant antigen. Subdominant T cell responses were enriched for a dysfunctional subset of CCR6+TCF1+ progenitor cells with Tc17 differentiation trajectory, and associated with poor response to ICB in both humans and mice. Therapeutic vaccination eliminated CCR6+TCF1+ cells and improved the subdominant response.

Contributed by Shishir Pant

Systematic Discovery and Validation of T Cell Targets Directed Against Oncogenic KRAS Mutations

Approximately 19% of cancer diagnoses are associated with a RAS mutation, and in roughly 75% of these cases, a mutation in KRAS has been detected. Choi et al. generated a panel of nine stably transduced cell lines. Each line expressed all four of the most common KRAS codon 12 mutations along with one HLA-1 allele. The peptides, which were bound to each HLA-I allele, were purified, sequenced, and tested for the ability to stimulate primary in vitro T cell responses. This work has doubled the number of known immunogenic KRAS-related neoantigens and provided a road map for systemic discovery of neoantigens in many different types of tumors.

Contributed by Margot O’Toole

Resident memory CD8+ T cells in regional lymph nodes mediate immunity to metastatic melanoma

In a mouse model of melanoma-associated vitiligo (MAV), Molodtsov and Khatwani et al. found that injected gp100-specific pmel T cells inhabited various organs, where they expressed resident memory (Trm) markers, but with tissue-specific transcriptional profiles. In naive recipients parabiosed with MAV/pmel mice, circulating memory T cell phenotypes conferred protection against lung, but not LN melanoma rechallenge, suggesting that Trm cells were required to restrict LN metastasis. Trm cells were detected in patient melanoma-infiltrated LNs, and a LN-Trm signature indicated better prognosis in patients with regional LN metastasis.

Contributed by Alex Najibi

Anticancer immunity induced by a synthetic tumor-targeted CD137 agonist

As most agonist costimulatory receptor antibodies have been clinically disappointing, Upadhyaya, Lahdenranta, Hurov et al. generated a novel class of synthetic, low molecular weight, tumor-targeted immune cell agonists (TICAs) by chemically linking bicyclic peptides (Bicycles) isolated by phage display. CD137 and OX40 agonist Bicycles were linked in different ratios to common tumor Ag Bicycles (e.g., EphA2). Agonist activity of TICA CD137/EphA2 (BCY12491) was specific for EphA2+ tumor cells and eliminated MC38 tumors in vivo in a CD8+ T cell-dependent manner with memory. Although plasma t1/2 of BCY12491 was short (1-2h), intermittent dosing was effective.

Contributed by Katherine Turner

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.