Weekly Digests
‹ Back to March

Resurrecting IL-2

March 21, 2018

IL-2 is a critical T cell cytokine that promotes effector T cell expansion and survival and improves antitumor functions, but as an immunotherapy, its use has been very limited due to induction of suppressive T cell responses and potentially severe toxicities. In an effort to circumvent IL-2’s less desirable effects, Sultan et al. and Sockolosky et al. have independently come up with new modifications to IL-2 administration, published in Cancer Immunology Research and Science, respectively.

BiVax, a previously developed peptide antigen (Ag) vaccine + poly-IC adjuvant combination developed by Sultan et al., can generate significant Ag-specific T cell responses against defined tumor antigens, but requires co-treatment with anti-PD-1 for optimal antitumor response. Based on their observations that expansion of Ag-specific T cells can be limited by competition with irrelevant T cells, the team evaluated the addition of IL-2 in the form of IL-2/anti-IL-2 complexes (IL2Cx) after vaccination and achieved enhanced Ag-specific T cell expansion in vivo. Interestingly, they found that IL2Cx administration enhanced the vaccine-induced antitumor effect only when it was administered with the vaccine boost (at least 5 days after the vaccine prime), indicating that Ag-specific T cells may need time to become sufficiently activated and upregulate IL-2 receptor expression before they can be prompted to proliferate.

With the proper timing, the addition of IL2Cx to BiVax improved the antitumor efficacy of Ag-specific T cells against established subcutaneous B16 melanoma, delaying tumor growth and extending overall survival compared to controls. Expectedly, the researchers found that the antitumor efficacy was due in part to the increased number of Ag-specific T cells, but they also noted that the IL2Cx treatment reduced expression of PD-1 on the stimulated T cells. Surprisingly, by comparison to a similar vaccine formulation, they came to realize that neither cell number nor PD-1 expression fully explained the increased antitumor efficacy. Through a series of experiments, the researchers found that IL2Cx also enhanced the ability of T cells to resist PD-1-induced negative signaling via increased expression of pSTAT5; this allowed the cells to remain functional even in tumor microenvironments expressing high levels of PD-L1 and PD-L2.

Next, the researchers demonstrated that the antibody/cytokine complex enhanced vaccine efficacy by extending the in vivo half-life of available IL-2 in the serum rather than by antibody-mediated targeting to IL-2 receptors. In this vein, they went on to show that half-life-extended pegylated IL-2 (PEG-IL2)was similarly effective. Mice treated with Bi-Vax + IL2Cx or PEG-IL2 were not cured and eventually developed tumors despite persistence of Ag-specific T cells and continued tumor expression of the antigen, suggesting that extended IL2Cx dosing might be required, although other explanations are possible.

In a separate study focusing on how to use IL-2 to improve expansion of engineered T cells, Sockolosky et al. created IL-2 cytokine-receptor orthogonal pairs that interact preferentially with one another, but not with their natural cytokine and receptor counterparts. To develop the pairs, the researchers focused their efforts on the interaction between IL-2 and the IL-2Rβ chain; of the three IL-2R chains, IL-2Rβ is required for signal transduction and can bind to IL-2 independently. Based on the known structure of the IL-2/IL-2R complex, the researchers generated a double mutant orthoIL-2Rβ incapable of binding wild-type IL-2. They then used yeast display to evolve IL-2 variants capable of binding to orthoIL-2Rβ but not wild-type IL-2Rβ. This yielded two standout orthoIL-2 mutant variants, 1G12 and 3A10.

To test their system in vivo, the researchers engineered T cells to express the mutant orthoIL-2Rβ and adoptively transferred them into immunocompetent mice. Using the orthoIL-2, they were able to direct IL-2 signaling to preferentially expand adoptively transferred T cells, with the 3A10 variant being more selective but slightly less potent, and the 1G12 variant being the opposite. Fusing each of the orthoIL-2 variants to albumin extended in vivo half-life; this increased the activity of both variants, but only the orthoIL-2 3A10 variant retained strong target specificity. Ex vivo evaluation of orthoIL-2Rβ T cells that had been expanded in vivo with orthoIL-2 showed that they produced significantly more IFNγ than wild-type T cells expanded with IL-2. While PD-1 levels were comparable between these groups, TIM-3 exhaustion was significantly lower on orthoIL-2Rβ T cells. This suggested that there may be additional off-target effects of IL-2 not induced with orthoIL-2.

Finally, to test clinical relevance, Sockolosky et al. engineered T cells with a tumor-specific TCR and orthoIL-2Rβ and adoptively transferred them into mice with B16-F10 melanoma. Administration of orthoIL-2 3A10 (or 1G12 at a dose that had minimal off-target activity on wild-type IL-2R) produced a significant tumor growth delay and survival advantage, comparable to the advantage produced with standard high-dose IL-2. Overall, Sockolosky et al. conclude that this approach could be used to redirect the specificity of IL-2 toward engineered T cells, enabling the selective expansion of only desired subsets in adoptive cell therapy with reduced off-target activity and toxicity. This strategy works on other cells expressing the common IL-2 γ-chain (such as Tregs or B cells) and so could be broadly useful as both a research tool and in the clinical setting.

by Lauren Hitchings

References:

Sultan H., Kumai T., Fesenkova V.I., Fan A.E., Wu J., Cho H.I., Kobayashi H., Harabuchi Y., Celis E. Sustained persistence of IL2 signaling enhances the antitumor effect of peptide vaccines through T-cell expansion and preventing PD-1 inhibition. Cancer Immunol Res. 2018 Feb 26.

Sockolosky J.T., Trotta E., Parisi G., Picton L., Su L.L., Le A.C., Chhabra A., Silveria S.L., George B.M., King I.C., Tiffany M.R., Jude K., Sibener L.V., Baker D., Shizuru J.A., Ribas A., Bluestone J.A., Garcia K.C. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science. 2018 Mar 2.

In the Spotlight...

A facile approach to enhance antigen response for personalized cancer vaccination

Building on their previous porous silicon microrod vaccine, Li et al. enhanced the vaccine’s immune-stimulating and antigen-incorporating effects by adsorbing polyethyleneimine to the microrods. This enhanced the accumulation of activated dendritic cells at the vaccine site and draining lymph nodes, and increased the number and activation state of antigen-specific CD8+ T cells in the peripheral blood and the tumor. A single dose of the easily assembled vaccine resulted in significant tumor control in multiple viral and neoantigen vaccine tumor models and was synergistic with anti-CTLA-4.

The multi-receptor inhibitor axitinib reverses tumor-induced immunosuppression and potentiates treatment with immune-modulatory antibodies in preclinical murine models

Läubli et al. show that the tyrosine kinase inhibitor axitinib not only blocks VEGF receptors, but also reduces the immunosuppressive environment of MC38 and LLC1 tumors by decreasing immune-inhibitory mast and myeloid cell populations. Depletion of CD4+ and CD8+ T cell populations prior to axitinib administration demonstrated that the axitinib effects are mediated via T cells. Axitinib reduced intratumoral PD1+/TIM-3+ exhausted CD8+ T cells, and synergized with checkpoint inhibitors (anti-PD-1 and/or anti-TIM-3) or a CD137 agonist.

A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy

Using multiple forms of anti-CTLA-4 antibody, Du et al. discovered that the antitumor mechanism of anti-CTLA-4 surprisingly requires none of the following: blockade of the CTLA-4/B7 interaction, inhibition of B7 trans-endocytosis, upregulation of B7 on dendritic cells, de novo CD8+ T cell priming, or CD4+ T cell activation. Instead, the antitumor effect of anti-CTLA-4 is dependent on the local depletion of Tregs via interactions with the Fc receptor on other host cells and the subsequent antibody-dependent cellular cytotoxicity.

Uncoupling therapeutic from immunotherapy-related adverse effects for safer and effective anti-CTLA-4 antibodies in CTLA4 humanized mice

Du et al. developed human CTLA-4 knock-in homo- and heterozygous mouse models and demonstrated that the immunotherapy-related adverse events (irAEs) could be uncoupled from antitumor immunity. The model recapitulated most irAEs observed with ipilimumab, including severe organ inflammation and anemia. irAEs were related to systemic T cell activation and increased autoreactive Teff/Treg ratio, while the antitumor effect depended on Treg depletion in the tumor. The CTLA-4h/h mouse model could prove useful for identifying safer anti-CTLA-4 therapies.

Molecular signatures of circulating melanoma cells for monitoring early response to immune checkpoint therapy

Hong et al. developed a non-invasive, blood-based method to quantify early response of melanoma patients to immune checkpoint blockade. Blood samples were microfluidically enriched for circulating tumor cells (CTCs) by efficient depletion of normal hematopoietic cells, and digital droplet-based PCR was used to detect and quantify a 19-gene melanoma RNA signature (CTC score). A decrease in CTC score within 7 weeks of treatment correlated with improved clinical outcomes, allowing for monitoring of early treatment-based changes.

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.