Abstract
IL-2 is a pleiotropic cytokine. In this issue of the JCI, Ren et al. report on the development of a low-affinity IL-2 paired with anti–PD-1 (PD-1–laIL-2) that reactivates intratumoral CD8+ T cells, but not CD4+ Treg cells. PD-1–laIL-2 treatment synergized with anti–PD-L1 therapy to overcome tumor resistance to immune checkpoint blockade (ICB) in tumor-bearing mice. Rejection of rechallenged tumors following PD-1–laIL-2 therapy demonstrated the establishment of a potent T cell memory response. Furthermore, PD-1–laIL-2 therapy manifested no obvious toxicity. These findings suggest the potential of PD-1–laIL-2 therapy in treating patients with cancer.
Targeting IL-2 signaling pathway for cancer therapy
IL-2 is produced primarily by activated CD4+ T cells and acts in a paracrine or autocrine fashion (1, 2). IL-2 receptor (IL-2R) signaling occurs through three subunits: alpha (CD25), beta (CD122), and gamma (CD132) (3). Intermediate-affinity dimeric IL-2 receptor consists of IL-2Rβ and IL-2Rγ on naive CD4+ and CD8+ T cells, memory T cells, and natural killer (NK) cells. TCR engagement or IL-2 stimulation induces the expression of IL-2Rα to form high-affinity trimeric IL-2 receptors that are highly expressed on Treg cells and recently activated effector T cells (4). IL-2 signaling has been an attractive immunotherapeutic target since IL-2 mediates effector T cell activation, including effector CD8+ T cells, which are vital for antitumor immunity. High-dose IL-2 was approved by the FDA in 1992 for treatment of certain types of cancer (5). However, IL-2 possesses a very short half-life and requires high doses to be effective, leading to toxicity and severe side effects, such as inflammation and vascular leak syndrome (6). Alternatively, low doses of IL-2 preferentially target IL-2Rα on Treg cells, restricting the immune response, and are associated with poor prognosis in patients with cancer (7, 8). Therefore, methods to target certain T cell subsets while reducing Treg cell binding have been a recent focus in the field of IL-2 therapy.
Manipulation of T cell phenotype by IL-2 therapy
To effectively manipulate effector T cells and reduce side effects of high-dose IL-2, IL-2 variants have been developed to stimulate specific T cell subsets through selective targeting of certain IL-2R chains. One strategy has been to introduce mutations in IL-2 to create mutants with preferential IL-2R chain binding. Mutants with reduced IL-2Rβ binding have been shown to target high-affinity IL-2 receptor expressed on effector T cells (Figure 1). These mutants have also exhibited reduced toxicity, possibly due to decreased binding of intermediate-affinity receptors on NK cells that lack IL-2Rα (1, 9). STK-012, a partial IL-2 agonist produced by Synthekine, employs a similar strategy by selectively binding IL-2Rα and IL-2Rβ subunits, but not IL-2Rγ. Effector T cells that may be specific for tumor epitopes can thus expand and readily attack the tumor while avoiding NK cell stimulation (10). However, undesirable Treg cell expansion remains a concern due to high IL-2Rα expression on Treg cells (7, 8). To address this issue, IL-2 mutants with reduced binding to IL-2Rα have also been generated. The cytokine company Nektar has engineered an IL-2 mutant with a bias toward IL-2Rβ and IL-2Rγ, rather than IL-2Rα, to reduce Treg cell binding (10). H9, an IL-2 superkine (sumIL-2) with enhanced IL-2Rβ binding without the need for IL-2Rα, was shown to increase expansion of cytotoxic memory T cells and NK cells while decreasing that of Treg cells (11). Interestingly, H9T, an engineered H9-based partial agonist with further reduced binding to IL-2Rγ, was also recently shown to promote CD8+ T cell proliferation that maintained a stem-like memory state and mediated greater antitumor immunity (12).
To enhance the activity of IL-2 in vivo and limit toxicity by reducing the necessary dose, IL-2 therapy has been combined with anti–IL-2 monoclonal antibodies (mAb). Interestingly, various anti–IL-2 mAbs differentially stimulate different immune cell subsets. Anti–mouse IL-2 mAbs S4B6 and JES6-5, as well as anti–human IL-2 mAb MAB602, complexed with recombinant IL-2, selectively stimulate memory CD8+ cells and NK cells in vivo to improve IL-2 cancer therapy (Figure 1) (13). On the other hand, anti–IL-2 mAb JES6-1 inhibits proliferation of CD8+ cells and NK cells yet maintains its ability to activate Treg cells and has been implicated as a potential treatment for autoimmune disease (14). Binding of these various mAbs to certain regions of IL-2, therefore blocking IL-2 binding to specific IL-2R chains, may explain these contrasting cell type affinities (1, 2).
IL-2–based fusion proteins are another IL-2 therapy strategy with a multitude of current preclinical and clinical trials (15, 16). Fusion of IL-2 to a fragment crystallizable (Fc) region has proven to be beneficial due to increased half-life, complement activation, and induction of antibody-dependent cellular cytotoxicity (ADCC) toward Treg cells (17–19). Furthermore, fusion of IL-2 to antigen-specific antibodies (termed an immunocytokine) allows for targeted delivery of IL-2 to cells and tissues expressing a protein of interest. Numerous IL-2 immunocytokines have been developed to target tumor-associated antigens expressed by cancer cells and their surrounding tissue (16). IL-2 is therefore honed to tumor tissues to enact its function. However, this strategy still lacks the ability to specifically target effector T cells within the tumor that are pertinent to anticancer immunity.
Targeting intratumoral effector T cells with IL-2 and anti–PD-1 therapy
Ren et al. (20) addressed this intratumoral T cell targeting gap by engineering an immunocytokine fusion protein consisting of low-affinity IL-2 (laIL-2) linked to an anti–PD-1 antibody (PD-1–laIL-2). laIL-2 exhibits reduced binding to IL-2Rα and IL-2Rβ to diminish unfavorable Treg cell binding in the tumor and periphery. Meanwhile, PD-1 is highly expressed on tumor-infiltrating CD8+ T cells. As a result, PD-1–laIL-2 possessed elevated avidity toward intratumoral CD8+ T cells, rather than Treg cells or peripheral CD4+ and CD8+ T cells. This specificity not only reduced the systemic toxicity, but also enhanced tumor control in A20 and MC38 tumor models, as well as A375 tumor-bearing humanized mice. In addition, PD-1–laIL-2 in combination with anti–PD-L1 therapy overcame tumor resistance to PD-L1 blockade therapy. Notably, this effect was dependent on intratumoral CD8+ T cells, whose proliferation was selectively induced by PD-1–laIL-2. Further investigation revealed that PD-1–laIL-2 seemed to selectively target intratumoral PD-1+TIM-3+CD8+ T cells, which are usually described as a functionally exhausted and/or terminally differentiated T cell subset. Therefore, PD-1–laIL-2 could reactivate PD-1+TIM-3+CD8+ T cells to enhance antitumor activity (Figure 1). Tumor rechallenge resulted in spontaneous rejection in tumor-bearing mice previously treated with PD-1–laIL-2. This effect was also dependent on the presence of CD8+ T cells, indicating these rejuvenated T cells are tumor antigen-specific and can mediate a strong memory response. These promising results suggest that PD-1–laIL-2 therapy may bring clinical benefits to patients with cancer.
Version 1. 02/01/2022
Electronic publication
Footnotes
Conflict of interest: WZ serves as a scientific consultant or board member at NGM Biopharmaceuticals Inc., Roivant, NextCure, CStone, CrownBio, and Intergalactic Therapeutics.
Copyright: © 2022, Holcomb et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.
Reference information: J Clin Invest. 2022;132(3):e156628. https://doi.org/10.1172/JCI156628.
See the related article at Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity.
Contributor Information
Erin A. Holcomb, Email: eholc@umich.edu.
Weiping Zou, Email: wzou@med.umich.edu.
References
- 1.Boyman O, et al. Potential use of IL-2/anti-IL-2 antibody immune complexes for the treatment of cancer and autoimmune disease. Expert Opin Biol Ther. 2006;6(12):1323–1331. doi: 10.1517/14712598.6.12.1323. [DOI] [PubMed] [Google Scholar]
- 2.Létourneau S, et al. IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25. Proc Natl Acad Sci U S A. 2010;107(5):2171–2176. doi: 10.1073/pnas.0909384107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wang X, et al. Structure of the quaternary complex of interleukin-2 with its alpha, beta, and gammac receptors. Science. 2005;310(5751):1159–1163. doi: 10.1126/science.1117893. [DOI] [PubMed] [Google Scholar]
- 4.Létourneau S, et al. IL-2- and CD25-dependent immunoregulatory mechanisms in the homeostasis of T-cell subsets. J Allergy Clin Immunol. 2009;123(4):758–762. doi: 10.1016/j.jaci.2009.02.011. [DOI] [PubMed] [Google Scholar]
- 5.Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol. 2014;192(12):5451–5458. doi: 10.4049/jimmunol.1490019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pachella LA, et al. The toxicity and benefit of various dosing strategies for interleukin-2 in metastatic melanoma and renal cell carcinoma. J Adv Pract Oncol. 2015;6(3):212–221. [PMC free article] [PubMed] [Google Scholar]
- 7.Wei S, et al. Interleukin-2 administration alters the CD4+FOXP3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res. 2007;67(15):7487–7494. doi: 10.1158/0008-5472.CAN-07-0565. [DOI] [PubMed] [Google Scholar]
- 8.Kryczek I, et al. Cutting edge: Th17 and regulatory T cell dynamics and the regulation by IL-2 in the tumor microenvironment. J Immunol. 2007;178(11):6730–6733. doi: 10.4049/jimmunol.178.11.6730. [DOI] [PubMed] [Google Scholar]
- 9.Shanafelt AB, et al. A T-cell-selective interleukin 2 mutein exhibits potent antitumor activity and is well tolerated in vivo. Nat Biotechnol. 2000;18(11):1197–1202. doi: 10.1038/81199. [DOI] [PubMed] [Google Scholar]
- 10.Garber K. Synthekine: beyond cytokines. Nat Biotechnol. doi: 10.1038/d41587.021.00006-6. [published online June 9, 2021]. [DOI] [PubMed] [Google Scholar]
- 11.Levin AM, et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature. 2012;484(7395):529–533. doi: 10.1038/nature10975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mo F, et al. An engineered IL-2 partial agonist promotes CD8+ T cell stemness. Nature. 2021;597(7877):544–548. doi: 10.1038/s41586-021-03861-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Boyman O, et al. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science. 2006;311(5769):1924–1927. doi: 10.1126/science.1122927. [DOI] [PubMed] [Google Scholar]
- 14.Webster KE, et al. In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J Exp Med. 2009;206(4):751–760. doi: 10.1084/jem.20082824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mullard A. Restoring IL-2 to its cancer immunotherapy glory. Nat Rev Drug Discov. 2021;20(3):163–165. doi: 10.1038/d41573-021-00034-6. [DOI] [PubMed] [Google Scholar]
- 16.MacDonald A, et al. Interleukin 2-based fusion proteins for the treatment of cancer. J Immunol Res. 2021;2021:7855808. doi: 10.1155/2021/7855808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hsu EJ, et al. A cytokine receptor-masked IL2 prodrug selectively activates tumor-infiltrating lymphocytes for potent antitumor therapy. Nat Commun. 2021;12(1):2768. doi: 10.1038/s41467-021-22980-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhu EF, et al. Synergistic innate and adaptive immune response to combination immunotherapy with anti-tumor antigen antibodies and extended serum half-life IL-2. Cancer Cell. 2015;27(4):489–501. doi: 10.1016/j.ccell.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Vazquez-Lombardi R, et al. Potent antitumour activity of interleukin-2-Fc fusion proteins requires Fc-mediated depletion of regulatory T-cells. Nat Commun. 2017;8:15373. doi: 10.1038/ncomms15373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ren Z, et al. Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity. J Clin Invest. 2021;132(3):e153604. doi: 10.1172/JCI153604. [DOI] [PMC free article] [PubMed] [Google Scholar]