Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer

Alexey Revenko, Larissa S Carnevalli, Charles Sinclair, Ben Johnson, Alison Peter, Molly Taylor, Lisa Hettrick, Melissa Chapman, Stephanie Klein, Anisha Solanki, Danielle Gattis, Andrew Watt, Adina M Hughes, Lukasz Magiera, Gozde Kar, Lucy Ireland, Deanna A Mele, Vasu Sah, Maneesh Singh, Josephine Walton, Maelle Mairesse, Matthew King, Mark Edbrooke, Paul Lyne, Simon T Barry, Stephen Fawell, Frederick W Goldberg, A Robert MacLeod, Alexey Revenko, Larissa S Carnevalli, Charles Sinclair, Ben Johnson, Alison Peter, Molly Taylor, Lisa Hettrick, Melissa Chapman, Stephanie Klein, Anisha Solanki, Danielle Gattis, Andrew Watt, Adina M Hughes, Lukasz Magiera, Gozde Kar, Lucy Ireland, Deanna A Mele, Vasu Sah, Maneesh Singh, Josephine Walton, Maelle Mairesse, Matthew King, Mark Edbrooke, Paul Lyne, Simon T Barry, Stephen Fawell, Frederick W Goldberg, A Robert MacLeod

Abstract

Background: The Regulatory T cell (Treg) lineage is defined by the transcription factor FOXP3, which controls immune-suppressive gene expression profiles. Tregs are often recruited in high frequencies to the tumor microenvironment where they can suppress antitumor immunity. We hypothesized that pharmacological inhibition of FOXP3 by systemically delivered, unformulated constrained ethyl-modified antisense oligonucleotides could modulate the activity of Tregs and augment antitumor immunity providing therapeutic benefit in cancer models and potentially in man.

Methods: We have identified murine Foxp3 antisense oligonucleotides (ASOs) and clinical candidate human FOXP3 ASO AZD8701. Pharmacology and biological effects of FOXP3 inhibitors on Treg function and antitumor immunity were tested in cultured Tregs and mouse syngeneic tumor models. Experiments were controlled by vehicle and non-targeting control ASO groups as well as by use of multiple independent FOXP3 ASOs. Statistical significance of biological effects was evaluated by one or two-way analysis of variance with multiple comparisons.

Results: AZD8701 demonstrated a dose-dependent knockdown of FOXP3 in primary Tregs, reduction of suppressive function and efficient target downregulation in humanized mice at clinically relevant doses. Surrogate murine FOXP3 ASO, which efficiently downregulated Foxp3 messenger RNA and protein levels in primary Tregs, reduced Treg suppressive function in immune suppression assays in vitro. FOXP3 ASO promoted more than 70% reduction in FOXP3 levels in Tregs in vitro and in vivo, strongly modulated Treg effector molecules (eg, ICOS, CTLA-4, CD25 and 4-1BB), and augmented CD8+ T cell activation and produced antitumor activity in syngeneic tumor models. The combination of FOXP3 ASOs with immune checkpoint blockade further enhanced antitumor efficacy.

Conclusions: Antisense inhibitors of FOXP3 offer a promising novel cancer immunotherapy approach. AZD8701 is being developed clinically as a first-in-class FOXP3 inhibitor for the treatment of cancer currently in Ph1a/b clinical trial (NCT04504669).

Keywords: Immunity, Cellular; Immunotherapy; Lymphocytes, Tumor-Infiltrating; Therapies, Investigational; Tumor Microenvironment.

Conflict of interest statement

Competing interests: The authors are paid employees of AstraZeneca or Ionis Pharmaceuticals, as indicated by their affiliations.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
AZD8701 is a highly potent clinical candidate ASO targeting human FOXP3. (A–C) Primary human Tregs were isolated from human PBMCs and cultured with AZD8701 or control ASO in duplicates for a total of 9 days, in the presence of Dynabeads human T-activated CD3/CD28 for the final 2 days of culture. (A) Line graph and (B) histogram show FOXP3 protein abundance in cultured Tregs as measured by flow cytometry. Histogram shows representative data for Tregs cultured with 1 µM ASO. (C) Histograms show the abundance of indicated proteins from a representative treatment with 1 µM AZD8701. (D–E) Contour plot and quantification of FOXP3 knockdown in human PBMC nTRegs and eTRegs with AZD8701 by flow cytometry. Data shown for three healthy donors. (F–G) iTregs were differentiated and cultured in the presence of ASOs in quadruplicates. Data represent ≥3 independent experiments and a total of ≥6 independent donors. (F) Line graph shows ability of iTregs to inhibit proliferation of effector cells in an in vitro suppression assay. (G) Histograms show representative CD25 or CellTrace Violet (CTV) staining on effector cells cultured at a 1:2 iTreg:Teffector ratio. (H) NSG mice were humanized by the infusion of human PBMC and treated systemically for four consecutive days with different AZD8701 doses. (I) FOXP3 messenger RNA expression was quantified by RT-qPCR. N=4 per group. (J) FOXP3 protein levels were quantified by flow cytometry in spleen, blood and bone marrow of humanized mice treated as in (F). N=7 per group. Data in figure is representative of ≥2 independent experiments. Error bars are ±SEM *, p≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance (ANOVA) with Dunnett’s post-test for E and J and two-way ANOVA with Dunnett’s post-test for F. Differences are calculated relative to control ASO (E, F and J) or saline (I). ASOs, antisense oligonucleotides; eTreg, effector Tregs; iTreg, inducible Tregs; nTreg, natural Tregs; PBS, phosphate buffered saline; Treg, regulatory T cells.
Figure 2
Figure 2
Antisense-mediated knockdown of mouse FOXP3 promotes loss of Treg suppressive phenotypic markers and function. (A) Primary Tregs were isolated from mouse spleen and cultured with ASOs (5 µM) in triplicates for 7 days. Bar chart shows relative FOXP3 mRNA expression measured by RT-qPCR, histogram shows FOXP3 protein expression measured by flow cytometry. (B) Inducible Tregs (iTregs) were differentiated and cultured in the presence of ASO for a total of 7 days. Heatmap shows expression of FOXP3-dependent mRNA genes as measured by Fluidigm. (C) Histograms show abundance of indicated proteins in primary Tregs. (D) iTregs were differentiated and cultured in the presence of ASOs and evaluated for their ability to inhibit proliferation of effector cells in an in vitro suppression assay in duplicates. Histograms show representative CellTrace Violet (CTV) staining on effector cells cultured at a 1:1 Treg:Teffector ratio. Data in figure represent ≥2 independent experiments. *, P≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance (ANOVA) with Dunnett’s post-test for (A) and two-way ANOVA with Dunnett’s post-test for (D). Differences are calculated relative to control ASO. ASOs, antisense oligonucleotides; mRNA, messenger RNA; Tregs, regulatory T cells.
Figure 3
Figure 3
Mouse FOXP3 ASOs promote FOXP3 knockdown in vivo, which associates with phenotypes indicative of immunopotentiation. (A) Mice were systemically treated with FOXP3 ASOs for 3 weeks at 100 mg/kg/week dose. CD4 T cells were isolated from spleens and FOXP3 messenger RNA was measured by RT-qPCR. ASOs selected for further in vivo evaluation are highlighted in red and blue. N=4 per group. (B) iTregs were differentiated in the presence of ASO in duplicates for 5 days, and FOXP3 protein abundance was measured by flow cytometry. (C) Primary splenic nTregs were cultured in the presence of indicated ASOs in duplicates for 7 days, and FOXP3 protein abundance was measured by flow cytometry. (D–G) 20 mg/kg and 50 mg/kg FOXP3 ASOs or 50 mg/kg control ASO were administered to BALB/c mice via subcutaneously route 5 days on 2 days off. N=5 per group. (D) The line graph shows the frequency of gated FOXP3+ cells within the splenic CD4+ population. Pseudocolor density plots show representative FOXP3+ gating. (E) Line graph shows programmed cell death 1 protein expression on detectable FOXP3+ cells. (F) Line graphs show the frequency of CD62LloCD44hi T-memory cells within the total CD4+ T-cell population. (G) Scatter bar charts show the frequency of CD69+ cells within the CD4+ or CD8+ populations at the week six time point. (H) ASOs were dosed (100 mg/kg two times per week) for 3 weeks before spleens from cohorts of mice were analyzed by flow cytometry at indicated time points following cessation of treatment. N=5 per group. Data represent two independent experiments. Error bars are SEM data in figure represent ≥2 independent experiments. *, P≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance (ANOVA) with Dunnett’s post-test for (A and G) and two-way ANOVA with Dunnett’s post-test for (D, E, F and H). Differences are calculated relative to control ASO (D–H) or saline (A). ASOs, antisense oligonucleotides; iTreg, inducible Tregs; nTreg, natural Tregs; Tregs, regulatory T cells.
Figure 5
Figure 5
FOXP3 ASOs promote antitumor efficacy when combined with αPD-L1 immune checkpoint blockade. (A) Mice were treated with mouse FOXP3 ASO (895317) (50 mg/kg BIW) and αPD-L1 (10 mg/kg BIW) alone or in combination from day 1 post A20 tumor-implantation and dosed two times per week for the duration of the experiment. N=14 per group. (B) Mean tumor and (C) individual tumor volumes and indicated number of complete responses (CR). (D-E) Tumor samples and peripheral blood (PB) were analyzed at day 20 by flow cytometry and RNA levels, respectively. (D) Frequency of CD8 +T cells and dendritic cells in tumor and (E) cytotoxic T cell and antigen-presenting cell gene markers in PB. *, P≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance (ANOVA) with Dunnett’s post-test for (D and E) and two-way ANOVA with Dunnett’s post-test for (B). Differences are calculated relative to PBS. ASOs, antisense oligonucleotides; BIW, two times per week; PBS, phosphate buffered saline; PD-1, programmed cell death 1; PD-L1, programmed cell death ligand 1.
Figure 4
Figure 4
Single-agent antitumor efficacy mediated by mouse FOXP3 ASOs. (A–F) ID8-VEGF or A20 tumor-bearing mice were systemically treated with FOXP3 ASOs starting at day 1 after implant at 50 mg/kg BIW until day 62 (ID8-VEGF) or four times per week until day 24 (A20). N=8 per group. (A) Tumors were dissociated and tumor-infiltrating Tregs were analyzed by flow cytometry. Helios +CD4 Tregs are shown in red. (B) FOXP3 protein expression was measured in Helios +CD4 Tregs. Line graphs show (C) ID8-VEGF and (E) A20 tumor volumes. The number of complete responses (CR) vs total number of animals in the group is indicated next to lines. (D) ID8-VEGF tumors were dissociated, and abundance of total tumor-infiltrating leukocytes (CD45 +cells) and CTL (CD8 +T cells) were analyzed by flow cytometry. (F) Total RNA from A20 tumors was analyzed for the mRNA expression of CTL marker CD8 and immune cell activation marker GzmB. (G) A20 tumor-bearing mice were treated with indicated ASO or vehicle control 50 mg/kg BIW and tumors analyzed by flow cytometry at day 13 or day 20 time points. N=10 per group. Bar charts show tumor or spleen Foxp3 and GzmB expression measured by RT-qPCR. (H) CD8+ T cells were depleted in vivo with an αCD8 blocking antibody in A20 tumor-bearing mice that were treated with indicated ASO or control at 50 mg/kg BIW. N=12 per group. Data represent >4 independent experiments in (A–F), two independent experiments in (G) and a single cell depletion experiments in (H). *, P≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.000 by one-way analysis of variance (ANOVA) with Dunnett’s post-test for (B, D, F and G) and two-way ANOVA with Dunnett’s post-test for (C, E and H). Differences are calculated relative to PBS (B–G) or as indicated on the panel (H). ASOs, antisense oligonucleotides; BIW, two times per week; mRNA, messenger RNA; PBS, phosphate buffered saline; VEGF, vascular endothelial growth factor.
Figure 6
Figure 6
FOXP3 ASOs reprogram Treg effector cell phenotype in vivo in combination with αPD-L1 immune checkpoint blockade. (A) Mice were treated with ASO (50 mg/kg BIW) and αPD-L1 (10 mg/kg BIW) alone or in combination from day 1 post A20 tumor-implantation. N=14 per group. Tumors were analyzed by mass cytometry at day 20 after implantation. (B) t-SNE analysis of tumor-infiltrating CD4+ T cells. (C) Relative changes of CD4 +T cell clusters compared with control group. (D) Expression map of T cells and Tregs markers. (E) Quantification of Treg activation expression markers. Error bars are SD *, p≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance with Dunnett’s post-test relative to control ASO. ASOs, antisense oligonucleotides; BIW, two times per week; t-SNE, t-distributed stochastic neighbor embedding; PD-L1, programmed cell death ligand 1; Treg, Regulatory T cells.
Figure 7
Figure 7
FOXP3 ASOs promote additive efficacy when combined with αPD-1 immune checkpoint blockade. Mice were treated with ASO 895310 (50 mg/kg BIW) and αPD-1 (10 mg/kg BIW) alone or in combination from day 1 post A20 tumor-implantation. ASO was dosed for the duration of the study. αPD-1 was dosed six times. N=12 per group. Mice were sacrificed when tumors exceeded 1500 mm3 or after 40 days. (A) Spider plots indicate tumor volumes. Survival panel shows percent surviving mice vs time. (B–G) Foxp3, CD8a/CD8b and Granzyme messenger RNA expression in tumors and spleens from terminal samples. (H) Model of ASO targeting of FOXP3 expression to reduce Treg immunosuppressive capacity and promote antitumor immunity. Data in figure represent two independent experiments. CR, complete responses. Error bars are ±SEM *, p≤0.05; **, p≤0.01; ***, p≤0.001; ****, p≤0.0001 by one-way analysis of variance with Dunnett’s post-test relative to PBS group. Survival analysis (A) was done by log-rank Mantel-Cox test. ASOs, antisense oligonucleotides; BIW, two times per week; IFN, interferon; PBS, phosphate buffered saline; PD-1, programmed cell death 1.

References

    1. Sakaguchi S, Yamaguchi T, Nomura T, et al. . Regulatory T cells and immune tolerance. Cell 2008;133:775–87. 10.1016/j.cell.2008.05.009
    1. Josefowicz SZ, Lu L-F, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012;30:531–64. 10.1146/annurev.immunol.25.022106.141623
    1. Whiteside TL. What are regulatory T cells (Treg) regulating in cancer and why? Semin Cancer Biol 2012;22:327–34. 10.1016/j.semcancer.2012.03.004
    1. Shang B, Liu Y, Jiang S-juan, et al. . Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep 2015;5:15179. 10.1038/srep15179
    1. Salama P, Phillips M, Grieu F, et al. . Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 2009;27:186–92. 10.1200/JCO.2008.18.7229
    1. Preston CC, Maurer MJ, Oberg AL, et al. . The ratios of CD8+ T cells to CD4+CD25+ FOXP3+ and FOXP3- T cells correlate with poor clinical outcome in human serous ovarian cancer. PLoS One 2013;8:e80063. 10.1371/journal.pone.0080063
    1. Plitas G, Konopacki C, Wu K, et al. . Regulatory T cells exhibit distinct features in human breast cancer. Immunity 2016;45:1122–34. 10.1016/j.immuni.2016.10.032
    1. Beyer M, Schultze JL. Regulatory T cells in cancer. Blood 2006;108:804–11. 10.1182/blood-2006-02-002774
    1. Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci 2018;1417:104–15. 10.1111/nyas.13625
    1. Owen DL, Mahmud SA, Sjaastad LE, et al. . Thymic regulatory T cells arise via two distinct developmental programs. Nat Immunol 2019;20:195–205. 10.1038/s41590-018-0289-6
    1. Sharma A, Subudhi SK, Blando J, et al. . Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3+ Regulatory T Cells (Tregs) in Human Cancers-Response. Clin Cancer Res 2019;25:3469–70. 10.1158/1078-0432.CCR-19-0402
    1. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003;299:1057–61. 10.1126/science.1079490
    1. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003;4:330–6. 10.1038/ni904
    1. Bennett CL, Christie J, Ramsdell F, et al. . The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001;27:20–1. 10.1038/83713
    1. Klages K, Mayer CT, Lahl K, et al. . Selective depletion of Foxp3+ regulatory T cells improves effective therapeutic vaccination against established melanoma. Cancer Res 2010;70:7788–99. 10.1158/0008-5472.CAN-10-1736
    1. Bos PD, Plitas G, Rudra D, et al. . Transient regulatory T cell ablation deters oncogene-driven breast cancer and enhances radiotherapy. J Exp Med 2013;210:2435–66. 10.1084/jem.20130762
    1. Crooke ST, Witztum JL, Bennett CF, et al. . RNA-Targeted therapeutics. Cell Metab 2018;27:714–39. 10.1016/j.cmet.2018.03.004
    1. Bennett CF, Baker BF, Pham N, et al. . Pharmacology of antisense drugs. Annu Rev Pharmacol Toxicol 2017;57:81–105. 10.1146/annurev-pharmtox-010716-104846
    1. Crooke ST. Molecular mechanisms of antisense oligonucleotides. Nucleic Acid Ther 2017;27:70–7. 10.1089/nat.2016.0656
    1. Seth PP, Siwkowski A, Allerson CR, et al. . Short antisense oligonucleotides with novel 2'-4' conformationaly restricted nucleoside analogues show improved potency without increased toxicity in animals. J Med Chem 2009;52:10–13. 10.1021/jm801294h
    1. Murray S, Ittig D, Koller E, et al. . TricycloDNA-modified oligo-2'-deoxyribonucleotides reduce scavenger receptor B1 mRNA in hepatic and extra-hepatic tissues--a comparative study of oligonucleotide length, design and chemistry. Nucleic Acids Res 2012;40:6135–43. 10.1093/nar/gks273
    1. Hong D, Kurzrock R, Kim Y, et al. . AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med 2015;7:ra185. 10.1126/scitranslmed.aac5272
    1. Reilley MJ, McCoon P, Cook C, et al. . STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. J Immunother Cancer 2018;6:119. 10.1186/s40425-018-0436-5
    1. Chowdhury S, Burris HA, Patel M, et al. . A phase I dose escalation, safety and pharmacokinetic (PK) study of AZD5312 (IONIS-ARRx), a first-in-class generation 2.5 antisense oligonucleotide targeting the androgen receptor (AR). Eur J Cancer 2016;69:S145. 10.1016/S0959-8049(16)33031-3
    1. Allan SE, Crome SQ, Crellin NK, et al. . Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol 2007;19:345–54. 10.1093/intimm/dxm014
    1. Wang J, Ioan-Facsinay A, van der Voort EIH, et al. . Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol 2007;37:129–38. 10.1002/eji.200636435
    1. Lozano T, Conde E, Martín-Otal C, et al. . TCR-induced FOXP3 expression by CD8+ T cells impairs their anti-tumor activity. Cancer Lett 2022;528:45–58. 10.1016/j.canlet.2021.12.030
    1. Curotto de Lafaille MA, Lafaille JJ, Lafaille Cde. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 2009;30:626–35. 10.1016/j.immuni.2009.05.002
    1. Webster KE, Walters S, Kohler RE, 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;206:751–60. 10.1084/jem.20082824
    1. Marson A, Kretschmer K, Frampton GM, et al. . Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature 2007;445:931–5. 10.1038/nature05478
    1. Zheng L, Sharma R, Gaskin F, et al. . A novel role of IL-2 in organ-specific autoimmune inflammation beyond regulatory T cell checkpoint: both IL-2 knockout and Fas mutation prolong lifespan of Scurfy mice but by different mechanisms. J Immunol 2007;179:8035–41. 10.4049/jimmunol.179.12.8035
    1. Gavin MA, Rasmussen JP, Fontenot JD, et al. . Foxp3-dependent programme of regulatory T-cell differentiation. Nature 2007;445:771–5. 10.1038/nature05543
    1. Kwon H-K, Chen H-M, Mathis D, et al. . FoxP3 scanning mutagenesis reveals functional variegation and mild mutations with atypical autoimmune phenotypes. Proc Natl Acad Sci U S A 2018;115:E253–62. 10.1073/pnas.1718599115
    1. Williams LM, Rudensky AY. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol 2007;8:277–84. 10.1038/ni1437
    1. Mayer CT, Ghorbani P, Kühl AA, et al. . Few Foxp3⁺ regulatory T cells are sufficient to protect adult mice from lethal autoimmunity. Eur J Immunol 2014;44:2990–3002. 10.1002/eji.201344315
    1. Barzaghi F, Passerini L, Gambineri E, et al. . Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun 2012;38:49–58. 10.1016/j.jaut.2011.12.009
    1. Elkord E, Al-Ramadi BK. Helios expression in FoxP3(+) T regulatory cells. Expert Opin Biol Ther 2012;12:1423–5. 10.1517/14712598.2012.711310
    1. Mosely SIS, Prime JE, Sainson RCA, et al. . Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res 2017;5:29–41. 10.1158/2326-6066.CIR-16-0114
    1. Taylor MA, Hughes AM, Walton J, et al. . Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discovery. J Immunother Cancer 2019;7:328. 10.1186/s40425-019-0794-7
    1. Dodagatta-Marri E, Meyer DS, Reeves MQ, et al. . α-PD-1 therapy elevates Treg/Th balance and increases tumor cell pSmad3 that are both targeted by α-TGFβ antibody to promote durable rejection and immunity in squamous cell carcinomas. J Immunother Cancer 2019;7:62. 10.1186/s40425-018-0493-9
    1. Kumagai S, Togashi Y, Kamada T, et al. . The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol 2020;21:1346–58. 10.1038/s41590-020-0769-3
    1. Finan C, Gaulton A, Kruger FA, et al. . The druggable genome and support for target identification and validation in drug development. Sci Transl Med 2017;9. 10.1126/scitranslmed.aag1166. [Epub ahead of print: 29 03 2017].
    1. Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol 2019;16:356–71. 10.1038/s41571-019-0175-7
    1. Arce Vargas F, Furness AJS, Solomon I, et al. . Fc-Optimized Anti-CD25 depletes tumor-infiltrating regulatory T cells and synergizes with PD-1 blockade to eradicate established tumors. Immunity 2017;46:577–86. 10.1016/j.immuni.2017.03.013
    1. Selby MJ, Engelhardt JJ, Quigley M, et al. . Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res 2013;1:32–42. 10.1158/2326-6066.CIR-13-0013
    1. Quezada SA, Peggs KS. Lost in translation: deciphering the mechanism of action of anti-human CTLA-4. Clin Cancer Res 2019;25:1130–2. 10.1158/1078-0432.CCR-18-2509
    1. Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol 2008;8:34–47. 10.1038/nri2206
    1. Di Giacomo AM, Biagioli M, Maio M. The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. Semin Oncol 2010;37:499–507. 10.1053/j.seminoncol.2010.09.007
    1. Klebanoff CA, Gattinoni L. Stubborn Tregs limit T-cell therapy. Blood 2012;120:2352–4. 10.1182/blood-2012-08-445239
    1. Zappasodi R, Sirard C, Li Y, et al. . Rational design of anti-GITR-based combination immunotherapy. Nat Med 2019;25:759–66. 10.1038/s41591-019-0420-8
    1. Maj T, Wang W, Crespo J, et al. . Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol 2017;18:1332–41. 10.1038/ni.3868
    1. Crooke ST, Wang S, Vickers TA, et al. . Cellular uptake and trafficking of antisense oligonucleotides. Nat Biotechnol 2017;35:230–7. 10.1038/nbt.3779
    1. Ali K, Soond DR, Pineiro R, et al. . Inactivation of PI(3)K p110δ breaks regulatory T-cell-mediated immune tolerance to cancer. Nature 2014;510:407–11. 10.1038/nature13444
    1. Carnevalli LS, Sinclair C, Taylor MA, et al. . PI3Kα/δ inhibition promotes anti-tumor immunity through direct enhancement of effector CD8+ T-cell activity. J Immunother Cancer 2018;6:158. 10.1186/s40425-018-0457-0
    1. Solomon I, Amann M, Goubier A, et al. . CD25-Treg-depleting antibodies preserving IL-2 signaling on effector T cells enhance effector activation and antitumor immunity. Nat Cancer 2020;1:1153–66. 10.1038/s43018-020-00133-0
    1. De Simone M, Arrigoni A, Rossetti G, et al. . Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity 2016;45:1135–47. 10.1016/j.immuni.2016.10.021
    1. Lukesova E, Boucek J, Rotnaglova E, et al. . High level of Tregs is a positive prognostic marker in patients with HPV-positive oral and oropharyngeal squamous cell carcinomas. Biomed Res Int 2014;2014:1–11. 10.1155/2014/303929
    1. Saito T, Nishikawa H, Wada H, et al. . Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016;22:679–84. 10.1038/nm.4086
    1. Huang AC, Orlowski RJ, Xu X, et al. . A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med 2019;25:454–61. 10.1038/s41591-019-0357-y
    1. Kim CG, Kim KH, Pyo K-H, et al. . Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann Oncol 2019;30:1104–13. 10.1093/annonc/mdz123
    1. Wei T, Zhong W, Li Q. Role of heterogeneous regulatory T cells in the tumor microenvironment. Pharmacol Res 2020;153:104659. 10.1016/j.phrs.2020.104659
    1. Miyao T, Floess S, Setoguchi R, et al. . Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity 2012;36:262–75. 10.1016/j.immuni.2011.12.012
    1. Overacre-Delgoffe AE, Chikina M, Dadey RE, et al. . Interferon-γ Drives T reg Fragility to Promote Anti-tumor Immunity. Cell 2017;169:1130–41. 10.1016/j.cell.2017.05.005
    1. Chaudhry A, Rudra D, Treuting P, et al. . CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 2009;326:986–91. 10.1126/science.1172702
    1. Levine AG, Mendoza A, Hemmers S, et al. . Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 2017;546:421–5. 10.1038/nature22360
    1. Ahmadzadeh M, Antony PA, Rosenberg SA. IL-2 and IL-15 each mediate de novo induction of FOXP3 expression in human tumor antigen-specific CD8 T cells. J Immunother 2007;30:294–302. 10.1097/CJI.0b013e3180336787
    1. Gavin MA, Torgerson TR, Houston E, et al. . Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc Natl Acad Sci U S A 2006;103:6659–64. 10.1073/pnas.0509484103
    1. Tran DQ, Ramsey H, Shevach EM. Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood 2007;110:2983–90. 10.1182/blood-2007-06-094656
    1. Proia TA, Singh M, Woessner R, et al. . STAT3 Antisense Oligonucleotide Remodels the Suppressive Tumor Microenvironment to Enhance Immune Activation in Combination with Anti-PD-L1. Clin Cancer Res 2020;26:6335–49. 10.1158/1078-0432.CCR-20-1066

Source: PubMed

3
Abonneren