Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway

Yu Kato, Kimiyo Tabata, Takayuki Kimura, Ayako Yachie-Kinoshita, Yoichi Ozawa, Kazuhiko Yamada, Junichi Ito, Sho Tachino, Yusaku Hori, Masahiro Matsuki, Yukiko Matsuoka, Samik Ghosh, Hiroaki Kitano, Kenichi Nomoto, Junji Matsui, Yasuhiro Funahashi, Yu Kato, Kimiyo Tabata, Takayuki Kimura, Ayako Yachie-Kinoshita, Yoichi Ozawa, Kazuhiko Yamada, Junichi Ito, Sho Tachino, Yusaku Hori, Masahiro Matsuki, Yukiko Matsuoka, Samik Ghosh, Hiroaki Kitano, Kenichi Nomoto, Junji Matsui, Yasuhiro Funahashi

Abstract

Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.

Conflict of interest statement

Lenvatinib is the marketed product. YK, KT, and YH obtained the patent entitled “Combination of a PD-1 antagonist and a VEGFR/FGFR/RET tyrosine kinase inhibitor for treating cancer” (WO2016140717A1). These do not interfere with our adherence to PLOS ONE policies regarding sharing data and materials.

Figures

Fig 1. Antitumor activity of lenvatinib in…
Fig 1. Antitumor activity of lenvatinib in immunocompetent and immunodeficient mice in the CT26 model.
A. Immunodeficient mice (Balb/cnu/nu) and immunocompetent mice (Balb/cwt/wt) inoculated with the CT26 cells were randomized into groups of 6 mice with an average tumor volume size (Day 1 mean TV: Balb/cnu/nu mice, 76.7 mm3; Balb/cwt/wt mice, 80.0 mm3), and were then treated with vehicle (blue circles) or 10 mg/kg lenvatinib (red squares) once daily (black arrows). Error bars indicate the SEM. B. The values of ΔT/C (%) were plotted for Balb/cnu/nu mice (red-filled squares) and Balb/cwt/wt mice (red-open squares). ****, P<0.0001; ***, P<0.001; Dunnett’s test vs vehicle, ***P<0.001, Dunnett’s test vs. Balb/cnu/nu mouse (n = 6). C. Mice, inoculated with CT26 cells, were randomized on Day 9 after CT26 inoculation with an average tumor volume size (Day 1 mean TV, 138 mm3) into groups of 6 or 7 mice, and were then treated with vehicle (blue bar) or 10 mg/kg lenvatinib (red bar). TILs were isolated on Day 8 after the start of treatment, and the percentage of TAMs gated by using Ly6C− F4/80+ (black square) in the CD11b+ Ly6G− population was analyzed and plotted. ****, P<0.0001, unpaired t-test vs. vehicle (n = 6 or 7). D. Immunohistochemical analysis of the TAM population in CT26 tumor tissues. CD11b is stained red, F4/80 is green, and DAPI is blue.
Fig 2. Antitumor activity of lenvatinib under…
Fig 2. Antitumor activity of lenvatinib under CD8+ T cell-depleted conditions in the CT26 model.
Mice, inoculated with CT26 cells, were treated with anti-CD8 antibody or control IgG on Day 6 and twice weekly thereafter. A. For flow cytometric analysis, the percentage of CD8+ T cells in the blood was determined. B. Two days after the first injection of anti-CD8 antibody or control IgG, mice were randomized into groups of 8 or 9 with an average tumor volume size (Day 1 mean TV: anti-CD8 antibody, 65.9 mm3; control IgG, 49.3 mm3), and then treated with vehicle (blue circles) or 10 mg/kg lenvatinib (red squares), indicated by the black arrow. Error bars represent the SEM. C. The percentage of ΔT/C (tumor size of each mouse treated with lenvatinib divided by the average tumor size of vehicle-treated mice) is shown for the control IgG (red-filled squares) and anti-CD8 antibody (red-open squares) treatment groups. **, P<0.01, Dunnett’s test vs. vehicle or control IgG (n = 8 or 9).
Fig 3. Antitumor effects of lenvatinib treatment…
Fig 3. Antitumor effects of lenvatinib treatment alone or in combination with anti-PD-1 in the CT26 model.
A. Mice were inoculated with CT26 cells and randomized into groups of 8 with an average tumor volume size (Day 1 mean TV, 32.6 mm3) and were then treated with vehicle (blue circles), 10 mg/kg lenvatinib (red squares) once daily, anti-PD-1 at 200 μg/mouse (green triangles) once every 3 days, or a combination of lenvatinib plus anti-PD-1 (purple circles). Lenvatinib treatment is indicated by the black arrow; anti-PD-1 treatment is indicated by black triangles. B. Tumor volumes for individual mice are shown for each treatment group. Error bars represent the SEM. **** P<0.0001, Dunnett’s test vs. vehicle on Day 19; ## P<0.01, #### P<0.0001 vs. combination.
Fig 4. Immune cell population analysis of…
Fig 4. Immune cell population analysis of lymphocytes in the CT26 tumor tissues.
A, Percentages of myeloid and TAM cell populations. Upper panel: TAMs, pDCs, granulocytes, and monocytes. Lower panel: M1, M2 in CD45+ cells, and the M1/M2 ratio. B. Upper panel: T cells, CD4+ T cells, and CD8+ T cells. Lower panel: PD-1+ Tim3+ CD8+ T cells, IFN-γ+ CD8+ T cells, and GzmB+CD8+ T cells. The gating strategy is shown in S1 Fig. *P <0.05, **P <0.01, ***P<0.001, unpaired t-test vs. vehicle; #P<0.05, unpaired t-test vs. combination (n = 6).
Fig 5. Weighted gene co-expression network analysis…
Fig 5. Weighted gene co-expression network analysis (WGCNA) to identify gene subsets modulated by combination treatment.
RNA-Seq analysis of tumor tissues followed by WGCNA was performed by using CT26 tumor tissues. A. Defined sample traits, each of which reflects a pattern of expression as indicated in the text. Gene subsets that correlate with one of traits (6) through (9) are considered to be possible target gene sets of the ‘potentiating effect’ of the two drugs. B. Modules from the WGCNA selected for trait correlation analysis. The modules highly correlated with one of the modules in A (r>0.65 and P<0.01) are shown with the number of genes included in each module in parentheses. The correlation strength of each module with each trait is shown in the heatmap where the most highly correlated trait is indicated by an asterisk. The significance (FDR-adjusted P value) of the top over-represented pathway in each module is shown in the bar graph, and the significantly enriched pathway is indicated (FDR-adjusted P value <0.05). The genes in the antiquewhite2 module represented in the type-I IFN signaling pathway are shown in the arrow box to the right. C. Eigengene expression pattern for the antiquewhite2 module.
Fig 6. Antitumor activity of lenvatinib under…
Fig 6. Antitumor activity of lenvatinib under IFN-γ-inhibited conditions in the CT26 model.
A. Mice, inoculated with CT26 cells, were treated with anti-IFN-γ antibody or control IgG on Day 8 after CT26 inoculation and twice weekly thereafter. The mice were randomized into groups of 7 with an average tumor volume size (Day 1 mean TV 71.1 mm3). The day after the first injection of anti-IFN-γ antibody or control IgG, mice were treated with vehicle (control IgG, dark blue; anti-IFN-γ antibody, light blue), 10 mg/kg lenvatinib (control IgG, dark red; anti-IFN-γ antibody, light red), anti-PD-1 (control IgG, dark green; anti-IFN-γ antibody, light green) or combination (control IgG, dark purple; anti-IFN-γ antibody, light purple). Lenvatinib treatment is indicated by the black arrow, and anti-PD-1 treatment is indicated as black triangles. Error bars represent the SEM. **P<0.01, ****P<0.0001, Dunnett’s test vs. control IgG (n = 7).

References

    1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565–70. Epub 2011/03/26. 10.1126/science.1203486 .
    1. Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol. 2013;14(12):1212–8. Epub 2013/11/19. 10.1038/ni.2762
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–54. Epub 2012/06/05. 10.1056/NEJMoa1200690
    1. McDermott DF, Atkins MB. PD-1 as a potential target in cancer therapy. Cancer Med. 2013;2(5):662–73. Epub 2014/01/10. 10.1002/cam4.106
    1. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. New England Journal of Medicine. 2016;375(19):1823–33.
    1. Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H, et al. Immunomodulatory Activity of Nivolumab in Metastatic Renal Cell Carcinoma. Clin Cancer Res. 2016;22(22):5461–71. Epub 2016/11/01. 10.1158/1078-0432.CCR-15-2839
    1. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369(2):134–44. Epub 2013/06/04. 10.1056/NEJMoa1305133
    1. Yamamoto Y, Matsui J, Matsushima T, Obaishi H, Miyazaki K, Nakamura K, et al. Lenvatinib, an angiogenesis inhibitor targeting VEGFR/FGFR, shows broad antitumor activity in human tumor xenograft models associated with microvessel density and pericyte coverage. Vasc Cell. 2014;6:18 Epub 2014/09/10. 10.1186/2045-824X-6-18
    1. Tohyama O, Matsui J, Kodama K, Hata-Sugi N, Kimura T, Okamoto K, et al. Antitumor activity of lenvatinib (e7080): an angiogenesis inhibitor that targets multiple receptor tyrosine kinases in preclinical human thyroid cancer models. J Thyroid Res. 2014;2014:638747 Epub 2014/10/09. 10.1155/2014/638747
    1. Okamoto K, Ikemori-Kawada M, Jestel A, von Konig K, Funahashi Y, Matsushima T, et al. Distinct binding mode of multikinase inhibitor lenvatinib revealed by biochemical characterization. ACS Med Chem Lett. 2015;6(1):89–94. Epub 2015/01/16. 10.1021/ml500394m
    1. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372(7):621–30. Epub 2015/02/12. 10.1056/NEJMoa1406470 .
    1. Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015;16(15):1473–82. Epub 2015/10/21. 10.1016/S1470-2045(15)00290-9 .
    1. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet. 2018. Epub 2018/02/13. 10.1016/s0140-6736(18)30207-1 .
    1. Tahara M, Schlumberger M, Elisei R, Habra MA, Kiyota N, Paschke R, et al. Exploratory analysis of biomarkers associated with clinical outcomes from the study of lenvatinib in differentiated cancer of the thyroid. Eur J Cancer. 2017;75:213–21. Epub 2017/02/27. 10.1016/j.ejca.2017.01.013 .
    1. Maaten L, Hinton G. Visualizing Data using t-SNE. Journal of Machine Learning Research. 2008;9:2579–605.
    1. Cesano A. nCounter((R)) PanCancer Immune Profiling Panel (NanoString Technologies, Inc., Seattle, WA). J Immunother Cancer. 2015;3:42 Epub 2015/12/18. 10.1186/s40425-015-0088-7
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–20. Epub 2014/04/04. 10.1093/bioinformatics/btu170
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. Epub 2012/10/30. 10.1093/bioinformatics/bts635
    1. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7(3):562–78. Epub 2012/03/03. 10.1038/nprot.2012.016
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559 Epub 2008/12/31. 10.1186/1471-2105-9-559
    1. Langfelder P, Zhang B, Horvath S. Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics. 2008;24(5):719–20. Epub 2007/11/21. 10.1093/bioinformatics/btm563 .
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. Epub 2009/01/10. 10.1038/nprot.2008.211 .
    1. Liang Y, Tang H, Guo J, Qiu X, Yang Z, Ren Z, et al. Targeting IFNα to tumor by anti-PD-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance. Nature Communications. 2018;9(1). 10.1038/s41467-018-06890-y
    1. McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nature Medicine. 2018;24(6):749–57. 10.1038/s41591-018-0053-3
    1. Kimura T, Kato Y, Ozawa Y, Kodama K, Ito J, Ichikawa K, et al. Immunomodulatory activity of lenvatinib plays a role in tumor growth inhibition in the Hepa1-6 mouse hepatocellular carcinoma syngeneic tumor model. Cancer Science. 2018;Forthcoming.
    1. Hettich M, Lahoti J, Prasad S, Niedermann G. Checkpoint Antibodies but not T Cell-Recruiting Diabodies Effectively Synergize with TIL-Inducing -Irradiation. Cancer Research. 2016;76(16):4673–83. 10.1158/0008-5472.CAN-15-3451
    1. Frey B, Rückert M, Weber J, Mayr X, Derer A, Lotter M, et al. Hypofractionated Irradiation Has Immune Stimulatory Potential and Induces a Timely Restricted Infiltration of Immune Cells in Colon Cancer Tumors. Frontiers in Immunology. 2017;8 10.3389/fimmu.2017.00231
    1. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212(2):139–48. Epub 2015/01/21. 10.1084/jem.20140559
    1. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 2016;165(1):35–44. Epub 2016/03/22. 10.1016/j.cell.2016.02.065
    1. Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol. 2009;86(5):1065–73. Epub 2009/09/11. 10.1189/jlb.0609385 .
    1. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49–61. Epub 2014/07/19. 10.1016/j.immuni.2014.06.010
    1. Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood. 1996;87(8):3336–43. Epub 1996/04/15. .
    1. Takase N, Koma Y, Urakawa N, Nishio M, Arai N, Akiyama H, et al. NCAM- and FGF-2-mediated FGFR1 signaling in the tumor microenvironment of esophageal cancer regulates the survival and migration of tumor-associated macrophages and cancer cells. Cancer Lett. 2016;380(1):47–58. Epub 2016/06/19. 10.1016/j.canlet.2016.06.009 .
    1. Ivashkiv LB. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nature Reviews Immunology. 2018;18(9):545–58. 10.1038/s41577-018-0029-z
    1. Lucarini V, Buccione C, Ziccheddu G, Peschiaroli F, Sestili P, Puglisi R, et al. Combining Type I Interferons and 5-Aza-2′-Deoxycitidine to Improve Anti-Tumor Response against Melanoma. Journal of Investigative Dermatology. 2017;137(1):159–69. 10.1016/j.jid.2016.08.024
    1. Frey B, Rückert M, Deloch L, Rühle PF, Derer A, Fietkau R, et al. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunological Reviews. 2017;280(1):231–48. 10.1111/imr.12572
    1. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nature Reviews Clinical Oncology. 2018;15(5):325–40. 10.1038/nrclinonc.2018.29
    1. Wu X, Li J, Connolly EM, Liao X, Ouyang J, Giobbie-Hurder A, et al. Combined Anti-VEGF and Anti–CTLA-4 Therapy Elicits Humoral Immunity to Galectin-1 Which Is Associated with Favorable Clinical Outcomes. Cancer Immunology Research. 2017;5(6):446–54. 10.1158/2326-6066.CIR-16-0385

Source: PubMed

3
S'abonner