Tumor-treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti-PD-1 therapy

Tali Voloshin, Noa Kaynan, Shiri Davidi, Yaara Porat, Anna Shteingauz, Rosa S Schneiderman, Einav Zeevi, Mijal Munster, Roni Blat, Catherine Tempel Brami, Shay Cahal, Aviran Itzhaki, Moshe Giladi, Eilon D Kirson, Uri Weinberg, Adrian Kinzel, Yoram Palti, Tali Voloshin, Noa Kaynan, Shiri Davidi, Yaara Porat, Anna Shteingauz, Rosa S Schneiderman, Einav Zeevi, Mijal Munster, Roni Blat, Catherine Tempel Brami, Shay Cahal, Aviran Itzhaki, Moshe Giladi, Eilon D Kirson, Uri Weinberg, Adrian Kinzel, Yoram Palti

Abstract

Tumor-treating fields (TTFields) are alternating electric fields in a specific frequency range (100-300 kHz) delivered to the human body through transducer arrays. In this study, we evaluated whether TTFields-mediated cell death can elicit antitumoral immunity and hence would be effectively combined with anti-PD-1 therapy. We demonstrate that in TTFields-treated cancer cells, damage-associated molecular patterns including high-mobility group B1 and adenosine triphosphate are released and calreticulin is exposed on the cell surface. Moreover, we show that TTFields treatment promotes the engulfment of cancer cells by dendritic cells (DCs) and DCs maturation in vitro, as well as recruitment of immune cells in vivo. Additionally, our study demonstrates that the combination of TTFields with anti-PD-1 therapy results in a significant decline of tumor volume and increase in the percentage of tumor-infiltrating leukocytes in two tumor models. In orthotopic lung tumors, these infiltrating leukocytes, specifically macrophages and DCs, showed elevated expression of PD-L1. Compatibly, cytotoxic T-cells isolated from these tumors demonstrated increased production of IFN-γ. In colon cancer tumors, T-cells infiltration was significantly increased following long treatment duration with TTFields plus anti-PD-1. Collectively, our results suggest that TTFields therapy can induce anticancer immune response. Furthermore, we demonstrate robust efficacy of concomitant application of TTFields and anti-PD-1 therapy. These data suggest that integrating TTFields with anti-PD-1 therapy may further enhance antitumor immunity, hence achieve better tumor control.

Keywords: Anti-PD-1; Autophagy; ER stress; Immunogenic cell death; Tumor-treating fields.

Conflict of interest statement

We wish to disclose that TV, NK, SD, YP, AS, MM, RSS, CTB, EZ, RB, SC, AI, MG, EDK, AK, and UW are employees of Novocure and hold stock in Novocure Ltd. Yoram Palti holds stocks in Novocure Ltd. Novocure holds several patents related to TTFields. The authors declare that there are no other conflict of interest.

Figures

Fig. 1
Fig. 1
TTFields kill cancer cells by triggering apoptosis followed by extracellular release of HMGB1. LLC-1 and murine CT-26 colon carcinoma cells were treated with TTFields for 24–72 h at optimal frequency. a Effect of TTFields treatment on cell counts. N ≥ 3, and data are presented as mean ± SD. b Elevation of percentage of early apoptotic cells (Annexin V +/7AAD-) and late apoptotic cells (Annexin V+/7AAD+) following treatment with TTFields. N ≥ 3. c Release of HMGB1 was monitored using ELISA assay. N ≥ 3, and data are presented as mean ± SD. P values were determined using one-way ANOVA followed by Dunnett’s post-test. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 2
Fig. 2
TTFields application mediate cell surface exposure of calreticulin. LLC-1 and CT-26 cells were treated with TTFields for 24–72 h at optimal frequency. A quantitative analysis of CRT surface exposure was performed using flow cytometry. a Representative plots of flow cytometry analysis of CRT, b CRT surface exposure among viable cells (7AAD-). c Median fluorescence intensity of CRT in viable cells. In b and cN ≥ 3, and data are presented as mean ± SD, d Upper panel—Immunoblot analysis of peIF2α. Lower panel—densitometric analysis (arbitrary units normalized on the expression of the housekeeping protein Vinculin). N ≥ 3, and data are presented as mean ± SEM; P values were determined using one-way ANOVA followed by Dunnett’s post-test. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 3
Fig. 3
TTFields induce autophagy-dependent reduction in intracellular ATP levels. LLC-1 cells were treated with TTFields for 24–72 h at optimal frequency. a Flow cytometry measurements of quinacrine staining in viable cells (7AAD-) as indicator of intracellular ATP levels (Upper panel). N ≥ 3, and data are presented as mean ± SD. Alternatively, ATP release induced by TTFields was measured by enzymatic methods (lower panel). N = 2, the results are from one representative experiment, and data are presented as mean ± SD. b Ultrastructural scanning transmission electron microscope analysis of LLC-1 cells treated with TTFields for 48 h. Autophagosomes (Red arrows) and autolysosomes (Blue arrows) are indicated. Magnification ×6000, ×15000. c Immunofluorescence staining for LC3 (green) and DAPI (blue; left panel). Magnification ×40. Quantification of LC3II signal per cell (right panel). N = 4, and the results are reported as mean ± SD of pooled independent experiments. d LLC-1 cells were either left untreated, or treated with TTFields for 24–72 h. CQ (20 µM) was added 4 h before cells were collected. Samples were immunoblotted for LC3 and GAPDH. N = 2, and the results are reported as mean ± SD of pooled independent experiments. P values were determined using one-way ANOVA followed by Dunnett’s post-test for (a-upper panel, d) or unpaired two-tailed t test for (a-lower panel, c). *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 4
Fig. 4
Culture of TTFields-treated cancer cell suspension induces maturation of bone marrow-derived dendritic cells. a, b Co-culture of cell tracker deep red labeled, untreated, or TTFields-treated LLC-1 cells and BMDCs. CD11c+Deep Red+events were quantified using flow cytometry. a Representative flow cytometry plot and b quantification of phagocytosis index. N = 3, and data are presented as mean ± SD. c–e Co-culture of untreated, TTFields-treated, F/T-treated, or LPS-treated LLC-1 cells and BMDCs. BMDCs maturation phenotype was assessed using flow cytometry. Quantification of activation markers: c MHC II (D-dim, I-intermediate, and B-bright expression), d CD80, and e CD40. N ≥ 4, and data are presented as mean ± SEM. f LLC-1 cells were either left untreated or treated with TTFields- (48 h), or F/T-treated cells and were intraperitoneally injected into wild-type C57/Bl6 mice. Equal volumes of PBS were injected in mice as negative controls. Peritoneal exudate cells were collected 48 h later and analyzed using flow cytometry. Graphs represent the number of CD45 + cells in peritoneal exudate cells. N = 2, bars indicate mean of two pooled independent experiments with 5–7 mice per group. Each circle represents one mouse. P values were determined using unpaired two-tailed t test for (b) or one-way ANOVA followed by Dunnett’s post-test (c–f). *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 5
Fig. 5
TTFields in combination with anti-PD-1 are therapeutically effective in murine lung cancer model. Ten-12-week-old male C57Bl/6 mice were injected directly into the lungs with Lewis lung carcinoma (LLC-1; 3 × 103 cells). a Application of TTFields to the mouse lungs was initiated 6 days afterward and was maintained for 7 days. Mice received an I.P. injection of anti-PD-1 (αPD-1) or Rat IgG2a, as indicated in the scheme. b At the end of the experiment, tumor volume was measured using Vernier calipers. N = 3, and the results are reported as mean ± SD of three pooled independent experiments with total of 5–8 mice per group. Tumors were harvested and analyzed for the percentages (ce) and PD-L1 expression levels (fh) of tumor (c, f) CD45+ cells, d, g dendritic cells (CD45+CD11c+), e, h macrophages (CD45+CD11b+F4/80+) using flow cytometry. Percentages of i CD3+CD8+, j CD3+CD4+, and k Foxp3+CD3+CD4+TILs. l, m TILs were harvested from LLC-1 tumor-bearing mice and stimulated with anti-CD3 and anti-CD28 before intracytoplasmic cytokine staining. Expression levels of IFN-γ+in CD3+CD8+ (l) and CD3+CD4+ (m). N = 2, and bars indicate mean of two pooled independent experiment with 5–8 mice per group. Each circle represents one mouse. P values were determined using the Kruskal–Wallis test followed by a Dunn’s post-test for (b) or unpaired two-tailed t test for (cm). *P < 0.05; **P < 0.01; ***P < 0.001. MFI Median fluorescence intensity
Fig. 6
Fig. 6
TTFields in combination with anti-PD-1 are therapeutically effective in murine colon cancer model. a Ten-week-old female Balb/c mice bearing 60 mm3 subcutaneous CT-26 tumors were treated with TTFields for 14 days, with a 3-day break (days 13–16). Mice received an I.P. injection of anti-PD-1 (αPD-1) or Rat IgG2a, as indicated in the scheme. b At the end of the experiment, tumor volume was measured using Vernier calipers. N = 2, and the results are reported as mean ± SD of two pooled independent experiments with total of 5–11 mice per group. Tumors were harvested and analyzed using flow cytometry for the percentages (c–e) and PD-L1 expression levels (fh) of tumor (c, f) CD45+ cells, d, g dendritic cells, e, h macrophages. Percentages of i CD3+CD8+, j CD3+CD4+, and k Foxp3+CD3+CD4+TILs. P values were determined using two-way ANOVA with Tukey’s post-test for (b) or unpaired two-tailed t test for (c–k). *P < 0.05; **P < 0.01; ***P < 0.001

References

    1. Vonderheide RH. The immune revolution: a case for priming, not checkpoint. Cancer Cell. 2018;33:563–569. doi: 10.1016/j.ccell.2018.03.008.
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. doi: 10.1016/j.ccell.2015.03.001.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi: 10.1038/nrc3239.
    1. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    1. Kirson ED, Gurvich Z, Schneiderman R, Dekel E, Itzhaki A, Wasserman Y, Schatzberger R, Palti Y. Disruption of cancer cell replication by alternating electric fields. Cancer Res. 2004;64:3288–3295. doi: 10.1158/0008-5472.CAN-04-0083.
    1. Giladi M, Schneiderman RS, Voloshin T, et al. Mitotic spindle disruption by alternating electric fields leads to improper chromosome segregation and mitotic catastrophe in cancer cells. Sci Rep. 2015;5:18046. doi: 10.1038/srep18046.
    1. Gera N, Yang A, Holtzman TS, Lee SX, Wong ET, Swanson KD. Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PLoS ONE. 2015;10:e0125269. doi: 10.1371/journal.pone.0125269.
    1. Kirson ED, Dbaly V, Tovarys F, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci USA. 2007;104:10152–10157. doi: 10.1073/pnas.0702916104.
    1. Porat Y, Giladi M, Schneiderman RS, et al. Determining the optimal inhibitory frequency for cancerous cells using tumor treating fields (TTFields) J Vis Exp. 2017 doi: 10.3791/55820.
    1. Yatim N, Cullen S, Albert ML. Dying cells actively regulate adaptive immune responses. Nat Rev Immunol. 2017;17:262–275. doi: 10.1038/nri.2017.9.
    1. Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol. 2009;9:353–363. doi: 10.1038/nri2545.
    1. Obeid M, Tesniere A, Ghiringhelli F, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523.
    1. Panaretakis T, Kepp O, Brockmeier U, et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009;28:578–590. doi: 10.1038/emboj.2009.1.
    1. Garg AD, Krysko DV, Verfaillie T, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012;31:1062–1079. doi: 10.1038/emboj.2011.497.
    1. Bezu L, Sauvat A, Humeau J, Leduc M, Kepp O, Kroemer G. eIF2alpha phosphorylation: a hallmark of immunogenic cell death. Oncoimmunology. 2018;7:e1431089. doi: 10.1080/2162402X.2018.1431089.
    1. Martins I, Wang Y, Michaud M, et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21:79–91. doi: 10.1038/cdd.2013.75.
    1. Martins I, Tesniere A, Kepp O, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8:3723–3728. doi: 10.4161/cc.8.22.10026.
    1. Michaud M, Martins I, Sukkurwala AQ, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–1577. doi: 10.1126/science.1208347.
    1. Silginer M, Weller M, Stupp R, Roth P. Biological activity of tumor-treating fields in preclinical glioma models. Cell Death Dis. 2017;8:e2753. doi: 10.1038/cddis.2017.171.
    1. Dupont N, Orhon I, Bauvy C, Codogno P. Autophagy and autophagic flux in tumor cells. Methods Enzymol. 2014;543:73–88. doi: 10.1016/B978-0-12-801329-8.00004-0.
    1. Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov. 2015;5:915–919. doi: 10.1158/-15-0563.
    1. Kirson ED, Giladi M, Gurvich Z, et al. Alternating electric fields (TTFields) inhibit metastatic spread of solid tumors to the lungs. Clin Exp Metastasis. 2009;26:633–640. doi: 10.1007/s10585-009-9262-y.
    1. Kleinovink JW, Fransen MF, Lowik CW, Ossendorp F. Photodynamic-immune checkpoint therapy eradicates local and distant tumors by CD8(+) T cells. Cancer Immunol Res. 2017;5:832–838. doi: 10.1158/2326-6066.CIR-17-0055.
    1. Hu ZI, McArthur HL, Ho AY. The abscopal effect of radiation therapy: What is it and how can we use it in breast cancer? Curr Breast Cancer Rep. 2017;9:45–51. doi: 10.1007/s12609-017-0234-y.
    1. Wong ET, Lok E, Swanson KD, Gautam S, Engelhard HH, Lieberman F, Taillibert S, Ram Z, Villano JL. Response assessment of NovoTTF-100A versus best physician’s choice chemotherapy in recurrent glioblastoma. Cancer Med. 2014;3:592–602. doi: 10.1002/cam4.210.
    1. Wong ET, Lok E, Gautam S, Swanson KD. Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113:232–241. doi: 10.1038/bjc.2015.238.
    1. Senovilla L, Vitale I, Martins I, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012;337:1678–1684. doi: 10.1126/science.1224922.
    1. Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, Zitvogel L, Kroemer G. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007;14:1848–1850. doi: 10.1038/sj.cdd.4402201.
    1. Stingele S, Stoehr G, Storchova Z. Activation of autophagy in cells with abnormal karyotype. Autophagy. 2013;9:246–248. doi: 10.4161/auto.22558.
    1. Elliott MR, Chekeni FB, Trampont PC, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461:282–286. doi: 10.1038/nature08296.
    1. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858.
    1. Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 2008;29:21–32. doi: 10.1016/j.immuni.2008.05.013.
    1. Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ, Chiorazzi N. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 2004;173:307–313. doi: 10.4049/jimmunol.173.1.307.
    1. Ribas A, Dummer R, Puzanov I, et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2017;170(1109–19):e10. doi: 10.1016/j.cell.2017.08.027.
    1. McNamara MJ, Hilgart-Martiszus I, Barragan Echenique DM, Linch SN, Kasiewicz MJ, Redmond WL. Interferon-gamma production by peripheral lymphocytes predicts survival of tumor-bearing mice receiving dual PD-1/CTLA-4 blockade. Cancer Immunol Res. 2016;4:650–657. doi: 10.1158/2326-6066.CIR-16-0022.
    1. Goldszmid RS, Caspar P, Rivollier A, White S, Dzutsev A, Hieny S, Kelsall B, Trinchieri G, Sher A. NK cell-derived interferon-gamma orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection. Immunity. 2012;36:1047–1059. doi: 10.1016/j.immuni.2012.03.026.
    1. Capasso A, Lang J, Pitts TM, et al. Characterization of immune responses to anti-PD-1 mono and combination immunotherapy in hematopoietic humanized mice implanted with tumor xenografts. J Immunother Cancer. 2019;7:37. doi: 10.1186/s40425-019-0518-z.
    1. Yu JW, Bhattacharya S, Yanamandra N, et al. Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironments. PLoS ONE. 2018;13:e0206223. doi: 10.1371/journal.pone.0206223.
    1. Lechner MG, Karimi SS, Barry-Holson K, Angell TE, Murphy KA, Church CH, Ohlfest JR, Hu P, Epstein AL. Immunogenicity of murine solid tumor models as a defining feature of in vivo behavior and response to immunotherapy. J Immunother. 2013;36:477–489. doi: 10.1097/01.cji.0000436722.46675.4a.
    1. Sun L, Clavijo PE, Robbins Y, et al. Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy. JCI Insight. 2019 doi: 10.1172/jci.insight.126853.
    1. Arlauckas SP, Garris CS, Kohler RH, et al. In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy. Sci Transl Med. 2017 doi: 10.1126/scitranslmed.aal3604.
    1. Georgoudaki AM, Prokopec KE, Boura VF, et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 2016;15:2000–2011. doi: 10.1016/j.celrep.2016.04.084.
    1. Roberts PC, Mottillo EP, Baxa AC, Heng HH, Doyon-Reale N, Gregoire L, Lancaster WD, Rabah R, Schmelz EM. Sequential molecular and cellular events during neoplastic progression: a mouse syngeneic ovarian cancer model. Neoplasia. 2005;7:944–956. doi: 10.1593/neo.05358.

Source: PubMed

3
Subskrybuj