DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity

Claire Vanpouille-Box, Amandine Alard, Molykutty J Aryankalayil, Yasmeen Sarfraz, Julie M Diamond, Robert J Schneider, Giorgio Inghirami, C Norman Coleman, Silvia C Formenti, Sandra Demaria, Claire Vanpouille-Box, Amandine Alard, Molykutty J Aryankalayil, Yasmeen Sarfraz, Julie M Diamond, Robert J Schneider, Giorgio Inghirami, C Norman Coleman, Silvia C Formenti, Sandra Demaria

Abstract

Radiotherapy is under investigation for its ability to enhance responses to immunotherapy. However, the mechanisms by which radiation induces anti-tumour T cells remain unclear. We show that the DNA exonuclease Trex1 is induced by radiation doses above 12-18 Gy in different cancer cells, and attenuates their immunogenicity by degrading DNA that accumulates in the cytosol upon radiation. Cytosolic DNA stimulates secretion of interferon-β by cancer cells following activation of the DNA sensor cGAS and its downstream effector STING. Repeated irradiation at doses that do not induce Trex1 amplifies interferon-β production, resulting in recruitment and activation of Batf3-dependent dendritic cells. This effect is essential for priming of CD8+ T cells that mediate systemic tumour rejection (abscopal effect) in the context of immune checkpoint blockade. Thus, Trex1 is an upstream regulator of radiation-driven anti-tumour immunity. Trex1 induction may guide the selection of radiation dose and fractionation in patients treated with immunotherapy.

Conflict of interest statement

The authors declare no competing financial interests related to this manuscript, but S.C.F. has received speaker compensation from Bristol-Myer Squibb, Sanofi, Regeneron, Varian, Elekta and Janssen, and S.D. currently serves as a consultant for Eisai, Inc, Lytix Biopharma and Nanobiotix.

Figures

Figure 1. Radiation-induced activation of type-I interferon…
Figure 1. Radiation-induced activation of type-I interferon pathway correlates with radiation's ability to induce abscopal responses in combination with anti-CTLA4.
(a,b) In mice with bilateral TSA tumours one tumour was irradiated (RT) and mice received anti-CTLA4 antibody as indicated (a). Growth of irradiated and abscopal tumours in mice treated with 0 Gy, 0 Gy+anti-CTLA4, 8GyX1, 8GyX1+anti-CTLA4, 30GyX1, 30GyX1+anti-CTLA4, 8GyX3 and 8GyX3+anti-CTLA4. Ratios indicate the number of mice free from the irradiated tumour. (Duplicate; asterisks indicate P values for the comparison of irradiated tumours in each group versus 0 Gy controls, *P<0.05; **P<0.005; hashs indicate P values for the comparison of tumours treated or not with anti-CTLA4 within each radiation level, ##P<0.005; two-way ANOVA; n=7). (b). (c) Heat map of gene expression in TSA tumours 24 h after radiation in vivo (n=3). (d) qRT-PCR (n=4) and IFNβ secretion (n=3) 24 h after in vitro irradiation of TSA cells (Triplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test). (e) Number of CD11c+CD8α+ DCs infiltrating TSA tumours 5 days after irradiation and CD70 mean fluorescence intensity (MFI) gated on CD11c+CD8α+ cells (n=5). (Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test). All data are mean±s.e.m.
Figure 2. IFNAR1 expression by TSA cancer…
Figure 2. IFNAR1 expression by TSA cancer cells is required for optimal therapeutic response of mice treated with 8GyX3 and anti-CTLA4.
(a) Upregulation of Ifnar1 expression measured by qRT-PCR 24 h after radiation in vitro in tetracycline-treated TSAshNS cells (black bars) is abrogated in TSAshIfnar1 cells (white bars) (Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3). (b) pTRIPZ lentiviral vector with tetracycline-inducible shRNA and TurboRFP fluorescent protein (tRFP) expression, and microscopic image of tetracycline-treated TSAshNS cells (magnification= × 20). (cf) Mice bearing irradiated TSAshIfnar1 or TSAshNS tumours and TSAshNS tumours in the abscopal site were treated with tetracycline and 0Gy (TSAshNS=black; TSAshIfnar1=dashed line), 8GyX3 (TSAshNS=green; TSAshIfnar1=yellow), 8GyX3+anti-CTLA4 (TSAshNS=blue; TSAshIfnar1=red). (c) Growth of irradiated and abscopal tumours. Duplicate; *P<0.05; **P<0.005: comparison of irradiated tumour outgrowth; two-way ANOVA; n=7. (d) Survival. Numbers indicates fraction of tumour-free mice. Log-rank (Mantel–Cox) test for the survival experiment; n=7 (e) IFNγ production by tumour-draining lymph node (TDLN) cells in response to the CD8 T cell epitopes AH1A5 (full circles) or control peptide MCMV (open circles). Each symbol represents one animal. Horizontal lines indicate the mean of antigen-specific (solid lines) or control (dashed lines) IFNγ concentration. Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3 (f) Individual tumour growth from (d) showing recurrence of all abscopal tumours in mice bearing irradiated TSAshIfnar1 tumours (red) while 3/7 mice with irradiated TSAshNS tumours (blue) remained tumour-free. All data are mean±s.e.m.
Figure 3. Trex1 is induced by high…
Figure 3. Trex1 is induced by high dose radiation and degrades IFN-inducing cytoplasmic DNA.
(ac) Cytoplasmic dsDNA accumulation (a), Trex1 gene expression (b), IFNβ secretion and Mx1 and Ifnar1 gene expression (c) in TSA cells treated with various radiation doses (n=3). (dg) TSAKI Trex1 cells were cultured without (grey bars) or with doxycycline (white bars) to induce Trex1 expression (Supplementary Fig. 3b) before measuring cytoplasmic dsDNA accumulation (d), Mx1 (e), Ifnar1 (f) and Ifnb1 (g) gene expression induced by radiation (n=3). (Duplicate; *P<0.05; **P<0.005; ***P<0.0005; ****P<0.0001: t-test). All data are mean±s.e.m.
Figure 4. Trex1 knockdown restores cytosolic dsDNA…
Figure 4. Trex1 knockdown restores cytosolic dsDNA accumulation and induction of ISGs in TSA cells treated with 20 Gy.
(a–c) TSAshNS and TSAshTrex1 cells were treated with doxycycline to induce Trex1 knockdown and tRFP expression, then exposed to a radiation dose of 8 Gy or 20 Gy. (a) After 24 h cells were fixed, permeabilized and stained with an antibody specific for dsDNA, followed by detection with Alexa fluor 488-conjugated secondary antibody. Representative micrographs show DAPI-stained nuclei (blue), cytoplasmic dsDNA (green), cytoplasmic tRFP (red), and the three channels combined. Magnification: × 400; duplicate; n=3 per group. White bars, 10 μm. (b,c) After 24 h cells were harvested for measurement of cytoplasmic dsDNA (b) or Trex1, Ifnb1 and Mx1 expression by qRT-PCR (c). Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3. All data are mean±s.e.m.
Figure 5. Cancer cell-intrinsic type-I IFN activation…
Figure 5. Cancer cell-intrinsic type-I IFN activation is mediated by cGAS-STING pathway and is required for radiation-induced abscopal responses.
(a) Doxycycline-inducible shRNA-mediated knockdown of cGAS (TSAshcGAS) or STING (TSAshSTING) in TSA cells completely abrogated IFNβ release and Ifnb1 and Mx1 gene expression induced by viral infection and 8GyX3 radiation in vitro. (bf) Mice with TSAshcGAS, or non-silencing shRNA (TSAshNS) in the irradiated tumour and TSAshNS in the abscopal tumour were treated with doxycycline, 8GyX3 and anti-CTLA4. IFNγ production by TDLN cells (b) and percentage of IFNγ+ CD8+ T cells in spleen (c) in response to CD8 epitope AH1A5 (full circles) or control peptide MCMV (open circles). Each symbol represents one animal. Horizontal lines indicate the mean of antigen-specific (solid lines) or control (dashed lines). (d) Representative fields (× 200) showing CD8+ cells (green), DAPI+ nuclei (blue), and RFP+ TSA cells (red), and mean number±s.d. of CD8+ cells per field in abscopal tumours harvested at day 22 from 8GyX3+anti-CTLA4-treated mice with irradiated TSAshcGAS (red squares) or TSAshNS (blue squares) tumours. White bars, 25 um. (e) Growth of irradiated and abscopal tumour in mice with TSAshNS cells treated with 0Gy (black), 8GyX3 (green), 8GyX3+anti-CTLA4 (blue), and mice with TSAshcGAS cells treated with 0Gy (dashed line), 8GyX3 (yellow), 8GyX3+anti-CTLA4 (red). (f) Survival of mice from 8GyX3+anti-CTLA4 that rejected the irradiated and abscopal tumour (n=4) and were rechallenged at day 100 with a tumorigenic inoculum of TSA cells, together with a group of naïve mice (n=5). (a-d) Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3. (e) Duplicate; *P<0.05; **P<0.005: comparison of irradiated tumour outgrowth; two-way ANOVA; n=7; ##P<0.005: comparison of abscopal tumour outgrowth; two-way ANOVA; n=7. All data are mean±s.e.m.
Figure 6. Trex1 regulates radiation-induced abscopal responses…
Figure 6. Trex1 regulates radiation-induced abscopal responses in combination with immune checkpoint inhibitors.
Half of the mice with doxycycline-inducible Trex1 in TSA cells (TSAKI Trex1) in the irradiated tumour and parental TSA (TSA-WT) in the abscopal tumour were given doxycycline and all mice were then treated with 8GyX3 and anti-CTLA4 (a,b) or anti-PD-1 (c,d). Growth of irradiated (a) and abscopal (b) tumour in mice treated with 0 Gy (black), 0 Gy+doxycycline (dashed line), 3 × 8 Gy (green), 3 × 8 Gy+doxycycline (yellow); 3 × 8 Gy+anti-CTLA4 (blue) and 3 × 8 Gy+anti-CTLA4+doxycycline (red). Growth of irradiated (c) and abscopal (d) tumour in mice treated with 0 Gy (black), 0 Gy+doxycycline (dashed line), 3 × 8 Gy (green), 3 × 8 Gy+doxycycline (yellow); 3 × 8 Gy+anti-PD-1 (blue) and 3 × 8 Gy+anti-PD-1+doxycycline (red). Mice with TSAshTrex1 (broken lines) or non-silencing shRNA (TSAshNS) (solid lines) in the irradiated tumour and TSAshNS in the abscopal tumour were treated with doxycycline to induce Trex1 knockdown (e,f). Growth of irradiated (e) and abscopal (f) tumour in mice treated with 0 Gy (black), 1 × 20 Gy (green), 1 × 20 Gy+anti-CTLA4 (blue), 2 × 20 Gy (yellow), 2 × 20 Gy+anti-CTLA4 (red). (Duplicate; *P<0.05; **P<0.005: comparison of irradiated tumour growth; two-way ANOVA; n=7; ##P<0.005: comparison of abscopal tumour growth; two-way ANOVA; n=7). All data are mean±s.e.m.
Figure 7. Threshold for Trex1 induction by…
Figure 7. Threshold for Trex1 induction by radiation in mouse cell lines.
Trex1 gene expressions and cytoplasmic dsDNA accumulations in TSA (a), MCA38 (b) and 4T1 (c) carcinoma cells measured 24 h after in vitro treatment. Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3. All data are mean±s.e.m.
Figure 8. Threshold for Trex1 induction by…
Figure 8. Threshold for Trex1 induction by radiation in human cell lines and induction of Trex1 and IFN-I pathway in a patient-derived lung adenocarcinoma xenograft.
(a,b) Trex1 expression and cytoplasmic dsDNA accumulations in MDA-MB-231 (a) and 4175TR (b) breast carcinoma cells 24 hrs after in vitro treatment. Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=3. All data are mean±s.e.m. (c) Trex1, Ifnb1 and Mx1 expression measured by qRT-PCR in a patient-derived tumour xenograft (PDTX) 24 h after mice treatment with tumour-directed radiation. Duplicate; *P<0.05; **P<0.005; ***P<0.0005: t-test; n=4. All data are mean±s.e.m.
Figure 9. Graphical summary.
Figure 9. Graphical summary.
Left, treatment with a radiation regimen that causes dsDNA accumulation in the cancer cells cytosol without inducing the DNAse Trex1 activates interferon type-I pathway via cGAS/STING. Downstream recruitment of DC and activation of CD8+ T cells is enabled and tumour rejection occurs in synergy with anti-CTLA4 or anti-PD-1 antibody. Right, in tumour treated with a dose of radiation above the threshold for Trex1 induction dsDNA is cleared from the cytosol precluding interferon-β release by the cancer cells. This leads to insufficient DC recruitment and activation and lack of CD8+ T cell activation resulting in absence of local and abscopal tumour regression in combination with immune checkpoint inhibitors.

References

    1. Sharma P. & Allison J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).
    1. Demaria S. et al.. Immune-mediated inhibition of metastases following treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 11, 728–734 (2005).
    1. Postow M. A. et al.. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 366, 925–931 (2012).
    1. Golden E. B., Demaria S., Schiff P. B., Chachoua A. & Formenti S. C. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol. Res. 1, 365–372 (2013).
    1. Twyman-Saint Victor C. et al.. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).
    1. Demaria S., Golden E. B. & Formenti S. C. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol. 1, 1325–1332 (2015).
    1. Lugade A. A. et al.. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J. Immunol. 174, 7516–7523 (2005).
    1. Lee Y. et al.. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 114, 589–595 (2009).
    1. Demaria S. & Formenti S. C. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front. Oncol. 2, 153 (2012).
    1. Demaria S., Coleman C. N. & Formenti S. C. Radiotherapy: changing the game in immunotherapy. Trends Cancer 2, 286–294 (2016).
    1. Cai X., Chiu Y. H. & Chen Z. J. The cGAS-cGAMP-STING pathway of cytosolic DNA sensing and signaling. Mol. Cell 54, 289–296 (2014).
    1. Dewan M. Z. et al.. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 15, 5379–5388 (2009).
    1. Fuertes M. B. et al.. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8a+ dendritic cells. J. Exp. Med. 208, 2005–2016 (2011).
    1. Keller A. M., Schildknecht A., Xiao Y., van den Broek M. & Borst J. Expression of costimulatory ligand CD70 on steady-state dendritic cells breaks CD8+ T cell tolerance and permits effective immunity. Immunity 29, 934–946 (2008).
    1. Ahn J. et al.. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 5, 5166 (2014).
    1. Yang Y. G., Lindahl T. & Barnes D. E. Trex1 exonuclease degrades ssDNA to prevent chronic checkpoint activation and autoimmune disease. Cell 131, 873–886 (2007).
    1. Stetson D. B., Ko J. S., Heidmann T. & Medzhitov R. Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell 134, 587–598 (2008).
    1. Gall A. et al.. Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity 36, 120–131 (2012).
    1. Sun L., Wu J., Du F., Chen X. & Chen Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).
    1. Gao P. et al.. Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153, 1094–1107 (2013).
    1. Woo S. R. et al.. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014).
    1. Deng L. et al.. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014).
    1. Spranger S., Bao R. & Gajewski T. F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).
    1. Pilones K. A., Aryankalayil J., Babb J. S. & Demaria S. Invariant natural killer T cells regulate anti-tumor immunity by controlling the population of dendritic cells in tumor and draining lymph nodes. J. Immunother. Cancer 2, 37 (2014).
    1. Pilones K. A., Vanpouille-Box C. & Demaria S. Combination of radiotherapy and immune checkpoint inhibitors. Semin. Radiat. Oncol. 25, 28–33 (2015).
    1. Biswas S. K. et al.. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation). Blood 107, 2112–2122 (2006).
    1. Matsumura S. et al.. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 181, 3099–3107 (2008).
    1. Demaria O. et al.. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc. Natl Acad. Sci. USA 112, 15408–15413 (2015).
    1. Apetoh L. et al.. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13, 1050–1059 (2007).
    1. Crow Y. J. & Manel N. Aicardi-Goutières syndrome and the type I interferonopathies. Nat. Rev. Immunol. 15, 429–440 (2015).
    1. Yan N., Regalado-Magdos A. D., Stiggelbout B., Lee-Kirsch M. A. & Lieberman J. The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1. Nat. Immunol. 11, 1005–1013 (2010).
    1. Golden E. B., Chachoua A., Fenton-Kerimian M. B., Demaria S. & Formenti S. C. Abscopal responses in metastatic non-small cell lung cancer (NSCLC ) patients treatded on a phase 2 study of combined radiation therapy and ipilimumab: evidence for the in situ vaccination hypothesis of radiation. Int. J. Radiat. Oncol. Biol. Phys. 93, S66–S67 (2015).
    1. Kang J., Demaria S. & Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J. Immunother. Cancer 4, 51 (2016).
    1. Xia T., Konno H., Ahn J. & Barber G. N. Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 14, 282–297 (2016).
    1. Lau L., Gray E. E., Brunette R. L. & Stetson D. B. DNA tumor virus oncogenes antagonize the cGAS-STING DNA-sensing pathway. Science 350, 568–571 (2015).
    1. Xia T., Konno H. & Barber G. N. Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Cancer Res. 76, 6747–6759 (2016).
    1. Aslakson C. J. & Miller F. R. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 52, 1399–1405 (1992).
    1. Rosato A. et al.. The cytotoxic T-lymphocyte response against a poorly immunogenic mammary adenocarcinoma is focused on a single immunodominant class I epitope derived from the gp70 Env product of an endogenous retrovirus. Cancer Res. 63, 2158–2163 (2003).
    1. Radoja S. et al.. CD8(+) tumor-infiltrating T cells are deficient in perforin-mediated cytolytic activity due to defective microtubule-organizing center mobilization and lytic granule exocytosis. J. Immunol. 167, 5042–5051 (2001).
    1. Vanpouille-Box C. et al.. TGFbeta is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 75, 2232–2242 (2015).
    1. Gao D. et al.. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903–906 (2013).

Source: PubMed

3
S'abonner