Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 7/2016

21.11.2015 | Focussed Research Review

Immune-modulating properties of ionizing radiation: rationale for the treatment of cancer by combination radiotherapy and immune checkpoint inhibitors

verfasst von: Anja Derer, Benjamin Frey, Rainer Fietkau, Udo S. Gaipl

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 7/2016

Einloggen, um Zugang zu erhalten

Abstract

Radiotherapy (RT) utilizes the DNA-damaging properties of ionizing radiation to control tumor growth and ultimately kill tumor cells. By modifying the tumor cell phenotype and the tumor microenvironment, it may also modulate the immune system. However, out-of-field reactions of RT mostly assume further immune activation. Here, the sequence of the applications of RT and immunotherapy is crucial, just as the dose and fractionation may be. Lower single doses may impact on tumor vascularization and immune cell infiltration in particular, while higher doses may impact on intratumoral induction and production of type I interferons. The induction of immunogenic cancer cell death seems in turn to be a common mechanism for most RT schemes. Dendritic cells (DCs) are activated by the released danger signals and by taking up tumor peptides derived from irradiated cells. DCs subsequently activate T cells, a process that has to be tightly controlled to ensure tolerance. Inhibitory pathways known as immune checkpoints exist for this purpose and are exploited by tumors to inhibit immune responses. Cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on T cells are two major checkpoints. The biological concepts behind the findings that RT in combination with anti-CTLA-4 and/or anti-PD-L1 blockade stimulates CD8+ T cell-mediated anti-tumor immunity are reviewed in detail. On this basis, we suggest clinically significant combinations and sequences of RT and immune checkpoint inhibition. We conclude that RT and immune therapies complement one another.
Literatur
5.
Zurück zum Zitat Apetoh L, Ghiringhelli F, Tesniere A et al (2007) Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 13:1050–1059. doi:10.1038/nm1622 CrossRefPubMed Apetoh L, Ghiringhelli F, Tesniere A et al (2007) Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 13:1050–1059. doi:10.​1038/​nm1622 CrossRefPubMed
10.
Zurück zum Zitat Frey B, Rubner Y, Kulzer L, Werthmoller N, Weiss EM, Fietkau R, Gaipl US (2014) Antitumor immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol Immunother 63:29–36. doi:10.1007/s00262-013-1474-y CrossRefPubMed Frey B, Rubner Y, Kulzer L, Werthmoller N, Weiss EM, Fietkau R, Gaipl US (2014) Antitumor immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol Immunother 63:29–36. doi:10.​1007/​s00262-013-1474-y CrossRefPubMed
11.
Zurück zum Zitat Gaipl US, Multhoff G, Scheithauer H, Lauber K, Hehlgans S, Frey B, Rodel F (2014) Kill and spread the word: stimulation of antitumor immune responses in the context of radiotherapy. Immunotherapy 6:597–610. doi:10.2217/imt.14.38 CrossRefPubMed Gaipl US, Multhoff G, Scheithauer H, Lauber K, Hehlgans S, Frey B, Rodel F (2014) Kill and spread the word: stimulation of antitumor immune responses in the context of radiotherapy. Immunotherapy 6:597–610. doi:10.​2217/​imt.​14.​38 CrossRefPubMed
13.
Zurück zum Zitat Werthmöller N, Frey B, Wunderlich R, Fietkau R, Gaipl US (2015) Modulation of radiochemoimmunotherapy-induced B16 melanoma cell death by the pan-caspase inhibitor zVAD-fmk induces anti-tumor immunity in a HMGB1-, nucleotide- and T-cell-dependent manner. Cell Death Dis 6:e1761. doi:10.1038/cddis.2015.129 CrossRefPubMedPubMedCentral Werthmöller N, Frey B, Wunderlich R, Fietkau R, Gaipl US (2015) Modulation of radiochemoimmunotherapy-induced B16 melanoma cell death by the pan-caspase inhibitor zVAD-fmk induces anti-tumor immunity in a HMGB1-, nucleotide- and T-cell-dependent manner. Cell Death Dis 6:e1761. doi:10.​1038/​cddis.​2015.​129 CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G (2010) Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11:700–714. doi:10.1038/nrm2970 CrossRefPubMed Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G (2010) Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11:700–714. doi:10.​1038/​nrm2970 CrossRefPubMed
19.
22.
Zurück zum Zitat Ganss R, Ryschich E, Klar E, Arnold B, Hammerling GJ (2002) Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res 62:1462–1470PubMed Ganss R, Ryschich E, Klar E, Arnold B, Hammerling GJ (2002) Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res 62:1462–1470PubMed
25.
Zurück zum Zitat Wunderlich R, Ernst A, Rodel F, Fietkau R, Ott O, Lauber K, Frey B, Gaipl US (2015) Low and moderate doses of ionizing radiation up to 2 Gy modulate transmigration and chemotaxis of activated macrophages, provoke an anti-inflammatory cytokine milieu, but do not impact upon viability and phagocytic function. Clin Exp Immunol 179:50–61. doi:10.1111/cei.12344 CrossRefPubMed Wunderlich R, Ernst A, Rodel F, Fietkau R, Ott O, Lauber K, Frey B, Gaipl US (2015) Low and moderate doses of ionizing radiation up to 2 Gy modulate transmigration and chemotaxis of activated macrophages, provoke an anti-inflammatory cytokine milieu, but do not impact upon viability and phagocytic function. Clin Exp Immunol 179:50–61. doi:10.​1111/​cei.​12344 CrossRefPubMed
30.
Zurück zum Zitat Pandha H, Pawelec G (2015) Immune checkpoint targeting as anti-cancer immunotherapy: promises, questions, challenges and the need for predictive biomarkers at ASCO 2015. Cancer Immunol Immunother 64:1071–1074. doi:10.1007/s00262-015-1748-7 CrossRefPubMed Pandha H, Pawelec G (2015) Immune checkpoint targeting as anti-cancer immunotherapy: promises, questions, challenges and the need for predictive biomarkers at ASCO 2015. Cancer Immunol Immunother 64:1071–1074. doi:10.​1007/​s00262-015-1748-7 CrossRefPubMed
31.
Zurück zum Zitat Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC (2005) Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 11:728–734PubMed Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC (2005) Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 11:728–734PubMed
33.
34.
Zurück zum Zitat Kim JE, Patel MA, Mangraviti A et al (2015) 143 the combination of anti-TIM-3 and anti-PD-1 checkpoint inhibitors with focused radiation resulted in a synergistic antitumor immune response in a preclinical glioma model. Neurosurgery 62(Suppl 1):212. doi:10.1227/01.neu.0000467105.60300.04 CrossRef Kim JE, Patel MA, Mangraviti A et al (2015) 143 the combination of anti-TIM-3 and anti-PD-1 checkpoint inhibitors with focused radiation resulted in a synergistic antitumor immune response in a preclinical glioma model. Neurosurgery 62(Suppl 1):212. doi:10.​1227/​01.​neu.​0000467105.​60300.​04 CrossRef
35.
Zurück zum Zitat Frey B, Rubner Y, Wunderlich R, Weiss EM, Pockley AG, Fietkau R, Gaipl US (2012) Induction of abscopal anti-tumor immunity and immunogenic tumor cell death by ionizing irradiation—implications for cancer therapies. Curr Med Chem 19:1751–1764CrossRefPubMed Frey B, Rubner Y, Wunderlich R, Weiss EM, Pockley AG, Fietkau R, Gaipl US (2012) Induction of abscopal anti-tumor immunity and immunogenic tumor cell death by ionizing irradiation—implications for cancer therapies. Curr Med Chem 19:1751–1764CrossRefPubMed
39.
Zurück zum Zitat Golden EB, Chhabra A, Chachoua A et al (2015) Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 16:795–803. doi:10.1016/s1470-2045(15)00054-6 CrossRefPubMed Golden EB, Chhabra A, Chachoua A et al (2015) Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 16:795–803. doi:10.​1016/​s1470-2045(15)00054-6 CrossRefPubMed
44.
Zurück zum Zitat Specht HM, Ahrens N, Blankenstein C et al (2015) Heat shock protein 70 (Hsp70) peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx)—from preclinical studies to a clinical phase ii trial. Front Immunol 6:162. doi:10.3389/fimmu.2015.00162 CrossRefPubMedPubMedCentral Specht HM, Ahrens N, Blankenstein C et al (2015) Heat shock protein 70 (Hsp70) peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx)—from preclinical studies to a clinical phase ii trial. Front Immunol 6:162. doi:10.​3389/​fimmu.​2015.​00162 CrossRefPubMedPubMedCentral
Metadaten
Titel
Immune-modulating properties of ionizing radiation: rationale for the treatment of cancer by combination radiotherapy and immune checkpoint inhibitors
verfasst von
Anja Derer
Benjamin Frey
Rainer Fietkau
Udo S. Gaipl
Publikationsdatum
21.11.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 7/2016
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-015-1771-8

Weitere Artikel der Ausgabe 7/2016

Cancer Immunology, Immunotherapy 7/2016 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.