Combining PARP Inhibition, Radiation, and Immunotherapy: A Possible Strategy to Improve the Treatment of Cancer?
Abstract
1. Introduction
1.1. The Immune System in Tumor Control
1.2. Combination of Treatments with Immunotherapies
2. Interactions and Synergy between PARPis and Immunotherapies in Tumor Control
2.1. PARPis as a Cytotoxic Treatment
2.2. PARPis and the Antitumor Immune Response
2.2.1. Anti-CTLA-4 and PARPis
2.2.2. Anti-PDL-1/PD-1 and PARPis
3. Immunotherapy and Radiation: A Synergistic Effect Mediated by Cytotoxic Lymphocytes
3.1. Ionizing Radiation Induces an Antitumor Immune Response Mediated by Cytotoxic Lymphocytes
3.2. Radiation and Immunotherapies Can Synergize to Control the Tumor
3.3. Radiation-Induced Immunosuppressive Effects Can Be Reversed with Immunotherapy
3.4. Inducing an Antitumor Immune Response with Proton or Carbon Ion Radiation
4. PARPis and Ionizing Radiation: A Promising Combination Therapy
5. Conclusions and Perspectives: A Rationale for Combining PARPis, Ionizing Radiation, and Immunotherapy
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
Abbreviations
APC | Antigen-presenting cell |
BRCA | BReast CAncer |
CCL2 | Chemokine (C-C motif) ligand 2 |
CCL5 | Chemokine (C-C motif) ligand 5 |
CTLs | specific activated CD8+ lymphocytes against tumor antigens |
CTLA-4 | Cytotoxic T lymphocyte-associated antigen 4 |
CXCL-16 | Chemokine (C-X-C motif) ligand 16 |
CXCL-10 | Chemokine (C-X-C motif) ligand 10 |
DC | Dendritic cell |
DSB | Double-strand break |
GBM | Glioblastoma |
Gy | Gray |
HMGB1 | High-mobility group box 1 |
MDSC | Myeloid-derived suppressor cells |
MHC | Major histocompatibility complex |
NSCLC | Non-small-cell lung cancer |
PARP | Poly(ADP-ribose) polymerase |
PARPi | PARP inhibitor |
PD-1 | Programmed cell death protein 1 |
PDL-1 | Programmed cell death ligand 1 |
TAA | Tumor-associated antigen |
TCR | T cell receptor |
TIL | Tumor-infiltrating lymphocyte |
TNBC | Triple-negative breast cancer |
TMZ | Temozolomide |
References
- Palucka, A.K.; Coussens, L.M. The Basis of Oncoimmunology. Cell 2016, 164, 1233–1247. [Google Scholar] [CrossRef] [PubMed]
- Badalamenti, G.; Fanale, D.; Incorvaia, L.; Barraco, N.; Listì, A.; Maragliano, R.; Vincenzi, B.; Calò, V.; Iovanna, J.L.; Bazan, V.; et al. Role of tumor-infiltrating lymphocytes in patients with solid tumors: Can a drop dig a stone? Cell. Immunol. 2018. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef] [PubMed]
- Geukes Foppen, M.H.; Donia, M.; Svane, I.M.; Haanen, J.B.A.G. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol. Oncol. 2015, 9, 1918–1935. [Google Scholar] [CrossRef] [PubMed]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [PubMed]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef] [PubMed]
- Candeias, S.M.; Gaipl, U.S. The Immune System in Cancer Prevention, Development and Therapy. Anticancer Agents Med. Chem. 2016, 16, 101–107. [Google Scholar] [CrossRef]
- Angell, H.; Galon, J. From the immune contexture to the Immunoscore: The role of prognostic and predictive immune markers in cancer. Curr. Opin. Immunol. 2013, 25, 261–267. [Google Scholar] [CrossRef]
- Taube, J.M.; Galon, J.; Sholl, L.M.; Rodig, S.J.; Cottrell, T.R.; Giraldo, N.A.; Baras, A.S.; Patel, S.S.; Anders, R.A.; Rimm, D.L.; et al. Implications of the tumor immune microenvironment for staging and therapeutics. Mod. Pathol. 2018, 31, 214–234. [Google Scholar] [CrossRef]
- Sharabi, A.B.; Lim, M.; DeWeese, T.L.; Drake, C.G. Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015, 16, e498–e509. [Google Scholar] [CrossRef]
- Walle, T.; Martinez Monge, R.; Cerwenka, A.; Ajona, D.; Melero, I.; Lecanda, F. Radiation effects on antitumor immune responses: Current perspectives and challenges. Ther. Adv. Med. Oncol. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
- Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed]
- Papa, A.; Caruso, D.; Strudel, M.; Tomao, S.; Tomao, F. Update on Poly-ADP-ribose polymerase inhibition for ovarian cancer treatment. J. Transl. Med. 2016, 14, 267. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed]
- Dudley, J.C.; Lin, M.-T.; Le, D.T.; Eshleman, J.R. Microsatellite Instability as a Biomarker for PD-1 Blockade. Clin. Cancer Res. 2016, 22, 813–820. [Google Scholar] [CrossRef] [PubMed]
- Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef]
- Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef]
- Greenwald, R.J.; Freeman, G.J.; Sharpe, A.H. The B7 family revisited. Annu. Rev. Immunol. 2005, 23, 515–548. [Google Scholar] [CrossRef]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef]
- Herzberg, B.; Campo, M.J.; Gainor, J.F. Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. Oncologist 2017, 22, 81–88. [Google Scholar] [CrossRef] [PubMed]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
- Khan, K.A.; Kerbel, R.S. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat. Rev. Clin. Oncol. 2018. [Google Scholar] [CrossRef] [PubMed]
- Gotwals, P.; Cameron, S.; Cipolletta, D.; Cremasco, V.; Crystal, A.; Hewes, B.; Mueller, B.; Quaratino, S.; Sabatos-Peyton, C.; Petruzzelli, L.; et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat. Rev. Cancer 2017, 17, 286–301. [Google Scholar] [CrossRef] [PubMed]
- Scott, C.L.; Swisher, E.M.; Kaufmann, S.H. Poly (ADP-ribose) polymerase inhibitors: Recent advances and future development. J. Clin. Oncol. 2015, 33, 1397–1406. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Yu, X. The role of poly(ADP-ribosyl)ation in DNA damage response and cancer chemotherapy. Oncogene 2015, 34, 3349–3356. [Google Scholar] [CrossRef]
- Scarpa, E.S.; Fabrizio, G.; Di Girolamo, M. A role of intracellular mono-ADP-ribosylation in cancer biology. FEBS J. 2013, 280, 3551–3562. [Google Scholar] [CrossRef]
- Dulaney, C.; Marcrom, S.; Stanley, J.; Yang, E.S. Poly(ADP-ribose) polymerase activity and inhibition in cancer. Semin. Cell Dev. Biol. 2017, 63, 144–153. [Google Scholar] [CrossRef]
- Morgan, R.D.; Clamp, A.R.; Evans, D.G.R.; Edmondson, R.J.; Jayson, G.C. PARP inhibitors in platinum-sensitive high-grade serous ovarian cancer. Cancer Chemother. Pharmacol. 2018. [Google Scholar] [CrossRef]
- Okuma, H.S.; Yonemori, K. BRCA Gene Mutations and Poly(ADP-Ribose) Polymerase Inhibitors in Triple-Negative Breast Cancer. Adv. Exp. Med. Biol. 2017, 1026, 271–286. [Google Scholar] [CrossRef]
- Mittica, G.; Ghisoni, E.; Giannone, G.; Genta, S.; Aglietta, M.; Sapino, A.; Valabrega, G. PARP Inhibitors in Ovarian Cancer. Recent Patents Anticancer Drug Discov. 2018. [Google Scholar] [CrossRef] [PubMed]
- Haddad, M.; Rhinn, H.; Bloquel, C.; Coqueran, B.; Szabó, C.; Plotkine, M.; Scherman, D.; Margaill, I. Anti-inflammatory effects of PJ34, a poly(ADP-ribose) polymerase inhibitor, in transient focal cerebral ischemia in mice. Br. J. Pharmacol. 2006, 149, 23–30. [Google Scholar] [CrossRef] [PubMed]
- Jijon, H.B.; Churchill, T.; Malfair, D.; Wessler, A.; Jewell, L.D.; Parsons, H.G.; Madsen, K.L. Inhibition of poly(ADP-ribose) polymerase attenuates inflammation in a model of chronic colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G641–G651. [Google Scholar] [CrossRef] [PubMed]
- Laudisi, F.; Sambucci, M.; Pioli, C. Poly (ADP-ribose) polymerase-1 (PARP-1) as immune regulator. Endocr. Metab. Immune Disord. Drug Targets 2011, 11, 326–333. [Google Scholar] [CrossRef]
- Aldinucci, A.; Gerlini, G.; Fossati, S.; Cipriani, G.; Ballerini, C.; Biagioli, T.; Pimpinelli, N.; Borgognoni, L.; Massacesi, L.; Moroni, F.; et al. A key role for poly(ADP-ribose) polymerase-1 activity during human dendritic cell maturation. J. Immunol. 2007, 179, 305–312. [Google Scholar] [CrossRef] [PubMed]
- Valdor, R.; Schreiber, V.; Saenz, L.; Martínez, T.; Muñoz-Suano, A.; Dominguez-Villar, M.; Ramírez, P.; Parrilla, P.; Aguado, E.; García-Cózar, F.; et al. Regulation of NFAT by poly(ADP-ribose) polymerase activity in T cells. Mol. Immunol. 2008, 45, 1863–1871. [Google Scholar] [CrossRef] [PubMed]
- Thorén, F.B.; Romero, A.I.; Hellstrand, K. Oxygen radicals induce poly(ADP-ribose) polymerase-dependent cell death in cytotoxic lymphocytes. J. Immunol. 2006, 176, 7301–7307. [Google Scholar] [CrossRef]
- Davalli, P.; Marverti, G.; Lauriola, A.; D’Arca, D. Targeting Oxidatively Induced DNA Damage Response in Cancer: Opportunities for Novel Cancer Therapies. Oxid. Med. Cell. Longev. 2018. [Google Scholar] [CrossRef]
- Huang, J.; Wang, L.; Cong, Z.; Amoozgar, Z.; Kiner, E.; Xing, D.; Orsulic, S.; Matulonis, U.; Goldberg, M.S. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1(-/-) murine model of ovarian cancer. Biochem. Biophys. Res. Commun. 2015, 463, 551–556. [Google Scholar] [CrossRef]
- Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef]
- Lança, T.; Costa, M.F.; Gonçalves-Sousa, N.; Rei, M.; Grosso, A.R.; Penido, C.; Silva-Santos, B. Protective role of the inflammatory CCR2/CCL2 chemokine pathway through recruitment of type 1 cytotoxic γδ T lymphocytes to tumor beds. J. Immunol. 2013, 190, 6673–6680. [Google Scholar] [CrossRef] [PubMed]
- Higuchi, T.; Flies, D.B.; Marjon, N.A.; Mantia-Smaldone, G.; Ronner, L.; Gimotty, P.A.; Adams, S.F. CTLA-4 Blockade Synergizes Therapeutically with PARP Inhibition in BRCA1-Deficient Ovarian Cancer. Cancer Immunol. Res. 2015, 3, 1257–1268. [Google Scholar] [CrossRef] [PubMed]
- Jiao, S.; Xia, W.; Yamaguchi, H.; Wei, Y.; Chen, M.-K.; Hsu, J.-M.; Hsu, J.L.; Yu, W.-H.; Du, Y.; Lee, H.-H.; et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin. Cancer Res. 2017. [Google Scholar] [CrossRef] [PubMed]
- Nebot-Bral, L.; Brandao, D.; Verlingue, L.; Rouleau, E.; Caron, O.; Despras, E.; El-Dakdouki, Y.; Champiat, S.; Aoufouchi, S.; Leary, A.; et al. Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. Eur. J. Cancer 2017, 84, 290–303. [Google Scholar] [CrossRef] [PubMed]
- Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2017, 168, 542. [Google Scholar] [CrossRef] [PubMed]
- Matsushita, H.; Hosoi, A.; Ueha, S.; Abe, J.; Fujieda, N.; Tomura, M.; Maekawa, R.; Matsushima, K.; Ohara, O.; Kakimi, K. Cytotoxic T lymphocytes block tumor growth both by lytic activity and IFNγ-dependent cell-cycle arrest. Cancer Immunol. Res. 2015, 3, 26–36. [Google Scholar] [CrossRef] [PubMed]
- Wall, L.; Burke, F.; Barton, C.; Smyth, J.; Balkwill, F. IFN-gamma induces apoptosis in ovarian cancer cells in vivo and in vitro. Clin. Cancer Res. 2003, 9, 2487–2496. [Google Scholar]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef]
- Du, X.; Tang, F.; Liu, M.; Su, J.; Zhang, Y.; Wu, W.; Devenport, M.; Lazarski, C.A.; Zhang, P.; Wang, X.; et al. A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy. Cell Res. 2018, 28, 416. [Google Scholar] [CrossRef]
- Sharma, A.; Subudhi, S.K.; Blando, J.; Scutti, J.; Vence, L.; Wargo, J.A.; Allison, J.P.; Ribas, A.; Sharma, P. Anti-CTLA-4 immunotherapy does not deplete FOXP3+ regulatory T cells (Tregs) in human cancers. Clin. Cancer Res. 2018. [Google Scholar] [CrossRef]
- Paradis, T.J.; Floyd, E.; Burkwit, J.; Cole, S.H.; Brunson, B.; Elliott, E.; Gilman, S.; Gladue, R.P. The anti-tumor activity of anti-CTLA-4 is mediated through its induction of IFN gamma. Cancer Immunol. Immunother. 2001, 50, 125–133. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.-M.; Cimino-Mathews, A.; Peer, C.J.; Zimmer, A.; Lipkowitz, S.; Annunziata, C.M.; Cao, L.; Harrell, M.I.; Swisher, E.M.; Houston, N.; et al. Safety and Clinical Activity of the Programmed Death-Ligand 1 Inhibitor Durvalumab in Combination with Poly (ADP-Ribose) Polymerase Inhibitor Olaparib or Vascular Endothelial Growth Factor Receptor 1-3 Inhibitor Cediranib in Women’s Cancers: A Dose-Escalation, Phase I Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 2193–2202. [Google Scholar] [CrossRef]
- Ko, E.C.; Formenti, S.C. Radiotherapy and checkpoint inhibitors: A winning new combination? Ther. Adv. Med. Oncol. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
- Frey, B.; Rückert, M.; Deloch, L.; Rühle, P.F.; Derer, A.; Fietkau, R.; Gaipl, U.S. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol. Rev. 2017, 280, 231–248. [Google Scholar] [CrossRef]
- Schaue, D.; Kachikwu, E.L.; McBride, W.H. Cytokines in radiobiological responses: A review. Radiat. Res. 2012, 178, 505–523. [Google Scholar] [CrossRef] [PubMed]
- Sharabi, A.B.; Nirschl, C.J.; Kochel, C.M.; Nirschl, T.R.; Francica, B.J.; Velarde, E.; Deweese, T.L.; Drake, C.G. Stereotactic Radiation Therapy Augments Antigen-Specific PD-1-Mediated Antitumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol. Res. 2015, 3, 345–355. [Google Scholar] [CrossRef] [PubMed]
- Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A.; Chakraborty, M.; Wansley, E.K.; Camphausen, K.; Luiten, R.M.; de Ru, A.H.; Neijssen, J.; et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef] [PubMed]
- Gameiro, S.R.; Jammeh, M.L.; Wattenberg, M.M.; Tsang, K.Y.; Ferrone, S.; Hodge, J.W. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 2014, 5, 403–416. [Google Scholar] [CrossRef]
- Vandenabeele, P.; Vandecasteele, K.; Bachert, C.; Krysko, O.; Krysko, D.V. Immunogenic Apoptotic Cell Death and Anticancer Immunity. Adv. Exp. Med. Biol. 2016, 930, 133–149. [Google Scholar] [CrossRef]
- Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef]
- Parker, J.J.; Jones, J.C.; Strober, S.; Knox, S.J. Characterization of direct radiation-induced immune function and molecular signaling changes in an antigen presenting cell line. Clin. Immunol. 2013, 148, 44–55. [Google Scholar] [CrossRef]
- Gupta, A.; Probst, H.C.; Vuong, V.; Landshammer, A.; Muth, S.; Yagita, H.; Schwendener, R.; Pruschy, M.; Knuth, A.; van den Broek, M. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J. Immunol. 2012, 189, 558–566. [Google Scholar] [CrossRef] [PubMed]
- Hallahan, D.; Kuchibhotla, J.; Wyble, C. Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer Res. 1996, 56, 5150–5155. [Google Scholar] [PubMed]
- Chow, M.T.; Luster, A.D. Chemokines in cancer. Cancer Immunol. Res. 2014, 2, 1125–1131. [Google Scholar] [CrossRef] [PubMed]
- Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef] [PubMed]
- Matsumura, S.; Demaria, S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat. Res. 2010, 173, 418–425. [Google Scholar] [CrossRef] [PubMed]
- Lugade, A.A.; Sorensen, E.W.; Gerber, S.A.; Moran, J.P.; Frelinger, J.G.; Lord, E.M. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J. Immunol. 2008, 180, 3132–3139. [Google Scholar] [CrossRef] [PubMed]
- Verma, V.; Cushman, T.R.; Tang, C.; Welsh, J.W. Toxicity of radiation and immunotherapy combinations. Adv. Radiat. Oncol. 2018, 3, 506–511. [Google Scholar] [CrossRef]
- Deng, L.; Liang, H.; Burnette, B.; Beckett, M.; Darga, T.; Weichselbaum, R.R.; Fu, Y.-X. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Investig. 2014, 124, 687–695. [Google Scholar] [CrossRef]
- Gong, X.; Li, X.; Jiang, T.; Xie, H.; Zhu, Z.; Zhou, F.; Zhou, C. Combined Radiotherapy and Anti-PD-L1 Antibody Synergistically Enhances Antitumor Effect in Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 1085–1097. [Google Scholar] [CrossRef]
- Derer, A.; Spiljar, M.; Bäumler, M.; Hecht, M.; Fietkau, R.; Frey, B.; Gaipl, U.S. Chemoradiation Increases PD-L1 Expression in Certain Melanoma and Glioblastoma Cells. Front. Immunol. 2016, 7, 610. [Google Scholar] [CrossRef]
- Demaria, S.; Kawashima, N.; Yang, A.M.; Devitt, M.L.; Babb, J.S.; Allison, J.P.; Formenti, S.C. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 2005, 11, 728–734. [Google Scholar] [PubMed]
- Twyman-Saint Victor, C.; Rech, A.J.; Maity, A.; Rengan, R.; Pauken, K.E.; Stelekati, E.; Benci, J.L.; Xu, B.; Dada, H.; Odorizzi, P.M.; et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015, 520, 373–377. [Google Scholar] [CrossRef]
- Park, S.S.; Dong, H.; Liu, X.; Harrington, S.M.; Krco, C.J.; Grams, M.P.; Mansfield, A.S.; Furutani, K.M.; Olivier, K.R.; Kwon, E.D. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol. Res. 2015, 3, 610–619. [Google Scholar] [CrossRef] [PubMed]
- Demaria, S.; Ng, B.; Devitt, M.L.; Babb, J.S.; Kawashima, N.; Liebes, L.; Formenti, S.C. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 862–870. [Google Scholar] [CrossRef] [PubMed]
- Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef] [PubMed]
- Rückert, M.; Deloch, L.; Fietkau, R.; Frey, B.; Hecht, M.; Gaipl, U.S. Immune modulatory effects of radiotherapy as basis for well-reasoned radioimmunotherapies. Strahlenther. Onkol. 2018, 194, 509–519. [Google Scholar] [CrossRef]
- Schmidt, E.V. Developing combination strategies using PD-1 checkpoint inhibitors to treat cancer. Semin. Immunopathol. 2018. [Google Scholar] [CrossRef]
- Samstein, R.M.; Riaz, N. The DNA damage response in immunotherapy and radiation. Adv. Radiat. Oncol. 2018, 3, 527–533. [Google Scholar] [CrossRef]
- Weichselbaum, R.R.; Liang, H.; Deng, L.; Fu, Y.-X. Radiotherapy and immunotherapy: A beneficial liaison? Nat. Rev. Clin. Oncol. 2017, 14, 365–379. [Google Scholar] [CrossRef]
- Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef] [PubMed]
- Vanpouille-Box, C.; Pilones, K.A.; Wennerberg, E.; Formenti, S.C.; Demaria, S. In situ vaccination by radiotherapy to improve responses to anti-CTLA-4 treatment. Vaccine 2015, 33, 7415–7422. [Google Scholar] [CrossRef]
- Burnette, B.C.; Liang, H.; Lee, Y.; Chlewicki, L.; Khodarev, N.N.; Weichselbaum, R.R.; Fu, Y.-X.; Auh, S.L. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011, 71, 2488–2496. [Google Scholar] [CrossRef] [PubMed]
- Wild, A.T.; Herman, J.M.; Dholakia, A.S.; Moningi, S.; Lu, Y.; Rosati, L.M.; Hacker-Prietz, A.; Assadi, R.K.; Saeed, A.M.; Pawlik, T.M.; et al. Lymphocyte-Sparing Effect of Stereotactic Body Radiation Therapy in Patients with Unresectable Pancreatic Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 571–579. [Google Scholar] [CrossRef] [PubMed]
- 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. 2013, 1, 365–372. [Google Scholar] [CrossRef] [PubMed]
- Grimaldi, A.M.; Simeone, E.; Giannarelli, D.; Muto, P.; Falivene, S.; Borzillo, V.; Giugliano, F.M.; Sandomenico, F.; Petrillo, A.; Curvietto, M.; et al. Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 2014, 3, e28780. [Google Scholar] [CrossRef] [PubMed]
- Ko, E.C.; Benjamin, K.T.; Formenti, S.C. Generating antitumor immunity by targeted radiation therapy: Role of dose and fractionation. Adv. Radiat. Oncol. 2018, 3, 486–493. [Google Scholar] [CrossRef]
- Tormoen, G.W.; Crittenden, M.R.; Gough, M.J. Role of the immunosuppressive microenvironment in immunotherapy. Adv. Radiat. Oncol. 2018, 3, 520–526. [Google Scholar] [CrossRef]
- Azad, A.; Yin Lim, S.; D’Costa, Z.; Jones, K.; Diana, A.; Sansom, O.J.; Kruger, P.; Liu, S.; McKenna, W.G.; Dushek, O.; et al. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol. Med. 2017, 9, 167–180. [Google Scholar] [CrossRef]
- Dovedi, S.J.; Adlard, A.L.; Lipowska-Bhalla, G.; McKenna, C.; Jones, S.; Cheadle, E.J.; Stratford, I.J.; Poon, E.; Morrow, M.; Stewart, R.; et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014, 74, 5458–5468. [Google Scholar] [CrossRef]
- Condamine, T.; Gabrilovich, D.I. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011, 32, 19–25. [Google Scholar] [CrossRef] [PubMed]
- Kachikwu, E.L.; Iwamoto, K.S.; Liao, Y.-P.; DeMarco, J.J.; Agazaryan, N.; Economou, J.S.; McBride, W.H.; Schaue, D. Radiation enhances regulatory T cell representation. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 1128–1135. [Google Scholar] [CrossRef] [PubMed]
- Sun, R.; Limkin, E.J.; Vakalopoulou, M.; Dercle, L.; Champiat, S.; Han, S.R.; Verlingue, L.; Brandao, D.; Lancia, A.; Ammari, S.; et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: An imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018. [Google Scholar] [CrossRef]
- Trowell, O.A. The sensitivity of lymphocytes to ionising radiation. J. Pathol. Bacteriol. 1952, 64, 687–704. [Google Scholar] [CrossRef] [PubMed]
- Song, W.Y.; Huh, S.N.; Liang, Y.; White, G.; Nichols, R.C.; Watkins, W.T.; Mundt, A.J.; Mell, L.K. Dosimetric comparison study between intensity modulated radiation therapy and three-dimensional conformal proton therapy for pelvic bone marrow sparing in the treatment of cervical cancer. J. Appl. Clin. Med. Phys. 2010, 11, 3255. [Google Scholar] [CrossRef] [PubMed]
- Gueulette, J.; Slabbert, J.P.; Böhm, L.; De Coster, B.M.; Rosier, J.F.; Octave-Prignot, M.; Ruifrok, A.; Schreuder, A.N.; Wambersie, A.; Scalliet, P.; et al. Proton RBE for early intestinal tolerance in mice after fractionated irradiation. Radiother. Oncol. 2001, 61, 177–184. [Google Scholar] [CrossRef]
- Uzawa, A.; Ando, K.; Koike, S.; Furusawa, Y.; Matsumoto, Y.; Takai, N.; Hirayama, R.; Watanabe, M.; Scholz, M.; Elsässer, T.; et al. Comparison of Biological Effectiveness of Carbon-Ion Beams in Japan and Germany. Int. J. Radiat. Oncol. 2009, 73, 1545–1551. [Google Scholar] [CrossRef]
- Durante, M.; Reppingen, N.; Held, K.D. Immunologically augmented cancer treatment using modern radiotherapy. Trends Mol. Med. 2013, 19, 565–582. [Google Scholar] [CrossRef]
- Gameiro, S.R.; Malamas, A.S.; Bernstein, M.B.; Tsang, K.Y.; Vassantachart, A.; Sahoo, N.; Tailor, R.; Pidikiti, R.; Guha, C.P.; Hahn, S.M.; et al. Tumor Cells Surviving Exposure to Proton or Photon Radiation Share a Common Immunogenic Modulation Signature, Rendering Them More Sensitive to T Cell-Mediated Killing. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 120–130. [Google Scholar] [CrossRef]
- Lee, K.-S.; Lee, D.-H.; Chun, S.-Y.; Nam, K.-S. Metastatic potential in MDA-MB-231 human breast cancer cells is inhibited by proton beam irradiation via the Akt/nuclear factor-κB signaling pathway. Mol. Med. Rep. 2014, 10, 1007–1012. [Google Scholar] [CrossRef]
- Ogata, T.; Teshima, T.; Kagawa, K.; Hishikawa, Y.; Takahashi, Y.; Kawaguchi, A.; Suzumoto, Y.; Nojima, K.; Furusawa, Y.; Matsuura, N. Particle irradiation suppresses metastatic potential of cancer cells. Cancer Res. 2005, 65, 113–120. [Google Scholar] [PubMed]
- Tamaki, T.; Iwakawa, M.; Ohno, T.; Imadome, K.; Nakawatari, M.; Sakai, M.; Tsujii, H.; Nakano, T.; Imai, T. Application of carbon-ion beams or gamma-rays on primary tumors does not change the expression profiles of metastatic tumors in an in vivo murine model. Int. J. Radiat. Oncol. Biol. Phys. 2009, 74, 210–218. [Google Scholar] [CrossRef] [PubMed]
- Ebner, D.K.; Kamada, T.; Yamada, S. Abscopal effect in recurrent colorectal cancer treated with carbon-ion radiation therapy: 2 case reports. Adv. Radiat. Oncol. 2017, 2, 333–338. [Google Scholar] [CrossRef] [PubMed]
- Mansour, W.Y.; Rhein, T.; Dahm-Daphi, J. The alternative end-joining pathway for repair of DNA double-strand breaks requires PARP1 but is not dependent upon microhomologies. Nucleic Acids Res. 2010, 38, 6065–6077. [Google Scholar] [CrossRef]
- Lee, H.-J.; Yoon, C.; Schmidt, B.; Park, D.J.; Zhang, A.Y.; Erkizan, H.V.; Toretsky, J.A.; Kirsch, D.G.; Yoon, S.S. Combining PARP-1 inhibition and radiation in Ewing sarcoma results in lethal DNA damage. Mol. Cancer Ther. 2013, 12, 2591–2600. [Google Scholar] [CrossRef] [PubMed]
- Tuli, R.; Surmak, A.J.; Reyes, J.; Armour, M.; Hacker-Prietz, A.; Wong, J.; DeWeese, T.L.; Herman, J.M. Radiosensitization of Pancreatic Cancer Cells In Vitro and In Vivo through Poly (ADP-ribose) Polymerase Inhibition with ABT-888. Transl. Oncol. 2014. [Google Scholar] [CrossRef]
- Lemasson, B.; Wang, H.; Galbán, S.; Li, Y.; Zhu, Y.; Heist, K.A.; Tsein, C.; Chenevert, T.L.; Rehemtulla, A.; Galbán, C.J.; et al. Evaluation of Concurrent Radiation, Temozolomide and ABT-888 Treatment Followed by Maintenance Therapy with Temozolomide and ABT-888 in a Genetically Engineered Glioblastoma Mouse Model. Neoplasia 2016, 18, 82–89. [Google Scholar] [CrossRef]
- Lesueur, P.; Chevalier, F.; Austry, J.-B.; Waissi, W.; Burckel, H.; Noël, G.; Habrand, J.-L.; Saintigny, Y.; Joly, F. Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: A systematic review of pre-clinical and clinical human studies. Oncotarget 2017, 8, 69105–69124. [Google Scholar] [CrossRef]
- McLachlan, J.; George, A.; Banerjee, S. The Current Status of PARP Inhibitors in Ovarian Cancer, The Current Status of PARP Inhibitors in Ovarian Cancer. Tumori J. 2016, 102, 433–440. [Google Scholar] [CrossRef]
- Paoluzzi, L.; Cacavio, A.; Ghesani, M.; Karambelkar, A.; Rapkiewicz, A.; Weber, J.; Rosen, G. Response to anti-PD1 therapy with nivolumab in metastatic sarcomas. Clin. Sarcoma Res. 2016, 6, 24. [Google Scholar] [CrossRef]
- Kostine, M.; Cleven, A.H.; de Miranda, N.F.C.C.; Italiano, A.; Cleton-Jansen, A.-M.; Bovée, J.V.M.G. Analysis of PD-L1, T-cell infiltrate and HLA expression in chondrosarcoma indicates potential for response to immunotherapy specifically in the dedifferentiated subtype. Mod. Pathol. 2016, 29, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
- Enderlin, M.; Kleinmann, E.V.; Struyf, S.; Buracchi, C.; Vecchi, A.; Kinscherf, R.; Kiessling, F.; Paschek, S.; Sozzani, S.; Rommelaere, J.; et al. TNF-alpha and the IFN-gamma-inducible protein 10 (IP-10/CXCL-10) delivered by parvoviral vectors act in synergy to induce antitumor effects in mouse glioblastoma. Cancer Gene Ther. 2009, 16, 149–160. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, A.; Schulte, A.; Schnack, C.; Hundhausen, C.; Reiss, K.; Brodway, N.; Held-Feindt, J.; Mentlein, R. Enhanced expression and shedding of the transmembrane chemokine CXCL16 by reactive astrocytes and glioma cells. J. Neurochem. 2005, 93, 1293–1303. [Google Scholar] [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Césaire, M.; Thariat, J.; Candéias, S.M.; Stefan, D.; Saintigny, Y.; Chevalier, F. Combining PARP Inhibition, Radiation, and Immunotherapy: A Possible Strategy to Improve the Treatment of Cancer? Int. J. Mol. Sci. 2018, 19, 3793. https://doi.org/10.3390/ijms19123793
Césaire M, Thariat J, Candéias SM, Stefan D, Saintigny Y, Chevalier F. Combining PARP Inhibition, Radiation, and Immunotherapy: A Possible Strategy to Improve the Treatment of Cancer? International Journal of Molecular Sciences. 2018; 19(12):3793. https://doi.org/10.3390/ijms19123793
Chicago/Turabian StyleCésaire, Mathieu, Juliette Thariat, Serge M. Candéias, Dinu Stefan, Yannick Saintigny, and François Chevalier. 2018. "Combining PARP Inhibition, Radiation, and Immunotherapy: A Possible Strategy to Improve the Treatment of Cancer?" International Journal of Molecular Sciences 19, no. 12: 3793. https://doi.org/10.3390/ijms19123793
APA StyleCésaire, M., Thariat, J., Candéias, S. M., Stefan, D., Saintigny, Y., & Chevalier, F. (2018). Combining PARP Inhibition, Radiation, and Immunotherapy: A Possible Strategy to Improve the Treatment of Cancer? International Journal of Molecular Sciences, 19(12), 3793. https://doi.org/10.3390/ijms19123793