Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Radiation Initiates Intratumoral Immune Responses
2.1. Conventional Radiation Induces Immunogenic Cell Death Molecules
2.2. Conventional Radiation Enhances (Neo)Antigen Expression
3. Dose Scheduling Effects of Conventional Radiation on Tumor Immunity
3.1. Single Dose Schedule
3.2. Fractionation Schedule
4. Window of Opportunity to Achieve Synergy between Radiotherapy and Immunotherapy
5. Immunotherapeutic Options and Conventional Radiotherapy
5.1. Vaccination
5.2. Adoptive Transfer
5.3. Agonists
5.4. Conjugated Cytokines
5.5. Immune Checkpoint Inhibitor Molecules and Signaling Pathways
6. Charged Particle Radiotherapy
Immune Modulatory Potential of Charged Particle Radiation
7. Charged Particle Radiation in Combination with Immunotherapy
8. Discussion and Perspectives
9. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Ceylan, C.; Hamacı, A.; Ayata, H.; Berberoglu, K.; Kılıç, A.; Güden, M.; Engin, K. Re-Irradiation of Locoregional NSCLC Recurrence Using Robotic Stereotactic Body Radiotherapy. Oncol. Res. Treat. 2017, 40, 207–214. [Google Scholar] [CrossRef] [PubMed]
- Formenti, S.C.; Demaria, S. Combining radiotherapy and cancer immunotherapy: A paradigm shift. J. Natl. Cancer Inst. 2013, 105, 256–265. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.E.; Patel, M.A.; Mangraviti, A.; Kim, E.S.; Theodros, D.; Velarde, E.; Liu, A.; Sankey, E.W.; Tam, A.; Xu, H.; et al. Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas. Clin. Cancer Res. 2017, 23, 124–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dovedi, S.J.; Lipowska-Bhalla, G.; Beers, S.A.; Cheadle, E.J.; Mu, L.; Glennie, M.J.; Illidge, T.M.; Honeychurch, J. Antitumor Efficacy of Radiation plus Immunotherapy Depends upon Dendritic Cell Activation of Effector CD8+ T Cells. Cancer Immunol. Res. 2016, 4, 621–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herrera, F.G.; Irving, M.; Kandalaft, L.E.; Coukos, G. Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol. 2019, 20, e417–e433. [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]
- Dokic, I.; Niklas, M.; Zimmermann, F.; Mairani, A.; Seidel, P.; Krunic, D.; Jakel, O.; Debus, J.; Greilich, S.; Abdollahi, A. Correlation of Particle Traversals with Clonogenic Survival Using Cell-Fluorescent Ion Track Hybrid Detector. Front. Oncol. 2015, 5, 275. [Google Scholar] [CrossRef] [Green Version]
- Niklas, M.; Greilich, S.; Melzig, C.; Akselrod, M.S.; Debus, J.; Jakel, O.; Abdollahi, A. Engineering cell-fluorescent ion track hybrid detectors. Radiat. Oncol. 2013, 8, 141. [Google Scholar] [CrossRef] [Green Version]
- Walsh, D.W.M.; Liew, H.; Schlegel, J.; Mairani, A.; Abdollahi, A.; Niklas, M. Carbon ion dosimetry on a fluorescent nuclear track detector using widefield microscopy. Phys. Med. Biol. 2020, 65, 21NT02. [Google Scholar] [CrossRef]
- Pantelias, A.; Zafiropoulos, D.; Cherubini, R.; Sarchiapone, L.; De Nadal, V.; Pantelias, G.E.; Georgakilas, A.G.; Terzoudi, G.I. Interphase Cytogenetic Analysis of G0 Lymphocytes Exposed to α-Particles, C-Ions, and Protons Reveals their Enhanced Effectiveness for Localized Chromosome Shattering-A Critical Risk for Chromothripsis. Cancers 2020, 12, 2336. [Google Scholar] [CrossRef] [PubMed]
- Dokic, I.; Mairani, A.; Niklas, M.; Zimmermann, F.; Chaudhri, N.; Krunic, D.; Tessonnier, T.; Ferrari, A.; Parodi, K.; Jakel, O.; et al. Next generation multi-scale biophysical characterization of high precision cancer particle radiotherapy using clinical proton, helium-, carbon- and oxygen ion beams. Oncotarget 2016, 7, 56676–56689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garnett, C.T.; Palena, C.; Chakraborty, M.; Tsang, K.Y.; Schlom, J.; Hodge, J.W. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004, 64, 7985–7994. [Google Scholar] [CrossRef] [Green Version]
- Dovedi, S.J.; Cheadle, E.J.; Popple, A.L.; Poon, E.; Morrow, M.; Stewart, R.; Yusko, E.C.; Sanders, C.M.; Vignali, M.; Emerson, R.O.; et al. Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade. Clin. Cancer Res. 2017, 23, 5514–5526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aliru, M.L.; Schoenhals, J.E.; Venkatesulu, B.P.; Anderson, C.C.; Barsoumian, H.B.; Younes, A.I.; Lakshmi, S.K.M.; Soeung, M.; Aziz, K.E.; Welsh, J.W.; et al. Radiation therapy and immunotherapy: What is the optimal timing or sequencing? Immunotherapy 2018, 10, 299–316. [Google Scholar] [CrossRef]
- Emens, L.A.; Ascierto, P.A.; Darcy, P.K.; Demaria, S.; Eggermont, A.M.M.; Redmond, W.L.; Seliger, B.; Marincola, F.M. Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur. J. Cancer 2017, 81, 116–129. [Google Scholar] [CrossRef]
- Wilkins, R.C.; Wilkinson, D.; Maharaj, H.P.; Bellier, P.V.; Cybulski, M.B.; McLean, J.R.N. Differential apoptotic response to ionizing radiation in subpopulations of human white blood cells. Mutat. Res. /Genet. Toxicol. Environ. Mutagenesis 2002, 513, 27–36. [Google Scholar] [CrossRef]
- Vanpouille-Box, C.; Diamond, J.M.; Pilones, K.A.; Zavadil, J.; Babb, J.S.; Formenti, S.C.; Barcellos-Hoff, M.H.; Demaria, S. TGFβ Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res. 2015, 75, 2232–2242. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-Ruiz, M.E.; Rodríguez, I.; Mayorga, L.; Labiano, T.; Barbes, B.; Etxeberria, I.; Ponz-Sarvise, M.; Azpilikueta, A.; Bolaños, E.; Sanmamed, M.F.; et al. TGFβ Blockade Enhances Radiotherapy Abscopal Efficacy Effects in Combination with Anti-PD1 and Anti-CD137 Immunostimulatory Monoclonal Antibodies. Mol. Cancer Ther. 2019, 18, 621–631. [Google Scholar] [CrossRef] [Green Version]
- Gong, J.; Le, T.Q.; Massarelli, E.; Hendifar, A.E.; Tuli, R. Radiation therapy and PD-1/PD-L1 blockade: The clinical development of an evolving anticancer combination. J. Immunother. Cancer 2018, 6, 46. [Google Scholar] [CrossRef]
- Di Maggio, F.M.; Minafra, L.; Forte, G.I.; Cammarata, F.P.; Lio, D.; Messa, C.; Gilardi, M.C.; Bravatà, V. Portrait of inflammatory response to ionizing radiation treatment. J. Inflamm. 2015, 12, 14. [Google Scholar] [CrossRef] [Green Version]
- Schaue, D.; Kachikwu, E.L.; McBride, W.H. Cytokines in radiobiological responses: A review. Radiat. Res. 2012, 178, 505–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Goldszmid, R.S.; Idoyaga, J.; Bravo, A.I.; Steinman, R.; Mordoh, J.; Wainstok, R. Dendritic cells charged with apoptotic tumor cells induce long-lived protective CD4+ and CD8+ T cell immunity against B16 melanoma. J. Immunol. 2003, 171, 5940–5947. [Google Scholar] [CrossRef] [Green Version]
- Malynn, B.A.; Romeo, D.T.; Wortis, H.H. Antigen-specific B cells efficiently present low doses of antigen for induction of T cell proliferation. J. Immunol. 1985, 135, 980–988. [Google Scholar]
- 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] [Green Version]
- Hellevik, T.; Pettersen, I.; Berg, V.; Winberg, J.O.; Moe, B.T.; Bartnes, K.; Paulssen, R.H.; Busund, L.T.; Bremnes, R.; Chalmers, A.; et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat. Oncol. 2012, 7, 59. [Google Scholar] [CrossRef] [Green Version]
- McLaughlin, M.; Patin, E.C.; Pedersen, M.; Wilkins, A.; Dillon, M.T.; Melcher, A.A.; Harrington, K.J. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat. Rev. Cancer 2020, 20, 203–217. [Google Scholar] [CrossRef]
- Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef]
- Blonska, M.; Agarwal, N.K.; Vega, F. Shaping of the tumor microenvironment: Stromal cells and vessels. Semin. Cancer Biol. 2015, 34, 3–13. [Google Scholar] [CrossRef]
- Liu, Y.; Dong, Y.; Kong, L.; Shi, F.; Zhu, H.; Yu, J. Abscopal effect of radiotherapy combined with immune checkpoint inhibitors. J. Hematol. Oncol. 2018, 11, 104. [Google Scholar] [CrossRef] [Green Version]
- Gómez, V.; Mustapha, R.; Ng, K.; Ng, T. Radiation therapy and the innate immune response: Clinical implications for immunotherapy approaches. Br. J. Clin. Pharmacol. 2020, 86, 1726–1735. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dewey, W.C.; Ling, C.C.; Meyn, R.E. Radiation-induced apoptosis: Relevance to radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 1995, 33, 781–796. [Google Scholar] [CrossRef]
- Wu, Q.; Allouch, A.; Martins, I.; Brenner, C.; Modjtahedi, N.; Deutsch, E.; Perfettini, J.-L. Modulating Both Tumor Cell Death and Innate Immunity Is Essential for Improving Radiation Therapy Effectiveness. Front. Immunol. 2017, 8, 613. [Google Scholar] [CrossRef]
- Eriksson, D.; Stigbrand, T. Radiation-induced cell death mechanisms. Tumour Biol. 2010, 31, 363–372. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Harding, S.M.; Natesan, R.; Tian, L.; Benci, J.L.; Li, W.; Minn, A.J.; Asangani, I.A.; Greenberg, R.A. Cell Cycle Checkpoints Cooperate to Suppress DNA- and RNA-Associated Molecular Pattern Recognition and Anti-Tumor Immune Responses. Cell Rep. 2020, 32, 108080. [Google Scholar] [CrossRef]
- Sachet, M.; Liang, Y.Y.; Oehler, R. The immune response to secondary necrotic cells. Apoptosis 2017, 22, 1189–1204. [Google Scholar] [CrossRef] [Green Version]
- Sia, J.; Szmyd, R.; Hau, E.; Gee, H.E. Molecular Mechanisms of Radiation-Induced Cancer Cell Death: A Primer. Front. Cell Dev. Biol. 2020, 8, 41. [Google Scholar] [CrossRef]
- Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
- Kepp, O.; Senovilla, L.; Vitale, I.; Vacchelli, E.; Adjemian, S.; Agostinis, P.; Apetoh, L.; Aranda, F.; Barnaba, V.; Bloy, N.; et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014, 3, e955691. [Google Scholar] [CrossRef] [Green Version]
- Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garg, A.D.; Galluzzi, L.; Apetoh, L.; Baert, T.; Birge, R.B.; Bravo-San Pedro, J.M.; Breckpot, K.; Brough, D.; Chaurio, R.; Cirone, M.; et al. Molecular and Translational Classifications of DAMPs in Immunogenic Cell Death. Front. Immunol. 2015, 6, 588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zappasodi, R.; Pupa, S.M.; Ghedini, G.C.; Bongarzone, I.; Magni, M.; Cabras, A.D.; Colombo, M.P.; Carlo-Stella, C.; Gianni, A.M.; Di Nicola, M. Improved clinical outcome in indolent B-cell lymphoma patients vaccinated with autologous tumor cells experiencing immunogenic death. Cancer Res. 2010, 70, 9062–9072. [Google Scholar] [CrossRef] [Green Version]
- 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, 1758834017742575. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Golden, E.B.; Frances, D.; Pellicciotta, I.; Demaria, S.; Helen Barcellos-Hoff, M.; Formenti, S.C. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014, 3, e28518. [Google Scholar] [CrossRef] [Green Version]
- Rubner, Y.; Muth, C.; Strnad, A.; Derer, A.; Sieber, R.; Buslei, R.; Frey, B.; Fietkau, R.; Gaipl, U.S. Fractionated radiotherapy is the main stimulus for the induction of cell death and of Hsp70 release of p53 mutated glioblastoma cell lines. Radiat. Oncol. 2014, 9, 89. [Google Scholar] [CrossRef] [Green Version]
- Bains, S.J.; Abrahamsson, H.; Flatmark, K.; Dueland, S.; Hole, K.H.; Seierstad, T.; Redalen, K.R.; Meltzer, S.; Ree, A.H. Immunogenic cell death by neoadjuvant oxaliplatin and radiation protects against metastatic failure in high-risk rectal cancer. Cancer Immunol. Immunother. 2020, 69, 355–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suzuki, Y.; Mimura, K.; Yoshimoto, Y.; Watanabe, M.; Ohkubo, Y.; Izawa, S.; Murata, K.; Fujii, H.; Nakano, T.; Kono, K. Immunogenic Tumor Cell Death Induced by Chemoradiotherapy in Patients with Esophageal Squamous Cell Carcinoma. Cancer Res. 2012, 72, 3967. [Google Scholar] [CrossRef] [Green Version]
- Gorchs, L.; Hellevik, T.; Bruun, J.A.; Camilio, K.A.; Al-Saad, S.; Stuge, T.B.; Martinez-Zubiaurre, I. Cancer-associated fibroblasts from lung tumors maintain their immunosuppressive abilities after high-dose irradiation. Front. Oncol. 2015, 5, 87. [Google Scholar] [CrossRef] [Green Version]
- Lhuillier, C.; Rudqvist, N.P.; Elemento, O.; Formenti, S.C.; Demaria, S. Radiation therapy and anti-tumor immunity: Exposing immunogenic mutations to the immune system. Genome Med. 2019, 11, 40. [Google Scholar] [CrossRef] [Green Version]
- Spiotto, M.; Fu, Y.X.; Weichselbaum, R.R. The intersection of radiotherapy and immunotherapy: Mechanisms and clinical implications. Sci. Immunol. 2016, 1. [Google Scholar] [CrossRef] [Green Version]
- Garrido, F.; Romero, I.; Aptsiauri, N.; Garcia-Lora, A.M. Generation of MHC class I diversity in primary tumors and selection of the malignant phenotype. Int. J. Cancer 2016, 138, 271–280. [Google Scholar] [CrossRef]
- Dillon, M.T.; Bergerhoff, K.F.; Pedersen, M.; Whittock, H.; Crespo-Rodriguez, E.; Patin, E.C.; Pearson, A.; Smith, H.G.; Paget, J.T.E.; Patel, R.R.; et al. ATR Inhibition Potentiates the Radiation-induced Inflammatory Tumor Microenvironment. Clin. Cancer Res. 2019, 25, 3392–3403. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Lin, W.; Xu, Y.; Chen, X.; Liu, J.; Weng, Y.; Zhuang, Q.; Lin, F.; Huang, Z.; Wu, S.; Ding, J.; et al. Radiation-induced small extracellular vesicles as “carriages” promote tumor antigen release and trigger antitumor immunity. Theranostics 2020, 10, 4871–4884. [Google Scholar] [CrossRef]
- Arnold, K.M.; Flynn, N.J.; Raben, A.; Romak, L.; Yu, Y.; Dicker, A.P.; Mourtada, F.; Sims-Mourtada, J. The Impact of Radiation on the Tumor Microenvironment: Effect of Dose and Fractionation Schedules. Cancer Growth Metastasis 2018, 11, 1179064418761639. [Google Scholar] [CrossRef] [PubMed]
- Lambin, P.; Lieverse, R.I.Y.; Eckert, F.; Marcus, D.; Oberije, C.; van der Wiel, A.M.A.; Guha, C.; Dubois, L.J.; Deasy, J.O. Lymphocyte-Sparing Radiotherapy: The Rationale for Protecting Lymphocyte-rich Organs When Combining Radiotherapy With Immunotherapy. Semin. Radiat. Oncol. 2020, 30, 187–193. [Google Scholar] [CrossRef] [PubMed]
- Davuluri, R.; Jiang, W.; Fang, P.; Xu, C.; Komaki, R.; Gomez, D.R.; Welsh, J.; Cox, J.D.; Crane, C.H.; Hsu, C.C.; et al. Lymphocyte Nadir and Esophageal Cancer Survival Outcomes After Chemoradiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 128–135. [Google Scholar] [CrossRef] [PubMed]
- Falcke, S.E.; Rühle, P.F.; Deloch, L.; Fietkau, R.; Frey, B.; Gaipl, U.S. Clinically Relevant Radiation Exposure Differentially Impacts Forms of Cell Death in Human Cells of the Innate and Adaptive Immune System. Int. J. Mol. Sci. 2018, 19, 3574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, H.; Deng, L.; Hou, Y.; Meng, X.; Huang, X.; Rao, E.; Zheng, W.; Mauceri, H.; Mack, M.; Xu, M.; et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat. Commun. 2017, 8, 1736. [Google Scholar] [CrossRef] [Green Version]
- Heylmann, D.; Ponath, V.; Kindler, T.; Kaina, B. Comparison of DNA repair and radiosensitivity of different blood cell populations. Sci. Rep. 2021, 11, 2478. [Google Scholar] [CrossRef] [PubMed]
- Conrad, S.; Ritter, S.; Fournier, C.; Nixdorff, K. Differential Effects of Irradiation with Carbon Ions and X-Rays on Macrophage Function. J. Radiat. Res. 2009, 50, 223–231. [Google Scholar] [CrossRef]
- Arina, A.; Beckett, M.; Fernandez, C.; Zheng, W.; Pitroda, S.; Chmura, S.J.; Luke, J.J.; Forde, M.; Hou, Y.; Burnette, B.; et al. Tumor-reprogrammed resident T cells resist radiation to control tumors. Nat. Commun. 2019, 10, 3959. [Google Scholar] [CrossRef] [Green Version]
- Galon, J.; Angell, H.K.; Bedognetti, D.; Marincola, F.M. The continuum of cancer immunosurveillance: Prognostic, predictive, and mechanistic signatures. Immunity 2013, 39, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Merrick, A.; Errington, F.; Milward, K.; O’Donnell, D.; Harrington, K.; Bateman, A.; Pandha, H.; Vile, R.; Morrison, E.; Selby, P.; et al. Immunosuppressive effects of radiation on human dendritic cells: Reduced IL-12 production on activation and impairment of naive T-cell priming. Br. J. Cancer 2005, 92, 1450–1458. [Google Scholar] [CrossRef]
- Heylmann, D.; Badura, J.; Becker, H.; Fahrer, J.; Kaina, B. Sensitivity of CD3/CD28-stimulated versus non-stimulated lymphocytes to ionizing radiation and genotoxic anticancer drugs: Key role of ATM in the differential radiation response. Cell Death Dis. 2018, 9, 1053. [Google Scholar] [CrossRef] [PubMed]
- Frey, B.; Rückert, M.; Weber, J.; Mayr, X.; Derer, A.; Lotter, M.; Bert, C.; Rödel, F.; Fietkau, R.; Gaipl, U.S. Hypofractionated Irradiation Has Immune Stimulatory Potential and Induces a Timely Restricted Infiltration of Immune Cells in Colon Cancer Tumors. Front. Immunol. 2017, 8, 231. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Zubiaurre, I.; Chalmers, A.J.; Hellevik, T. Radiation-Induced Transformation of Immunoregulatory Networks in the Tumor Stroma. Front. Immunol. 2018, 9, 1679. [Google Scholar] [CrossRef]
- Sun, G.-Y.; Wang, S.-L.; Song, Y.-W.; Jin, J.; Wang, W.-H.; Liu, Y.-P.; Ren, H.; Fang, H.; Tang, Y.; Zhao, X.-R.; et al. Radiation-Induced Lymphopenia Predicts Poorer Prognosis in Patients With Breast Cancer: A Post Hoc Analysis of a Randomized Controlled Trial of Postmastectomy Hypofractionated Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 277–285. [Google Scholar] [CrossRef]
- Genard, G.; Lucas, S.; Michiels, C. Reprogramming of Tumor-Associated Macrophages with Anticancer Therapies: Radiotherapy versus Chemo- and Immunotherapies. Front. Immunol. 2017, 8, 828. [Google Scholar] [CrossRef] [Green Version]
- Chiang, C.S.; Fu, S.Y.; Wang, S.C.; Yu, C.F.; Chen, F.H.; Lin, C.M.; Hong, J.H. Irradiation promotes an m2 macrophage phenotype in tumor hypoxia. Front. Oncol. 2012, 2, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, H.J.; Griffin, R.J.; Hui, S.; Levitt, S.H.; Song, C.W. Radiation-induced vascular damage in tumors: Implications of vascular damage in ablative hypofractionated radiotherapy (SBRT and SRS). Radiat. Res. 2012, 177, 311–327. [Google Scholar] [CrossRef] [Green Version]
- Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
- Lugade, A.A.; Moran, J.P.; Gerber, S.A.; Rose, R.C.; Frelinger, J.G.; Lord, E.M. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J. Immunol. 2005, 174, 7516–7523. [Google Scholar] [CrossRef] [Green Version]
- Wood, J.; Yasmin-Karim, S.; Mueller, R.; Viswanathan, A.N.; Ngwa, W. Single Radiotherapy Fraction with Local Anti-CD40 Therapy Generates Effective Abscopal Responses in Mouse Models of Cervical Cancer. Cancers 2020, 12, 1026. [Google Scholar] [CrossRef] [Green Version]
- Cao, M.; Cabrera, R.; Xu, Y.; Liu, C.; Nelson, D. Different radiosensitivity of CD4(+)CD25(+) regulatory T cells and effector T cells to low dose gamma irradiation in vitro. Int. J. Radiat. Biol. 2011, 87, 71–80. [Google Scholar] [CrossRef] [Green Version]
- Deloch, L.; Derer, A.; Hartmann, J.; Frey, B.; Fietkau, R.; Gaipl, U.S. Modern Radiotherapy Concepts and the Impact of Radiation on Immune Activation. Front. Oncol. 2016, 6, 141. [Google Scholar] [CrossRef] [Green Version]
- Tsai, M.H.; Cook, J.A.; Chandramouli, G.V.; DeGraff, W.; Yan, H.; Zhao, S.; Coleman, C.N.; Mitchell, J.B.; Chuang, E.Y. Gene expression profiling of breast, prostate, and glioma cells following single versus fractionated doses of radiation. Cancer Res. 2007, 67, 3845–3852. [Google Scholar] [CrossRef] [Green Version]
- Benci, J.L.; Xu, B.; Qiu, Y.; Wu, T.J.; Dada, H.; Twyman-Saint Victor, C.; Cucolo, L.; Lee, D.S.M.; Pauken, K.E.; Huang, A.C.; et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell 2016, 167, 1540–1554.e1512. [Google Scholar] [CrossRef] [Green Version]
- Schaue, D.; Ratikan, J.A.; Iwamoto, K.S.; McBride, W.H. Maximizing tumor immunity with fractionated radiation. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, 1306–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Ruysscher, D.; Waer, M.; Vandeputte, M.; Aerts, R.; Vantongelen, K.; van der Schueren, E. Changes of lymphocyte subsets after local irradiation for early stage breast cancer and seminoma testis: Long-term increase of activated (HLA-DR+) T cells and decrease of “naïve” (CD4-CD45R) T lymphocytes. Eur. J. Cancer 1992, 28, 1729–1734. [Google Scholar] [CrossRef]
- Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef]
- Pilones, K.A.; Charpentier, M.; Garcia-Martinez, E.; Daviaud, C.; Kraynak, J.; Aryankalayil, J.; Formenti, S.C.; Demaria, S. Radiotherapy Cooperates with IL15 to Induce Antitumor Immune Responses. Cancer Immunol. Res. 2020, 8, 1054–1063. [Google Scholar] [CrossRef]
- Harding, S.M.; Benci, J.L.; Irianto, J.; Discher, D.E.; Minn, A.J.; Greenberg, R.A. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017, 548, 466–470. [Google Scholar] [CrossRef] [Green Version]
- Chon, H.; Yoon, K.; Lee, D.; Kwon, D.H.; Cho, Y.H. Single-fraction versus hypofractionated stereotactic radiosurgery for medium-sized brain metastases of 2.5 to 3 cm. J. Neuro-Oncol. 2019, 145, 49–56. [Google Scholar] [CrossRef]
- Brooks, E.D.; Sun, B.; Zhao, L.; Komaki, R.; Liao, Z.; Jeter, M.; Welsh, J.W.; O’Reilly, M.S.; Gomez, D.R.; Hahn, S.M.; et al. Stereotactic Ablative Radiation Therapy is Highly Safe and Effective for Elderly Patients With Early-stage Non-Small Cell Lung Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2017, 98, 900–907. [Google Scholar] [CrossRef] [Green Version]
- Levy, A.; Rivera, S. 1-week hypofractionated adjuvant whole-breast radiotherapy: Towards a new standard? Lancet 2020, 395, 1588–1589. [Google Scholar] [CrossRef]
- Khalife, M.; Shahid, K.; Dabney, R.S.; Phan, A.T. Stereotactic body radiation therapy and immunotherapy. Clin. Adv. Hematol. Oncol. 2019, 17, 518–523. [Google Scholar]
- Bernstein, M.B.; Krishnan, S.; Hodge, J.W.; Chang, J.Y. Immunotherapy and stereotactic ablative radiotherapy (ISABR): A curative approach? Nat. Rev. Clin. Oncol. 2016, 13, 516–524. [Google Scholar] [CrossRef]
- Trovo, M.; Giaj-Levra, N.; Furlan, C.; Bortolin, M.T.; Muraro, E.; Polesel, J.; Minatel, E.; Tedeschi, R.; Filippi, A.R.; Alongi, F.; et al. Stereotactic body radiation therapy and intensity modulated radiation therapy induce different plasmatic cytokine changes in non-small cell lung cancer patients: A pilot study. Clin. Transl. Oncol. 2016, 18, 1003–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chvetsov, A.V.; Rajendran, J.G.; Zeng, J.; Patel, S.A.; Bowen, S.R.; Kim, E.Y. Theoretical effectiveness of cell survival in fractionated radiotherapy with hypoxia-targeted dose escalation. Med. Phys. 2017, 44, 1975–1982. [Google Scholar] [CrossRef]
- Ko, E.C.; Raben, D.; Formenti, S.C. The Integration of Radiotherapy with Immunotherapy for the Treatment of Non-Small Cell Lung Cancer. Clin. Cancer Res. 2018, 24, 5792–5806. [Google Scholar] [CrossRef] [Green Version]
- Senthi, S.; Lagerwaard, F.J.; Haasbeek, C.J.; Slotman, B.J.; Senan, S. Patterns of disease recurrence after stereotactic ablative radiotherapy for early stage non-small-cell lung cancer: A retrospective analysis. Lancet. Oncol. 2012, 13, 802–809. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Zegers, C.M.L.; Rekers, N.H.; Quaden, D.H.F.; Lieuwes, N.G.; Yaromina, A.; Germeraad, W.T.V.; Wieten, L.; Biessen, E.A.L.; Boon, L.; Neri, D.; et al. Radiotherapy Combined with the Immunocytokine L19-IL2 Provides Long-lasting Antitumor Effects. Clin. Cancer Res. 2015, 21, 1151–1160. [Google Scholar] [CrossRef] [Green Version]
- Rekers, N.H.; Zegers, C.M.L.; Germeraad, W.T.V.; Dubois, L.; Lambin, P. Long-lasting antitumor effects provided by radiotherapy combined with the immunocytokine L19-IL2. Oncoimmunology 2015, 4, e1021541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alfonso, J.C.L.; Papaxenopoulou, L.A.; Mascheroni, P.; Meyer-Hermann, M.; Hatzikirou, H. On the Immunological Consequences of Conventionally Fractionated Radiotherapy. Iscience 2020, 23, 100897. [Google Scholar] [CrossRef] [Green Version]
- Hettich, M.; Lahoti, J.; Prasad, S.; Niedermann, G. Checkpoint Antibodies but not T Cell–Recruiting Diabodies Effectively Synergize with TIL-Inducing γ-Irradiation. Cancer Res. 2016, 76, 4673–4683. [Google Scholar] [CrossRef] [Green Version]
- Young, K.H.; Baird, J.R.; Savage, T.; Cottam, B.; Friedman, D.; Bambina, S.; Messenheimer, D.J.; Fox, B.; Newell, P.; Bahjat, K.S.; et al. Optimizing Timing of Immunotherapy Improves Control of Tumors by Hypofractionated Radiation Therapy. PLoS ONE 2016, 11, e0157164. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Lieverse, R.I.Y.; Van Limbergen, E.J.; Oberije, C.J.G.; Troost, E.G.C.; Hadrup, S.R.; Dingemans, A.C.; Hendriks, L.E.L.; Eckert, F.; Hiley, C.; Dooms, C.; et al. Stereotactic ablative body radiotherapy (SABR) combined with immunotherapy (L19-IL2) versus standard of care in stage IV NSCLC patients, ImmunoSABR: A multicentre, randomised controlled open-label phase II trial. BMC Cancer 2020, 20, 557. [Google Scholar] [CrossRef]
- Perez, C.R.; De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat. Commun. 2019, 10, 5408. [Google Scholar] [CrossRef]
- Boudreau, J.E.; Bonehill, A.; Thielemans, K.; Wan, Y. Engineering dendritic cells to enhance cancer immunotherapy. Mol. Ther. 2011, 19, 841–853. [Google Scholar] [CrossRef] [Green Version]
- Vandenberk, L.; Garg, A.D.; Verschuere, T.; Koks, C.; Belmans, J.; Beullens, M.; Agostinis, P.; De Vleeschouwer, S.; Van Gool, S.W. Irradiation of necrotic cancer cells, employed for pulsing dendritic cells (DCs), potentiates DC vaccine-induced antitumor immunity against high-grade glioma. Oncoimmunology 2016, 5, e1083669. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-Otero, P.; Paiva, B.; Engelhardt, M.; Prósper, F.; San Miguel, J.F. Is immunotherapy here to stay in multiple myeloma? Haematologica 2017, 102, 423–432. [Google Scholar] [CrossRef] [Green Version]
- Jongbloed, S.L.; Kassianos, A.J.; McDonald, K.J.; Clark, G.J.; Ju, X.; Angel, C.E.; Chen, C.J.; Dunbar, P.R.; Wadley, R.B.; Jeet, V.; et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J. Exp. Med. 2010, 207, 1247–1260. [Google Scholar] [CrossRef] [Green Version]
- Rapp, M.; Grauer, O.M.; Kamp, M.; Sevens, N.; Zotz, N.; Sabel, M.; Sorg, R.V. A randomized controlled phase II trial of vaccination with lysate-loaded, mature dendritic cells integrated into standard radiochemotherapy of newly diagnosed glioblastoma (GlioVax): Study protocol for a randomized controlled trial. Trials 2018, 19, 293. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-Ruiz, M.; Perez-Gracia, J.; Rodríguez, I.; Alfaro, C.; Oñate, C.; Pérez, G.; Gil-Bazo, I.; Benito, A.; Inogés, S.; De Cerio, A.L.-D.; et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann. Oncol. 2018, 29, 1312–1319. [Google Scholar] [CrossRef] [PubMed]
- Finkelstein, S.E.; Iclozan, C.; Bui, M.M.; Cotter, M.J.; Ramakrishnan, R.; Ahmed, J.; Noyes, D.R.; Cheong, D.; Gonzalez, R.J.; Heysek, R.V.; et al. Combination of external beam radiotherapy (EBRT) with intratumoral injection of dendritic cells as neo-adjuvant treatment of high-risk soft tissue sarcoma patients. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 924–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, J.; Demaria, S.; Formenti, S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J. Immunother. Cancer 2016, 4, 51. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, S.A.; Restifo, N.P.; Yang, J.C.; Morgan, R.A.; Dudley, M.E. Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat. Rev. Cancer 2008, 8, 299–308. [Google Scholar] [CrossRef]
- De Groot, R.; Van Loenen, M.M.; Guislain, A.; Nicolet, B.P.; Freen-Van Heeren, J.J.; Verhagen, O.; Van Den Heuvel, M.M.; De Jong, J.; Burger, P.; Van Der Schoot, C.E.; et al. Polyfunctional tumor-reactive T cells are effectively expanded from non-small cell lung cancers, and correlate with an immune-engaged T cell profile. Oncoimmunology 2019, 8, e1648170. [Google Scholar] [CrossRef]
- Hinrichs, C.S.; Borman, Z.A.; Cassard, L.; Gattinoni, L.; Spolski, R.; Yu, Z.; Sanchez-Perez, L.; Muranski, P.; Kern, S.J.; Logun, C.; et al. Adoptively transferred effector cells derived from naïve rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc. Natl. Acad. Sci. USA 2009, 106, 17469–17474. [Google Scholar] [CrossRef] [Green Version]
- Lai, J.Z.; Zhu, Y.Y.; Ruan, M.; Chen, L.; Zhang, Q.Y. Local Irradiation Sensitized Tumors to Adoptive T Cell Therapy via Enhancing the Cross-Priming, Homing, and Cytotoxicity of Antigen-Specific CD8 T Cells. Front. Immunol. 2019, 10, 2857. [Google Scholar] [CrossRef] [Green Version]
- Ménager, J.; Gorin, J.B.; Maurel, C.; Drujont, L.; Gouard, S.; Louvet, C.; Chérel, M.; Faivre-Chauvet, A.; Morgenstern, A.; Bruchertseifer, F.; et al. Combining α-Radioimmunotherapy and Adoptive T Cell Therapy to Potentiate Tumor Destruction. PLoS ONE 2015, 10, e0130249. [Google Scholar] [CrossRef]
- Wei, S.; Egenti, M.U.; Teitz-Tennenbaum, S.; Zou, W.; Chang, A.E. Effects of tumor irradiation on host T-regulatory cells and systemic immunity in the context of adoptive T-cell therapy in mice. J. Immunother. 2013, 36, 124–132. [Google Scholar] [CrossRef] [Green Version]
- Jin, L.; Tao, H.; Karachi, A.; Long, Y.; Hou, A.Y.; Na, M.; Dyson, K.A.; Grippin, A.J.; Deleyrolle, L.P.; Zhang, W.; et al. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat. Commun. 2019, 10, 4016. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.W.; Jeong, J.U.; Lee, K.H.; Uong, T.N.T.; Rhee, J.H.; Ahn, S.J.; Kim, S.K.; Cho, D.; Quang Nguyen, H.P.; Pham, C.T.; et al. Combined NK Cell Therapy and Radiation Therapy Exhibit Long-Term Therapeutic and Antimetastatic Effects in a Human Triple Negative Breast Cancer Model. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 115–125. [Google Scholar] [CrossRef]
- DeSelm, C.; Palomba, M.L.; Yahalom, J.; Hamieh, M.; Eyquem, J.; Rajasekhar, V.K.; Sadelain, M. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape. Mol. Ther. 2018, 26, 2542–2552. [Google Scholar] [CrossRef] [Green Version]
- Sim, A.J.; Jain, M.D.; Figura, N.B.; Chavez, J.C.; Shah, B.D.; Khimani, F.; Lazaryan, A.; Krivenko, G.; Davila, M.L.; Liu, H.D.; et al. Radiation Therapy as a Bridging Strategy for CAR T Cell Therapy With Axicabtagene Ciloleucel in Diffuse Large B-Cell Lymphoma. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 1012–1021. [Google Scholar] [CrossRef]
- Vascotto, F.; Petschenka, J.; Walzer, K.C.; Vormehr, M.; Brkic, M.; Strobl, S.; Rösemann, R.; Diken, M.; Kreiter, S.; Türeci, Ö.; et al. Intravenous delivery of the toll-like receptor 7 agonist SC1 confers tumor control by inducing a CD8+ T cell response. Oncoimmunology 2019, 8, 1601480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larkin, B.; Ilyukha, V.; Sorokin, M.; Buzdin, A.; Vannier, E.; Poltorak, A. Cutting Edge: Activation of STING in T Cells Induces Type I IFN Responses and Cell Death. J. Immunol. 2017, 199, 397–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knee, D.A.; Hewes, B.; Brogdon, J.L. Rationale for anti-GITR cancer immunotherapy. Eur. J. Cancer 2016, 67, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Von Karstedt, S.; Montinaro, A.; Walczak, H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat. Rev. Cancer 2017, 17, 352–366. [Google Scholar] [CrossRef]
- Cohen, A.D.; Diab, A.; Perales, M.A.; Wolchok, J.D.; Rizzuto, G.; Merghoub, T.; Huggins, D.; Liu, C.; Turk, M.J.; Restifo, N.P.; et al. Agonist anti-GITR antibody enhances vaccine-induced CD8(+) T-cell responses and tumor immunity. Cancer Res. 2006, 66, 4904–4912. [Google Scholar] [CrossRef] [Green Version]
- Fu, J.; Kanne, D.B.; Leong, M.; Glickman, L.H.; McWhirter, S.M.; Lemmens, E.; Mechette, K.; Leong, J.J.; Lauer, P.; Liu, W.; et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci. Transl. Med. 2015, 7, 283ra252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dewan, M.Z.; Vanpouille-Box, C.; Kawashima, N.; DiNapoli, S.; Babb, J.S.; Formenti, S.C.; Adams, S.; Demaria, S. Synergy of topical toll-like receptor 7 agonist with radiation and low-dose cyclophosphamide in a mouse model of cutaneous breast cancer. Clin. Cancer Res. 2012, 18, 6668–6678. [Google Scholar] [CrossRef] [Green Version]
- Dovedi, S.J.; Adlard, A.L.; Ota, Y.; Murata, M.; Sugaru, E.; Koga-Yamakawa, E.; Eguchi, K.; Hirose, Y.; Yamamoto, S.; Umehara, H.; et al. Intravenous administration of the selective toll-like receptor 7 agonist DSR-29133 leads to anti-tumor efficacy in murine solid tumor models which can be potentiated by combination with fractionated radiotherapy. Oncotarget 2016, 7, 17035–17046. [Google Scholar] [CrossRef] [Green Version]
- Yokouchi, H.; Yamazaki, K.; Chamoto, K.; Kikuchi, E.; Shinagawa, N.; Oizumi, S.; Hommura, F.; Nishimura, T.; Nishimura, M. Anti-OX40 monoclonal antibody therapy in combination with radiotherapy results in therapeutic antitumor immunity to murine lung cancer. Cancer Sci. 2008, 99, 361–367. [Google Scholar] [CrossRef] [PubMed]
- Mason, K.A.; Neal, R.; Hunter, N.; Ariga, H.; Ang, K.; Milas, L. CpG oligodeoxynucleotides are potent enhancers of radio- and chemoresponses of murine tumors. Radiother. Oncol. 2006, 80, 192–198. [Google Scholar] [CrossRef]
- Monjazeb, A.M.; Kent, M.S.; Grossenbacher, S.K.; Mall, C.; Zamora, A.E.; Mirsoian, A.; Chen, M.; Kol, A.; Shiao, S.L.; Reddy, A.; et al. Blocking Indolamine-2,3-Dioxygenase Rebound Immune Suppression Boosts Antitumor Effects of Radio-Immunotherapy in Murine Models and Spontaneous Canine Malignancies. Clin. Cancer Res. 2016, 22, 4328–4340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meng, Y.; Carpentier, A.F.; Chen, L.; Boisserie, G.; Simon, J.M.; Mazeron, J.J.; Delattre, J.Y. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. Int. J. Cancer 2005, 116, 992–997. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.H.; Gratzinger, D.; Harrison, C.; Brody, J.D.; Czerwinski, D.K.; Ai, W.Z.; Morales, A.; Abdulla, F.; Xing, L.; Navi, D.; et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: A phase 1/2 study. Blood 2012, 119, 355–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brody, J.D.; Ai, W.Z.; Czerwinski, D.K.; Torchia, J.A.; Levy, M.; Advani, R.H.; Kim, Y.H.; Hoppe, R.T.; Knox, S.J.; Shin, L.K.; et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: A phase I/II study. J. Clin. Oncol. 2010, 28, 4324–4332. [Google Scholar] [CrossRef] [PubMed]
- Payne, R.; Glenn, L.; Hoen, H.; Richards, B.; Smith, J.W., 2nd; Lufkin, R.; Crocenzi, T.S.; Urba, W.J.; Curti, B.D. Durable responses and reversible toxicity of high-dose interleukin-2 treatment of melanoma and renal cancer in a Community Hospital Biotherapy Program. J. Immunother. Cancer 2014, 2, 13. [Google Scholar] [CrossRef] [Green Version]
- Conlon, K.C.; Miljkovic, M.D.; Waldmann, T.A. Cytokines in the Treatment of Cancer. J. Interferon Cytokine Res. 2018, 39, 6–21. [Google Scholar] [CrossRef]
- Alwan, L.M.; Grossmann, K.; Sageser, D.; Van Atta, J.; Agarwal, N.; Gilreath, J.A. Comparison of acute toxicity and mortality after two different dosing regimens of high-dose interleukin-2 for patients with metastatic melanoma. Target. Oncol. 2014, 9, 63–71. [Google Scholar] [CrossRef]
- Kontermann, R.E. Antibody-cytokine fusion proteins. Arch. Biochem. Biophys. 2012, 526, 194–205. [Google Scholar] [CrossRef]
- List, T.; Neri, D. Immunocytokines: A review of molecules in clinical development for cancer therapy. Clin. Pharmacol. 2013, 5, 29–45. [Google Scholar] [CrossRef] [Green Version]
- Létourneau, S.; van Leeuwen, E.M.; Krieg, C.; Martin, C.; Pantaleo, G.; Sprent, J.; Surh, C.D.; Boyman, O. IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25. Proc. Natl. Acad. Sci. USA 2010, 107, 2171–2176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mortara, L.; Balza, E.; Bruno, A.; Poggi, A.; Orecchia, P.; Carnemolla, B. Anti-cancer Therapies Employing IL-2 Cytokine Tumor Targeting: Contribution of Innate, Adaptive and Immunosuppressive Cells in the Anti-tumor Efficacy. Front. Immunol. 2018, 9, 2905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lieverse, R.I.Y.; Marcus, D.; van der Wiel, A.M.A.; Van Limbergen, E.J.; Theys, J.; Yaromina, A.; Lambin, P.; Dubois, L.J. Human fibronectin extra domain B as a biomarker for targeted therapy in cancer. Mol. Oncol. 2020, 14, 1555–1568. [Google Scholar] [CrossRef] [PubMed]
- Palata, O.; Hradilova Podzimkova, N.; Nedvedova, E.; Umprecht, A.; Sadilkova, L.; Palova Jelinkova, L.; Spisek, R.; Adkins, I. Radiotherapy in Combination With Cytokine Treatment. Front. Oncol. 2019, 9, 367. [Google Scholar] [CrossRef] [PubMed]
- Van den Heuvel, M.M.; Verheij, M.; Boshuizen, R.; Belderbos, J.; Dingemans, A.M.; De Ruysscher, D.; Laurent, J.; Tighe, R.; Haanen, J.; Quaratino, S. NHS-IL2 combined with radiotherapy: Preclinical rationale and phase Ib trial results in metastatic non-small cell lung cancer following first-line chemotherapy. J. Transl. Med. 2015, 13, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris, Z.S.; Guy, E.I.; Francis, D.M.; Gressett, M.M.; Werner, L.R.; Carmichael, L.L.; Yang, R.K.; Armstrong, E.A.; Huang, S.; Navid, F.; et al. In Situ Tumor Vaccination by Combining Local Radiation and Tumor-Specific Antibody or Immunocytokine Treatments. Cancer Res. 2016, 76, 3929–3941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Voeller, J.; Erbe, A.K.; Slowinski, J.; Rasmussen, K.; Carlson, P.M.; Hoefges, A.; VandenHeuvel, S.; Stuckwisch, A.; Wang, X.; Gillies, S.D.; et al. Combined innate and adaptive immunotherapy overcomes resistance of immunologically cold syngeneic murine neuroblastoma to checkpoint inhibition. J. Immunother. Cancer 2019, 7, 344. [Google Scholar] [CrossRef]
- Kujawski, M.; Sherman, M.; Hui, S.; Zuro, D.; Lee, W.H.; Yazaki, P.; Sherman, A.; Szpikowska, B.; Chea, J.; Lasiewski, D.; et al. Potent immunomodulatory effects of an anti-CEA-IL-2 immunocytokine on tumor therapy and effects of stereotactic radiation. Oncoimmunology 2020, 9, 1724052. [Google Scholar] [CrossRef] [Green Version]
- Rekers, N.H.; Olivo Pimentel, V.; Yaromina, A.; Lieuwes, N.G.; Biemans, R.; Zegers, C.M.L.; Germeraad, W.T.V.; Van Limbergen, E.J.; Neri, D.; Dubois, L.J.; et al. The immunocytokine L19-IL2: An interplay between radiotherapy and long-lasting systemic anti-tumour immune responses. Oncoimmunology 2018, 7, e1414119. [Google Scholar] [CrossRef] [Green Version]
- Van Limbergen, E.J.; Hoeben, A.; Lieverse, R.; Houben, R.; Overhof, C.; Postma, A.; Zindler, J.; Verhelst, F.; Dubois, L.J.; De Ruysscher, D.; et al. Toxicity of L19-Interleukin 2 combined with Stereotactic Body Radiotherapy: A phase 1 study. Int. J. Radiat. Oncol. Biol. Phys. 2021, 109, 1421–1430. [Google Scholar] [CrossRef]
- Zorn, E.; Nelson, E.A.; Mohseni, M.; Porcheray, F.; Kim, H.; Litsa, D.; Bellucci, R.; Raderschall, E.; Canning, C.; Soiffer, R.J.; et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006, 108, 1571–1579. [Google Scholar] [CrossRef] [Green Version]
- Fallon, J.; Tighe, R.; Kradjian, G.; Guzman, W.; Bernhardt, A.; Neuteboom, B.; Lan, Y.; Sabzevari, H.; Schlom, J.; Greiner, J.W. The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget 2014, 5, 1869–1884. [Google Scholar] [CrossRef] [Green Version]
- Eckert, F.; Jelas, I.; Oehme, M.; Huber, S.M.; Sonntag, K.; Welker, C.; Gillies, S.D.; Strittmatter, W.; Zips, D.; Handgretinger, R.; et al. Tumor-targeted IL-12 combined with local irradiation leads to systemic tumor control via abscopal effects in vivo. Oncoimmunology 2017, 6, e1323161. [Google Scholar] [CrossRef] [Green Version]
- Sardana, R.; Mishra, S.K.; Williamson, S.R.; Mohanty, A.; Mohanty, S.K. Immune checkpoints and their inhibitors: Reappraisal of a novel diagnostic and therapeutic dimension in the urologic malignancies. Semin. Oncol. 2020, 47, 367–379. [Google Scholar] [CrossRef]
- Vanneste, B.G.L.; Van Limbergen, E.J.; Dubois, L.; Samarska, I.V.; Wieten, L.; Aarts, M.J.B.; Marcelissen, T.; De Ruysscher, D. Immunotherapy as sensitizer for local radiotherapy. Oncoimmunology 2020, 9, 1832760. [Google Scholar] [CrossRef]
- Ullah, M.; Aoudjeghout, W.; Pimpie, C.; Pocard, M.; Mirshahi, M. Mitosis in Cancer Cell Increases Immune Resistance via High Expression of HLA-G and PD-L1. Cancers 2020, 12, 2661. [Google Scholar] [CrossRef]
- Herter-Sprie, G.S.; Koyama, S.; Korideck, H.; Hai, J.; Deng, J.; Li, Y.Y.; Buczkowski, K.A.; Grant, A.K.; Ullas, S.; Rhee, K.; et al. Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer. JCI Insight 2016, 1, e87415. [Google Scholar] [CrossRef] [PubMed]
- Oweida, A.; Lennon, S.; Calame, D.; Korpela, S.; Bhatia, S.; Sharma, J.; Graham, C.; Binder, D.; Serkova, N.; Raben, D.; et al. Ionizing radiation sensitizes tumors to PD-L1 immune checkpoint blockade in orthotopic murine head and neck squamous cell carcinoma. Oncoimmunology 2017, 6, e1356153. [Google Scholar] [CrossRef] [PubMed]
- Zeng, J.; See, A.P.; Phallen, J.; Jackson, C.M.; Belcaid, Z.; Ruzevick, J.; Durham, N.; Meyer, C.; Harris, T.J.; Albesiano, E.; et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 343–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Rudqvist, N.P.; Pilones, K.A.; Lhuillier, C.; Wennerberg, E.; Sidhom, J.W.; Emerson, R.O.; Robins, H.S.; Schneck, J.; Formenti, S.C.; Demaria, S. Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells. Cancer Immunol. Res. 2018, 6, 139–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Son, C.H.; Bae, J.H.; Shin, D.Y.; Lee, H.R.; Choi, Y.J.; Jo, W.S.; Ho Jung, M.; Kang, C.D.; Yang, K.; Park, Y.S. CTLA-4 blockade enhances antitumor immunity of intratumoral injection of immature dendritic cells into irradiated tumor in a mouse colon cancer model. J. Immunother. 2014, 37, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Formenti, S.C.; Rudqvist, N.P.; Golden, E.; Cooper, B.; Wennerberg, E.; Lhuillier, C.; Vanpouille-Box, C.; Friedman, K.; Ferrari de Andrade, L.; Wucherpfennig, K.W.; et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat. Med. 2018, 24, 1845–1851. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, C.; Welsh, J.W.; De Groot, P.; Massarelli, E.; Chang, J.Y.; Hess, K.R.; Basu, S.; Curran, M.A.; Cabanillas, M.E.; Subbiah, V.; et al. Ipilimumab with Stereotactic Ablative Radiation Therapy: Phase I Results and Immunologic Correlates from Peripheral T Cells. Clinical Cancer Res. 2017, 23, 1388–1396. [Google Scholar] [CrossRef] [Green Version]
- Roger, A.; Finet, A.; Boru, B.; Beauchet, A.; Mazeron, J.J.; Otzmeguine, Y.; Blom, A.; Longvert, C.; de Maleissye, M.F.; Fort, M.; et al. Efficacy of combined hypo-fractionated radiotherapy and anti-PD-1 monotherapy in difficult-to-treat advanced melanoma patients. Oncoimmunology 2018, 7, e1442166. [Google Scholar] [CrossRef]
- Luke, J.J.; Lemons, J.M.; Karrison, T.G.; Pitroda, S.P.; Melotek, J.M.; Zha, Y.; Al-Hallaq, H.A.; Arina, A.; Khodarev, N.N.; Janisch, L.; et al. Safety and Clinical Activity of Pembrolizumab and Multisite Stereotactic Body Radiotherapy in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2018, 36, 1611–1618. [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]
- Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef]
- Hui, R.; Özgüroğlu, M.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Yokoi, T.; Chiappori, A.; Lee, K.H.; De Wit, M.; et al. Patient-reported outcomes with durvalumab after chemoradiotherapy in stage III, unresectable non-small-cell lung cancer (PACIFIC): A randomised, controlled, phase 3 study. Lancet Oncol. 2019, 20, 1670–1680. [Google Scholar] [CrossRef]
- Watanabe, T.; Gaedicke, S.; Guffart, E.; Firat, E.; Niedermann, G. Adding Indoximod to Hypofractionated Radiotherapy with Anti-PD-1 Checkpoint Blockade Enhances Early NK and CD8(+) T-Cell-Dependent Tumor Activity. Clin. Cancer Res. 2020, 26, 945–956. [Google Scholar] [CrossRef] [Green Version]
- Cohen, E.E.; Ferris, R.L.; Psyrri, A.; Haddad, R.; Tahara, M.; Bourhis, J.; Harrington, K.J.; Chang, P.M.H.; Lin, J.C.; Razaq, M.; et al. 910O Primary results of the phase III JAVELIN head & neck 100 trial: Avelumab plus chemoradiotherapy (CRT) followed by avelumab maintenance vs CRT in patients with locally advanced squamous cell carcinoma of the head and neck (LA SCCHN). Ann. Oncol. 2020, 31, S658. [Google Scholar] [CrossRef]
- Bourhis, J.; Sire, C.; Tao, Y.; Martin, L.; Alfonsi, M.; Prevost, J.B.; Rives, M.; Lafond, C.; Tourani, J.M.; Biau, J.; et al. LBA38 Pembrolizumab versus cetuximab, concomitant with radiotherapy (RT) in locally advanced head and neck squamous cell carcinoma (LA-HNSCC): Results of the GORTEC 2015-01 “PembroRad” randomized trial. Ann. Oncol. 2020, 31, S1168. [Google Scholar] [CrossRef]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
- Qin, S.; Xu, L.; Yi, M.; Yu, S.; Wu, K.; Luo, S. Novel immune checkpoint targets: Moving beyond PD-1 and CTLA-4. Mol. Cancer 2019, 18, 155. [Google Scholar] [CrossRef] [PubMed]
- Poggio, M.; Hu, T.; Pai, C.C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell 2019, 177, 414–427.e413. [Google Scholar] [CrossRef] [Green Version]
- Moynihan, K.D.; Opel, C.F.; Szeto, G.L.; Tzeng, A.; Zhu, E.F.; Engreitz, J.M.; Williams, R.T.; Rakhra, K.; Zhang, M.H.; Rothschilds, A.M.; et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat. Med. 2016, 22, 1402–1410. [Google Scholar] [CrossRef]
- Olivo Pimentel, V.; Marcus, D.; van der Wiel, A.M.; Lieuwes, N.G.; Biemans, R.; Lieverse, R.I.; Neri, D.; Theys, J.; Yaromina, A.; Dubois, L.J.; et al. Releasing the brakes of tumor immunity with anti-PD-L1 and pushing its accelerator with L19-IL2 cures poorly immunogenic tumors when combined with radiotherapy. J. Immunother. Cancer 2021, 9. [Google Scholar] [CrossRef]
- Bai, R.; Lv, Z.; Xu, D.; Cui, J. Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark. Res. 2020, 8, 34. [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, 19, 1180–1191. [Google Scholar] [CrossRef]
- Jang, B.S.; Kim, I.A. A radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with invasive breast carcinoma in The Cancer Genome Atlas (TCGA) dataset. Radiother. Oncol. 2017, 124, 403–410. [Google Scholar] [CrossRef] [PubMed]
- Debus, C.; Afshar-Oromieh, A.; Floca, R.; Ingrisch, M.; Knoll, M.; Debus, J.; Haberkorn, U.; Abdollahi, A. Feasibility and robustness of dynamic (18)F-FET PET based tracer kinetic models applied to patients with recurrent high-grade glioma prior to carbon ion irradiation. Sci. Rep. 2018, 8, 14760. [Google Scholar] [CrossRef]
- Debus, C.; Waltenberger, M.; Floca, R.; Afshar-Oromieh, A.; Bougatf, N.; Adeberg, S.; Heiland, S.; Bendszus, M.; Wick, W.; Rieken, S.; et al. Impact of (18)F-FET PET on Target Volume Definition and Tumor Progression of Recurrent High Grade Glioma Treated with Carbon-Ion Radiotherapy. Sci. Rep. 2018, 8, 7201. [Google Scholar] [CrossRef]
- Zhou, C.; Jones, B.; Moustafa, M.; Schwager, C.; Bauer, J.; Yang, B.; Cao, L.; Jia, M.; Mairani, A.; Chen, M.; et al. Quantitative assessment of radiation dose and fractionation effects on normal tissue by utilizing a novel lung fibrosis index model. Radiat. Oncol. 2017, 12, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, C.; Jones, B.; Moustafa, M.; Yang, B.; Brons, S.; Cao, L.; Dai, Y.; Schwager, C.; Chen, M.; Jaekel, O.; et al. Determining RBE for development of lung fibrosis induced by fractionated irradiation with carbon ions utilizing fibrosis index and high-LET BED model. Clin. Transl. Radiat. Oncol. 2019, 14, 25–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, C.; Moustafa, M.R.; Cao, L.; Kriegsmann, M.; Winter, M.; Schwager, C.; Jones, B.; Wang, S.; Bauerle, T.; Zhou, P.K.; et al. Modeling and multiscale characterization of the quantitative imaging based fibrosis index reveals pathophysiological, transcriptome and proteomic correlates of lung fibrosis induced by fractionated irradiation. Int. J. Cancer 2019, 144, 3160–3173. [Google Scholar] [CrossRef] [Green Version]
- Nowrouzi, A.; Sertorio, M.G.; Akbarpour, M.; Knoll, M.; Krunic, D.; Kuhar, M.; Schwager, C.; Brons, S.; Debus, J.; Wells, S.I.; et al. Personalized Assessment of Normal Tissue Radiosensitivity via Transcriptome Response to Photon, Proton and Carbon Irradiation in Patient-Derived Human Intestinal Organoids. Cancers 2020, 12, 469. [Google Scholar] [CrossRef] [Green Version]
- Mohamad, O.; Sishc, B.J.; Saha, J.; Pompos, A.; Rahimi, A.; Story, M.D.; Davis, A.J.; Kim, D.W.N. Carbon Ion Radiotherapy: A Review of Clinical Experiences and Preclinical Research, with an Emphasis on DNA Damage/Repair. Cancers 2017, 9, 66. [Google Scholar] [CrossRef] [Green Version]
- Mein, S.; Klein, C.; Kopp, B.; Magro, G.; Harrabi, S.; Karger, C.P.; Haberer, T.; Debus, J.; Abdollahi, A.; Dokic, I.; et al. Assessment of RBE-Weighted Dose Models for Carbon Ion Therapy Toward Modernization of Clinical Practice at HIT: In Vitro, in Vivo, and in Patients. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 779–791. [Google Scholar] [CrossRef]
- Bronk, L.; Guan, F.; Patel, D.; Ma, D.; Kroger, B.; Wang, X.; Tran, K.; Yiu, J.; Stephan, C.; Debus, J.; et al. Mapping the Relative Biological Effectiveness of Proton, Helium and Carbon Ions with High-Throughput Techniques. Cancers 2020, 12, 3658. [Google Scholar] [CrossRef]
- Winter, M.; Dokic, I.; Schlegel, J.; Warnken, U.; Debus, J.; Abdollahi, A.; Schnolzer, M. Deciphering the Acute Cellular Phosphoproteome Response to Irradiation with X-rays, Protons and Carbon Ions. Mol. Cell Proteom. 2017, 16, 855–872. [Google Scholar] [CrossRef] [Green Version]
- Dai, Y.; Wei, Q.; Schwager, C.; Hanne, J.; Zhou, C.; Herfarth, K.; Rieken, S.; Lipson, K.E.; Debus, J.; Abdollahi, A. Oncogene addiction and radiation oncology: Effect of radiotherapy with photons and carbon ions in ALK-EML4 translocated NSCLC. Radiat. Oncol. 2018, 13, 1. [Google Scholar] [CrossRef]
- Dai, Y.; Wei, Q.; Schwager, C.; Moustafa, M.; Zhou, C.; Lipson, K.E.; Weichert, W.; Debus, J.; Abdollahi, A. Synergistic effects of crizotinib and radiotherapy in experimental EML4-ALK fusion positive lung cancer. Radiother. Oncol. 2015, 114, 173–181. [Google Scholar] [CrossRef] [PubMed]
- Ando, K.; Koike, S.; Uzawa, A.; Takai, N.; Fukawa, T.; Furusawa, Y.; Aoki, M.; Miyato, Y. Biological gain of carbon-ion radiotherapy for the early response of tumor growth delay and against early response of skin reaction in mice. J. Radiat. Res. 2005, 46, 51–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shirai, K.; Kubota, Y.; Ohno, T.; Saitoh, J.I.; Abe, T.; Mizukami, T.; Mori, Y.; Kawamura, H.; Akahane, K.; Nakano, T. Carbon-ion Radiotherapy for Isolated Lymph Node Metastasis After Surgery or Radiotherapy for Lung Cancer. Front. Oncol. 2019, 9, 731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sato, K.; Shimokawa, T.; Imai, T. Difference in Acquired Radioresistance Induction Between Repeated Photon and Particle Irradiation. Front. Oncol. 2019, 9, 1213. [Google Scholar] [CrossRef] [PubMed]
- Tommasino, F.; Durante, M. Proton radiobiology. Cancers 2015, 7, 353–381. [Google Scholar] [CrossRef]
- Tinganelli, W.; Durante, M. Carbon Ion Radiobiology. Cancers 2020, 12, 3022. [Google Scholar] [CrossRef]
- Durante, M.; Loeffler, J.S. Charged particles in radiation oncology. Nat. Rev. Clin. Oncol. 2010, 7, 37–43. [Google Scholar] [CrossRef] [PubMed]
- Terasawa, T.; Dvorak, T.; Ip, S.; Raman, G.; Lau, J.; Trikalinos, T.A. Systematic review: Charged-particle radiation therapy for cancer. Ann. Intern. Med. 2009, 151, 556–565. [Google Scholar] [CrossRef] [Green Version]
- Debus, J.; Abdollahi, A. For the next trick: New discoveries in radiobiology applied to glioblastoma. Am. Soc. Clin. Oncol. Educ. Book 2014, e95–e99. [Google Scholar] [CrossRef] [Green Version]
- Onishi, M.; Okonogi, N.; Oike, T.; Yoshimoto, Y.; Sato, H.; Suzuki, Y.; Kamada, T.; Nakano, T. High linear energy transfer carbon-ion irradiation increases the release of the immune mediator high mobility group box 1 from human cancer cells. J. Radiat. Res. 2018, 59, 541–546. [Google Scholar] [CrossRef] [Green Version]
- Murshed, H. Fundamentals of Radiation Oncology, 3rd ed.; Elsevier Science: San Diego, CA, USA, 2019; p. 746. [Google Scholar]
- Yoshimoto, Y.; Kono, K.; Suzuki, Y. Anti-tumor immune responses induced by radiotherapy: A review. Fukushima J. Med. Sci. 2015, 61, 13–22. [Google Scholar] [CrossRef] [Green Version]
- Ando, K.; Fujita, H.; Hosoi, A.; Ma, L.; Wakatsuki, M.; Seino, K.-i.; Kakimi, K.; Imai, T.; Shimokawa, T.; Nakano, T. Intravenous dendritic cell administration enhances suppression of lung metastasis induced by carbon-ion irradiation. J. Radiat. Res. 2017, 58, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed] [Green Version]
- Huang, Y.; Dong, Y.; Zhao, J.; Zhang, L.; Kong, L.; Lu, J.J. Comparison of the effects of photon, proton and carbon-ion radiation on the ecto-calreticulin exposure in various tumor cell lines. Ann. Transl. Med. 2019, 7, 542. [Google Scholar] [CrossRef]
- Garg, A.D.; De Ruysscher, D.; Agostinis, P. Immunological metagene signatures derived from immunogenic cancer cell death associate with improved survival of patients with lung, breast or ovarian malignancies: A large-scale meta-analysis. Oncoimmunology 2016, 5, e1069938. [Google Scholar] [CrossRef] [Green Version]
- Bao, C.; Sun, Y.; Dwarakanath, B.; Dong, Y.; Huang, Y.; Wu, X.; Guha, C.; Kong, L.; Lu, J.J. Carbon ion triggered immunogenic necroptosis of nasopharyngeal carcinoma cells involving necroptotic inhibitor BCL-x. J. Cancer 2021, 12, 1520–1530. [Google Scholar] [CrossRef]
- Alan Mitteer, R.; Wang, Y.; Shah, J.; Gordon, S.; Fager, M.; Butter, P.P.; Jun Kim, H.; Guardiola-Salmeron, C.; Carabe-Fernandez, A.; Fan, Y. Proton beam radiation induces DNA damage and cell apoptosis in glioma stem cells through reactive oxygen species. Sci. Rep. 2015, 5, 13961. [Google Scholar] [CrossRef] [Green Version]
- Georgakilas, A.G.; O’Neill, P.; Stewart, R.D. Induction and repair of clustered DNA lesions: What do we know so far? Radiat. Res. 2013, 180, 100–109. [Google Scholar] [CrossRef]
- Oonishi, K.; Cui, X.; Hirakawa, H.; Fujimori, A.; Kamijo, T.; Yamada, S.; Yokosuka, O.; Kamada, T. Different effects of carbon ion beams and X-rays on clonogenic survival and DNA repair in human pancreatic cancer stem-like cells. Radiother. Oncol. 2012, 105, 258–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morishita, M.; Muramatsu, T.; Suto, Y.; Hirai, M.; Konishi, T.; Hayashi, S.; Shigemizu, D.; Tsunoda, T.; Moriyama, K.; Inazawa, J. Chromothripsis-like chromosomal rearrangements induced by ionizing radiation using proton microbeam irradiation system. Oncotarget 2016, 7, 10182–10192. [Google Scholar] [CrossRef] [PubMed]
- Rode, A.; Maass, K.K.; Willmund, K.V.; Lichter, P.; Ernst, A. Chromothripsis in cancer cells: An update. Int. J. Cancer 2016, 138, 2322–2333. [Google Scholar] [CrossRef] [PubMed]
- Helm, A.; Lee, R.; Durante, M.; Ritter, S. The Influence of C-Ions and X-rays on Human Umbilical Vein Endothelial Cells. Front. Oncol. 2016, 6, 5. [Google Scholar] [CrossRef] [Green Version]
- Litvinchuk, A.V.; Vachelová, J.; Michaelidesová, A.; Wagner, R.; Davídková, M. Dose-dependent micronuclei formation in normal human fibroblasts exposed to proton radiation. Radiat. Environ. Biophys. 2015, 54, 327–334. [Google Scholar] [CrossRef]
- Autsavapromporn, N.; Plante, I.; Liu, C.; Konishi, T.; Usami, N.; Funayama, T.; Azzam, E.I.; Murakami, T.; Suzuki, M. Genetic changes in progeny of bystander human fibroblasts after microbeam irradiation with X-rays, protons or carbon ions: The relevance to cancer risk. Int. J. Radiat. Biol. 2015, 91, 62–70. [Google Scholar] [CrossRef]
- Carter, R.J.; Nickson, C.M.; Thompson, J.M.; Kacperek, A.; Hill, M.A.; Parsons, J.L. Complex DNA Damage Induced by High Linear Energy Transfer Alpha-Particles and Protons Triggers a Specific Cellular DNA Damage Response. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 776–784. [Google Scholar] [CrossRef] [Green Version]
- Tarcic, O.; Pateras, I.S.; Cooks, T.; Shema, E.; Kanterman, J.; Ashkenazi, H.; Boocholez, H.; Hubert, A.; Rotkopf, R.; Baniyash, M.; et al. RNF20 Links Histone H2B Ubiquitylation with Inflammation and Inflammation-Associated Cancer. Cell Rep. 2016, 14, 1462–1476. [Google Scholar] [CrossRef] [Green Version]
- Melzig, C.; Golestaneh, A.F.; Mier, W.; Schwager, C.; Das, S.; Schlegel, J.; Lasitschka, F.; Kauczor, H.U.; Debus, J.; Haberkorn, U.; et al. Combined external beam radiotherapy with carbon ions and tumor targeting endoradiotherapy. Oncotarget 2018, 9, 29985–30004. [Google Scholar] [CrossRef] [Green Version]
- Chiblak, S.; Tang, Z.; Campos, B.; Gal, Z.; Unterberg, A.; Debus, J.; Herold-Mende, C.; Abdollahi, A. Radiosensitivity of Patient-Derived Glioma Stem Cell 3-Dimensional Cultures to Photon, Proton, and Carbon Irradiation. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 112–119. [Google Scholar] [CrossRef]
- Lupu-Plesu, M.; Claren, A.; Martial, S.; N’Diaye, P.D.; Lebrigand, K.; Pons, N.; Ambrosetti, D.; Peyrottes, I.; Feuillade, J.; Hérault, J.; et al. Effects of proton versus photon irradiation on (lymph)angiogenic, inflammatory, proliferative and anti-tumor immune responses in head and neck squamous cell carcinoma. Oncogenesis 2017, 6, e354. [Google Scholar] [CrossRef]
- Nielsen, S.; Bassler, N.; Grzanka, L.; Swakon, J.; Olko, P.; Horsman, M.R.; Sørensen, B.S. Proton scanning and X-ray beam irradiation induce distinct regulation of inflammatory cytokines in a preclinical mouse model. Int. J. Radiat. Biol. 2020, 96, 1238–1244. [Google Scholar] [CrossRef]
- Nielsen, S.; Bassler, N.; Grzanka, L.; Laursen, L.; Swakon, J.; Olko, P.; Andreassen, C.N.; Alsner, J.; Singers Sørensen, B. Comparison of Coding Transcriptomes in Fibroblasts Irradiated With Low and High LET Proton Beams and Cobalt-60 Photons. Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 1203–1211. [Google Scholar] [CrossRef]
- Nielsen, S.; Bassler, N.; Grzanka, L.; Swakon, J.; Olko, P.; Andreassen, C.N.; Overgaard, J.; Alsner, J.; Sørensen, B.S. Differential gene expression in primary fibroblasts induced by proton and cobalt-60 beam irradiation. Acta Oncol. 2017, 56, 1406–1412. [Google Scholar] [CrossRef]
- Chouaib, S.; Umansky, V.; Kieda, C. The role of hypoxia in shaping the recruitment of proangiogenic and immunosuppressive cells in the tumor microenvironment. Contemp. Oncol. 2018, 22, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Labiano, S.; Palazon, A.; Melero, I. Immune response regulation in the tumor microenvironment by hypoxia. Semin. Oncol. 2015, 42, 378–386. [Google Scholar] [CrossRef]
- Park, J.E.; Dutta, B.; Tse, S.W.; Gupta, N.; Tan, C.F.; Low, J.K.; Yeoh, K.W.; Kon, O.L.; Tam, J.P.; Sze, S.K. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 2019, 38, 5158–5173. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Hasmim, M.; Messai, Y.; Terry, S.; Kieda, C.; Janji, B.; Chouaib, S. Hypoxia: A key player in antitumor immune response. A Review in the Theme: Cellular Responses to Hypoxia. Am. J. Physiol. Cell Physiol. 2015, 309, C569–C579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barsoum, I.B.; Smallwood, C.A.; Siemens, D.R.; Graham, C.H. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014, 74, 665–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed]
- Subtil, F.S.; Wilhelm, J.; Bill, V.; Westholt, N.; Rudolph, S.; Fischer, J.; Scheel, S.; Seay, U.; Fournier, C.; Taucher-Scholz, G.; et al. Carbon ion radiotherapy of human lung cancer attenuates HIF-1 signaling and acts with considerably enhanced therapeutic efficiency. FASEB J. 2014, 28, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
- Wozny, A.S.; Lauret, A.; Battiston-Montagne, P.; Guy, J.B.; Beuve, M.; Cunha, M.; Saintigny, Y.; Blond, E.; Magne, N.; Lalle, P.; et al. Differential pattern of HIF-1α expression in HNSCC cancer stem cells after carbon ion or photon irradiation: One molecular explanation of the oxygen effect. Br. J. Cancer 2017, 116, 1340–1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klein, C.; Dokic, I.; Mairani, A.; Mein, S.; Brons, S.; Haring, P.; Haberer, T.; Jakel, O.; Zimmermann, A.; Zenke, F.; et al. Overcoming hypoxia-induced tumor radioresistance in non-small cell lung cancer by targeting DNA-dependent protein kinase in combination with carbon ion irradiation. Radiat. Oncol. 2017, 12, 208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chiblak, S.; Tang, Z.; Lemke, D.; Knoll, M.; Dokic, I.; Warta, R.; Moustafa, M.; Mier, W.; Brons, S.; Rapp, C.; et al. Carbon irradiation overcomes glioma radioresistance by eradicating stem cells and forming an antiangiogenic and immunopermissive niche. JCI Insight 2019, 4, e123837. [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]
- Mohamad, O.; Tabuchi, T.; Nitta, Y.; Nomoto, A.; Sato, A.; Kasuya, G.; Makishima, H.; Choy, H.; Yamada, S.; Morishima, T.; et al. Risk of subsequent primary cancers after carbon ion radiotherapy, photon radiotherapy, or surgery for localised prostate cancer: A propensity score-weighted, retrospective, cohort study. Lancet Oncol. 2019, 20, 674–685. [Google Scholar] [CrossRef]
- Matsunaga, A.; Ueda, Y.; Yamada, S.; Harada, Y.; Shimada, H.; Hasegawa, M.; Tsujii, H.; Ochiai, T.; Yonemitsu, Y. Carbon-ion beam treatment induces systemic antitumor immunity against murine squamous cell carcinoma. Cancer 2010, 116, 3740–3748. [Google Scholar] [CrossRef]
- 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]
- Brenneman, R.J.; Sharifai, N.; Fischer-Valuck, B.; Hassanzadeh, C.; Guzelian, J.; Chrisinger, J.S.A.; Michalski, J.M.; Oppelt, P.; Baumann, B.C. Abscopal Effect Following Proton Beam Radiotherapy in a Patient With Inoperable Metastatic Retroperitoneal Sarcoma. Front. Oncol. 2019, 9, 922. [Google Scholar] [CrossRef]
- Takahashi, Y.; Yasui, T.; Minami, K.; Tamari, K.; Hayashi, K.; Otani, K.; Seo, Y.; Isohashi, F.; Koizumi, M.; Ogawa, K. Carbon ion irradiation enhances the antitumor efficacy of dual immune checkpoint blockade therapy both for local and distant sites in murine osteosarcoma. Oncotarget 2019, 10, 633–646. [Google Scholar] [CrossRef] [Green Version]
- Shimokawa, T.; Ma, L.; Ando, K.; Sato, K.; Imai, T. The Future of Combining Carbon-Ion Radiotherapy with Immunotherapy: Evidence and Progress in Mouse Models. Int. J. Part. Ther. 2016, 3, 61–70. [Google Scholar] [CrossRef] [Green Version]
- Ohkubo, Y.; Iwakawa, M.; Seino, K.; Nakawatari, M.; Wada, H.; Kamijuku, H.; Nakamura, E.; Nakano, T.; Imai, T. Combining carbon ion radiotherapy and local injection of α-galactosylceramide-pulsed dendritic cells inhibits lung metastases in an in vivo murine model. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 1524–1531. [Google Scholar] [CrossRef]
- Helm, A.; Tinganelli, W.; Simoniello, P.; Kurosawa, F.; Fournier, C.; Shimokawa, T.; Durante, M. Reduction of Lung Metastases in a Mouse Osteosarcoma Model Treated With Carbon Ions and Immune Checkpoint Inhibitors. Int. J. Radiat. Oncol. Biol. Phys. 2020, 109, 594–602. [Google Scholar] [CrossRef]
- Brownstein, J.M.; Wisdom, A.J.; Castle, K.D.; Mowery, Y.M.; Guida, P.; Lee, C.L.; Tommasino, F.; Tessa, C.; Scifoni, E.; Gao, J.; et al. Characterizing the Potency and Impact of Carbon Ion Therapy in a Primary Mouse Model of Soft Tissue Sarcoma. Mol. Cancer Ther. 2018, 17, 858–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGrail, D.J.; Federico, L.; Li, Y.; Dai, H.; Lu, Y.; Mills, G.B.; Yi, S.; Lin, S.-Y.; Sahni, N. Multi-omics analysis reveals neoantigen-independent immune cell infiltration in copy-number driven cancers. Nat. Commun. 2018, 9, 1317. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Usatorre, A.; Carmona, S.J.; Godfroid, C.; Yacoub Maroun, C.; Labiano, S.; Romero, P. Enhanced Phenotype Definition for Precision Isolation of Precursor Exhausted Tumor-Infiltrating CD8 T Cells. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theys, J.; Lambin, P. Clostridium to treat cancer: Dream or reality? Ann. Transl. Med. 2015, 3, S21. [Google Scholar] [CrossRef] [PubMed]
- Uribe-Herranz, M.; Rafail, S.; Beghi, S.; Gil-de-Gómez, L.; Verginadis, I.; Bittinger, K.; Pustylnikov, S.; Pierini, S.; Perales-Linares, R.; Blair, I.A.; et al. Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response. J. Clin. Investig. 2020, 130, 466–479. [Google Scholar] [CrossRef]
- Lhuillier, C.; Vanpouille-Box, C.; Galluzzi, L.; Formenti, S.C.; Demaria, S. Emerging biomarkers for the combination of radiotherapy and immune checkpoint blockers. Semin. Cancer Biol. 2018, 52, 125–134. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Identifier | Pathology | RT Dose | IO | Dose | Status | Study Type | |
---|---|---|---|---|---|---|---|
PrRT | NCT02648997 | Meningiomas | Unknown | Nivolumab * Ipilimumab * | N: 1 mg/kg for 3 weeks I: 3 mg/kg for 3 weeks | Recruiting | Open-label Phase-II |
NCT03267836 | Meningiomas | fRT; 5 × 0.04 Gy Total 0.2 Gy | Avelumab * | Concurrent RT, 10 mg/kg, every 2 weeks for 3 months | Recruiting | Phase I | |
NCT03539198 | Head and neck cancer | fRT; 5× Total 35–45 Gy | Nivolumab * | Before and after RT, Q2/week for 2 weeks | Recruiting | Observational | |
NCT03764787 | Unknown | Unknown | a-PD-1 | Unknown, for 1 year | Not yet recruiting | Phase I/II | |
NCT03765190 | Neoplasm metastasis | Unknown | a-PD-1 | Unknown | Not yet recruiting | Phase I/II | |
NCT03818776 | Non-small cell lung cancer | fRT; 20–23× Total 60–69 Gy (cardiac sparing) | Durvalumab | 1500 mg Q4W, max. 12 months (to 13 doses/cycles) | Recruiting | Early Phase I | |
NCT03087760 | Non-small cell lung cancer | Reirradiation, unknown | Pembroluzimab | Unknown | Recruiting | Phase II | |
NCT02444741 | Non-small cell lung cancer | fRT, 15× low dose, Total unkown | Pembroluzimab | Unknown dose for 21 days, up to 16 cycles | Recruiting | Phase I/II | |
CIRT | NCT04143984 | Locally recurrent nasopharyngeal carcinoma | fRT; 21 × 3 Gy Total 63 Gy | Camrelizumab * | C: 200 mg i.v. every 2 weeks for a year maximum | Not yet recruiting | Phase II/III |
CIRT | NCT03705403 **, [102] | Non-small cell lung cancer | SABR | Darleukin | C: 15 Mio IU, 6 cycles, 3 infusions within one cycle, every 3 weeks | Not yet recruiting | Phase II |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Marcus, D.; Lieverse, R.I.Y.; Klein, C.; Abdollahi, A.; Lambin, P.; Dubois, L.J.; Yaromina, A. Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers 2021, 13, 1468. https://doi.org/10.3390/cancers13061468
Marcus D, Lieverse RIY, Klein C, Abdollahi A, Lambin P, Dubois LJ, Yaromina A. Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers. 2021; 13(6):1468. https://doi.org/10.3390/cancers13061468
Chicago/Turabian StyleMarcus, Damiënne, Relinde I. Y. Lieverse, Carmen Klein, Amir Abdollahi, Philippe Lambin, Ludwig J. Dubois, and Ala Yaromina. 2021. "Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy" Cancers 13, no. 6: 1468. https://doi.org/10.3390/cancers13061468
APA StyleMarcus, D., Lieverse, R. I. Y., Klein, C., Abdollahi, A., Lambin, P., Dubois, L. J., & Yaromina, A. (2021). Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers, 13(6), 1468. https://doi.org/10.3390/cancers13061468