Skip to main content
The British Journal of Radiology logoLink to The British Journal of Radiology
. 2020 Feb 14;93(1107):20190224. doi: 10.1259/bjr.20190224

Harnessing radiation to improve immunotherapy: better with particles?

Marco Durante 1,2,1,2,, Silvia Formenti 3
PMCID: PMC7066943  PMID: 31317768

Abstract

The combination of radiotherapy and immunotherapy is one of the most promising strategies for cancer treatment. Recent clinical results support the pre-clinical experiments pointing to a benefit for the combined treatment in metastatic patients. Charged particle therapy (using protons or heavier ions) is considered one of the most advanced radiotherapy techniques, but its cost remains higher than conventional X-ray therapy. The most important question to be addressed to justify a more widespread use of particle therapy is whether they can be more effective than X-rays in combination with immunotherapy. Protons and heavy ions have physical advantages compared to X-rays that lead to a reduced damage to the immune cells, that are required for an effective immune response. Moreover, densely ionizing radiation may have biological advantages, due to different cell death pathways and release of cytokine mediators of inflammation. We will discuss results in esophageal cancer patients showing that charged particles can reduce the damage to blood lymphocytes compared to X-rays, and preliminary in vitro studies pointing to an increased release of immune-stimulating cytokines after heavy ion exposure. Pre-clinical and clinical studies are ongoing to test these hypotheses.

Introduction

Radiotherapy is an effective and largely used treatment to achieve local control of solid tumors.1 Conventional radiotherapy use X-rays generated by electron accelerators (linacs), but high-energy charged particles produced by larger cyclotrons or synchrotrons improve precision of dose delivery and enable to spare more normal tissue compared to photons.2 Around 170,000 patients have been treated worldwide with protons, and 25,000 with C-ions (source: PTCOG, www.ptcog.ch). This remains a small fraction compared to the millions of patients treated with conventional X-rays. The main reason for this difference is the high cost of the particle accelerators compared to conventional linacs.3 Currently, the cost effectiveness of particle therapy remains under scrutiny.4

Even with high local control rates achievable with particle therapy, this advantage fails to translate in improved cancer survival in most cancers, because of distant metastasis. Local radiotherapy can trigger an immune response that can immunize the host, leading to immune destruction of distant, unirradiated metastasis.5 This phenomenon, described as an abscopal effect, is rare and the mechanisms are unclear.6

In recent years the introduction of checkpoint inhibitors has proven a breakthrough strategy, capable to induce powerful tumor rejections and improved survival in metastatic patients.7 Excellent results have been obtained with immune checkpoint inhibitors such as anti-CTLA4 and anti-PD1 antibodies in malignant melanoma.8 However, severe immune-related side-effects complicate the use of immunotherapy and limits its use in cancer patients.9

It is widely acknowledged that the combination of immunotherapy with local therapies can further improve the survival in most solid cancers.10 Because radiation has the potential to activate an anti tumor immune response,11 it is an optimal candidate for combinations with immunotherapy.12–18 Animal experiments have shown that the superior activity of radiation and dual immune checkpoint blockade is mediated by non-redundant immune mechanisms in cancer.19 Following initial positive results of this combination,20–22 hundreds of trials have been launched to test safety and efficacy of radiation and immunotherapy in different tumors types, including lung cancer,23 the first cause of cancer-related death in United States for both males and females.24 The PACIFIC trial has shown significant improvements in disease free survival25 and overall survival26 in stage III non-small-cell lung cancer (NSCLC) patients treated with Durvalumab after chemoradiotherapy. Prospective randomized trials in stage IV patients are still missing and will be essential to quantify the benefit of combined radiotherapy and immunotherapy in lung cancer. In the pilot/feasibility trial NCT02221739 at Weill Cornell Medicine, a 30% disease control was achieved in stage IV NSCLC patients refractory to anti CTLA-4 alone or in combination with chemotherapy by combining Ipilimumab with focal hypofractionated radiotherapy of a single metastasis.27 Several strategies are under study to further improve these results, including modifying the dose per fraction28 and the number of metastasis irradiated.29

Considering the success and promise of the combination of X-rays with checkpoint inhibitors, the question is whether particle therapy can present additional advantages, and result in better outcomes.30 This question is relevant to the future of particle therapy, in view of its higher cost. We will discuss here the potential physical and biological advantages of particle therapy in combination with immunotherapy.

Physical advantages

Particle therapy is based on the different depth-dose distribution of charged particles compared to photons.31 Using protons or heavier ions, much more normal tissue can be spared in virtually every tumor site, while preserving dose conformality on the tumor (see e.g. Figure 1 for esophageal cancer32). Sparing of the normal tissue, and in particular reducing the exposure of circulating T-lymphocytes and other immune cells, present advantages to immunotherapy. Non-proliferating peripheral blood lymphocytes are very radiosensitive33 and during radiotherapy lymphopenia may occur, which is often correlated to a negative prognosis.34 In fact, both radiotherapy and chemotherapy damage circulating immune cells,35 compromising the host’s immunity.36 Consistently, blocking immune response in mice prevents tumor control when radiation therapy is combined with checkpoint immunotherapy.37

Figure 1. .

Figure 1. 

Coronal and transverse images of 3D-CRT plan (left), IMRT plan (middle), and proton plan (right) for esophageal cancer. The plans clearly show the large tissue sparing using protons. Treatment plans produced at Loma Linda University Medical Center (CA) details in ref.,32 reproduced with permissions.

Damage to peripheral blood lymphocytes can be measured during radiotherapy using the chromosomal aberration assay (Figure 2). When lymphocytes are irradiated in vitro, charged particles induce more chromosomal aberrations than X-rays at the same dose level, and the relative biological effectiveness (RBE) increases by increasing the particle linear energy transfer (LET).38 However, in vivo, the damage mainly depends on the size of the irradiated volume (or integral dose), and on the presence of lymph nodes in the field.39 For sites such as the esophagus, an increased field size leads to a proportional increase of the lymph nodes exposed. In fact, the chromosomal aberrations detected in circulating lymphocytes in esophageal cancer patients are proportional to the radiotherapy target volume.40 As a result of the reduced normal tissue volume irradiated with particles (Figure 1), less chromosomal aberrations were measured in patients treated with C-ions than with X-rays for esophageal cancer,.41 Similar results were reported for patients treated with C-ions or IMRT for prostate cancer.42,43

Figure 2. .

Figure 2. 

Damage in peripheral blood lymphocytes of a patient during the course of radiotherapy. The karyotype by mFISH shows a translocation involving chromosomes 4 and 8.

Damaged lymphocytes eventually die and can lead to lymphopenia. The absolute lymphocyte count decreases during the course of radiotherapy. In sites like the esophagus, it can be expected that the use of protons reduce s the risk of severe lymphopenia.44 In fact, for the decrease in lymphocyte counts during the radiotherapy course is less pronounced using protons45 or C-ions41 than with X-rays (Figure 3). Even if the data in Figure 3 come from different centers and different chemotherapy drugs, the tendency to spare more effectively lymphocytes using particles is supported for esophageal cancer patients. These results suggest that the physical characteristics of charged particle lead to sparing of immune cells that can then be engaged in the immune response triggered by immunotherapy.

Figure 3. .

Figure 3. 

Median values of lymphocyte count in esophageal cancer patients during the course of radiotherapy. Data for protons (1.8 Gy RBE/fraction) and IMRT (1.8 Gy/fraction) are from ref.,45 data for 3DCRT (1.6–2.0 Gy/fraction) and C-ions (2.7–3.6 Gy RBE/fraction) from ref.41

While a reduced lymphopenia is certainly an advantage for the patients, whether lymphocyte sparing boosts the immune response remains to be demonstrated. In esophageal cancer patients receiving pre-operative chemotherapy, a significant increases in T-cell receptor diversity was observed in peripheral T-cells but not in tumor-infiltrating lymphocytes.46 In general, chemotherapy-induced depletion of immune cells can be followed by a ‘rebound overshoot’ effect, leading to transiently enlarged immune cell numbers, then relaxing to normality.47 Whether this transient excess of immune cells supports immunotherapy or not remains to be clarified, and is certainly important to clarify the potential physical advantages of charged particles in radioimmunotherapy.

Biological advantages

In addition to the physical advantages, charged particles have different biological effects compared to X-rays, caused by the different kind of DNA lesions induced by densely ionizing radiation.48 High-LET radiation induce more clustered DNA lesions, that are difficult to repair,49 and that trigger different DNA damage repair signals.50–52

DNA repair signaling pathways are strongly related to the immune response. Recently, it has been directly shown that that PD-L1 expression in cancer cells is upregulated in response to DNA double-strand breakage, through the ATM/ATR/Chk1 kinase pathway.53 Similar upregulation of PD-L1 has been recently shown in melanoma cells exposed to UV radiation.54 Activation of different DNA damage response pathways at high-LET, such as resection,55 may have different effects on the expression of immune receptors. It presently not known whether the radiation-induced upregulation of PD-L1 will actually translate into response to checkpoint inhibitors.56

DNA damage eventually leads to cell death through different pathways (such as apoptosis, necrosis, mitotic catastrophe or senescence),57,58 and to the consequent release of small molecules such as ATP, calreticulin, and HMGB159 that can trigger the immune response (Figure 4).60 In vitro studies with different human tumor cell lines have shown that proton radiation induces calreticulin cell-surface expression, increasing sensitivity to cytotoxic T-lymphocyte killing of tumor cells.61 Increased extracellular concentration of HMGB1 has been measured after irradiation of human cancer cells with heavy ions,62 and the release is positively correlated to the particle LET (Figure 5). HMGB1 is upregulated in the serum of esophageal cancer patients following chemoradiotherapy,63 and these results are therefore suggestive of an increased efficiency of densely ionizing radiation compared to X-rays.

Figure 4. .

Figure 4. 

An illustration of immune-mediated effects of ionizing radiation. The green arrow points to the release of HGMB1, that interacts with the toll-like receptor TLR4 activating the dendritic cell maturation. Cartoon from ref.,60 reproduced with permission.

Figure 5. .

Figure 5. 

Concentration of HGMB1 in the medium following irradiation of HeLa cells with sham (0 Gy) radiation (black), 4 Gy C-ions in the plateau region of the Bragg curve, 13 keV/μm (red) and 2 Gy C-ions in the spread-out-Bragg-peak, 70 keV/μm (green). The two doses were chosen to achieve the same 10% survival in irradiated HeLa cells. Data from ref.62

In addition to the damage to nuclear DNA, recent evidence points to the sensing of cytoplasmic DNA as a key factor in eliciting immune response.64 Double stranded DNA (dsDNA) fragments generated by exposure to sparsely ionizing radiation are extruded from the nucleus65,66 and accumulate in the cytosol where dsDNA sensors, cGAS/STING are activated to transcribe type I interferon genes and jumpstart an immune response. Interferon is a powerful promoter of dendritic cells recruitment and activation, enabling cross-presentation of radiation induced neoantigens and immune response.67,68 Consistently, metastatic NSCLC patients responding to focal radiation and CTLA4 blockade demonstrated an increased serum level of interferon-βcompared to baseline.27 These results suggest that the cGAS/STING activation may be an essential pathway for a successful combination of radiation and immunotherapy.69,70 Increased cytosolic DNA is observed after irradiation of human cancer cells, but at high doses per fraction (>10 Gy) cells activate the DNA exonuclease Trex1, that degrades cytosolic DNA thus blocking the STING activation.71 Type-I interferon activation induced by radiation therefore reflects a balance between sufficient dsDNA induction to stimulate cGAS/STING and Trex1 activation.72 How this pathway is affected by charged particles remains unknown, and experiments are needed to clarify whether the production of smaller DNA fragments by densely ionizing radiation can lead to an enhanced response.73

Preliminary animal studies have shown that charged particles effectively induce abscopal responses for instance by reducing lung metastasis after irradiation of the primary tumor.74–76 However, these results remain inconclusive and a direct comparison of X-rays, protons, and C-ions is warranted.77

Conclusions

The potential advantages in using protons or heavy ions in combination with immunotherapy are based on both their physical and biological properties, warranting experimental verification. Since many pre-clinical and clinical trials are ongoing, it is expected that within a few years the role of charged particles for a combined use with modern immunotherapy of cancer will be defined. Several new proton therapy centers are under construction in Europe, and many of these centers will dedicate “beamtime” to radiobiology research.78 Often these research efforts are directed to measurements of proton RBE,79 but one of the key questions to be answered is whether particles are more effective than X-rays when used in combination with immunotherapy.30 Considering the high cost of the proton or heavy ion therapy facilities and the success of radioimmunotherapy trials, this research is perhaps the most important to decide the future of particle therapy.

Footnotes

Acknowledgments: This work was partly supported by EU Horizon2020 grant 73,0983 (INSPIRE). The research in radioimmunotherapy at GSI is in the frame of FAIR Phase-0 supported by the GSI Helmholtzzentrum für Schwerionenforschung in Darmstadt (Germany).

Contributor Information

Marco Durante, Email: M.Durante@gsi.de.

Silvia Formenti, Email: formenti@med.cornell.edu.

REFERENCES

  • 1.Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, et al. Radiation oncology in the era of precision medicine. Nat Rev Cancer 2016; 16: 234–49. doi: 10.1038/nrc.2016.18 [DOI] [PubMed] [Google Scholar]
  • 2.Durante M, Orecchia R, Loeffler JS. Charged-Particle therapy in cancer: clinical uses and future perspectives. Nat Rev Clin Oncol 2017; 14: 483–95. doi: 10.1038/nrclinonc.2017.30 [DOI] [PubMed] [Google Scholar]
  • 3.Bortfeld TR, Loeffler JS. Three ways to make proton therapy affordable. Nature 2017; 549: 451–3. doi: 10.1038/549451a [DOI] [PubMed] [Google Scholar]
  • 4.Mitin T, Zietman AL. Promise and pitfalls of heavy-particle therapy. JCO 2014; 32: 2855–63. doi: 10.1200/JCO.2014.55.1945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys 2005; 63: 655–66. doi: 10.1016/j.ijrobp.2005.06.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Abuodeh Y, Venkat P, Kim S. Systematic review of case reports on the abscopal effect. Curr Probl Cancer 2016; 40: 25–37. doi: 10.1016/j.currproblcancer.2015.10.001 [DOI] [PubMed] [Google Scholar]
  • 7.Kirkwood JM, Butterfield LH, Tarhini AA, Zarour H, Kalinski P, Ferrone S. Immunotherapy of cancer in 2012. CA Cancer J Clin 2012; 62: 309–35. doi: 10.3322/caac.20132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015; 372: 2521–32. doi: 10.1056/NEJMoa1503093 [DOI] [PubMed] [Google Scholar]
  • 9.Postow MA, Sidlow R, Hellmann MD, Longo DL. Immune-Related adverse events associated with immune checkpoint blockade. N Engl J Med (eds 2018; 378: 158–68. doi: 10.1056/NEJMra1703481 [DOI] [PubMed] [Google Scholar]
  • 10.Whiteside TL, Demaria S, Rodriguez-Ruiz ME, Zarour HM, Melero I. Emerging opportunities and challenges in cancer immunotherapy. Clin Cancer Res 2016; 22: 1845–55. doi: 10.1158/1078-0432.CCR-16-0049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol 2009; 10: 718–26. doi: 10.1016/S1470-2045(09)70082-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer 2018; 18: 313–22. doi: 10.1038/nrc.2018.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 2015; 1: 1325–32. doi: 10.1001/jamaoncol.2015.2756 [DOI] [PubMed] [Google Scholar]
  • 14.Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst 2013; 105: 256–65. doi: 10.1093/jnci/djs629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Weichselbaum RR, Liang H, Deng L, Fu Y-X. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol 2017; 14: 365–79. doi: 10.1038/nrclinonc.2016.211 [DOI] [PubMed] [Google Scholar]
  • 16.Durante M, Reppingen N, Held KD. Immunologically augmented cancer treatment using modern radiotherapy. Trends Mol Med 2013; 19: 565–82. doi: 10.1016/j.molmed.2013.05.007 [DOI] [PubMed] [Google Scholar]
  • 17.Van Limbergen EJ, De Ruysscher DK, Olivo Pimentel V, Marcus D, Berbee M, Hoeben A, et al. Combining radiotherapy with immunotherapy: the past, the present and the future. Br J Radiol 2017; 90: 20170157. doi: 10.1259/bjr.20170157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Herrera FG, Bourhis J, Coukos G. Radiotherapy combination opportunities Leveraging immunity for the next oncology practice. CA Cancer J Clin 2017; 67: 65–85. doi: 10.3322/caac.21358 [DOI] [PubMed] [Google Scholar]
  • 19.Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015; 520: 373–7. doi: 10.1038/nature14292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 2012; 366: 925–31. doi: 10.1056/NEJMoa1112824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hiniker SM, Chen DS, Knox SJ. Abscopal effect in a patient with melanoma. N Engl J Med 2012; 366: 2035–6. doi: 10.1056/NEJMc1203984 [DOI] [PubMed] [Google Scholar]
  • 22.Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 2015; 16: 795–803. doi: 10.1016/S1470-2045(15)00054-6 [DOI] [PubMed] [Google Scholar]
  • 23.Ko EC, Raben D, Formenti SC. The integration of radiotherapy with immunotherapy for the treatment of non-small cell lung cancer. Clin Cancer Res 2018; 24: 5792–806. doi: 10.1158/1078-0432.CCR-17-3620 [DOI] [PubMed] [Google Scholar]
  • 24.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69: 7–34. doi: 10.3322/caac.21551 [DOI] [PubMed] [Google Scholar]
  • 25.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 2017; 377: 1919–29. doi: 10.1056/NEJMoa1709937 [DOI] [PubMed] [Google Scholar]
  • 26.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Overall survival with Durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med 2018; 379: 2342–50. doi: 10.1056/NEJMoa1809697 [DOI] [PubMed] [Google Scholar]
  • 27.Formenti SC, Rudqvist N-P, Golden E, Cooper B, Wennerberg E, Lhuillier C, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med 2018; 24: 1845–51. doi: 10.1038/s41591-018-0232-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Formenti SC. Optimizing dose per fraction: a new chapter in the story of the Abscopal effect? Int J Radiat Oncol Biol Phys 2017; 99: 677–9. doi: 10.1016/j.ijrobp.2017.07.028 [DOI] [PubMed] [Google Scholar]
  • 29.Brooks ED, Chang JY. Time to abandon single-site irradiation for inducing abscopal effects. Nat Rev Clin Oncol 2019; 16: 123–35. doi: 10.1038/s41571-018-0119-7 [DOI] [PubMed] [Google Scholar]
  • 30.Durante M, Brenner DJ, Formenti SC. Does heavy ion therapy work through the immune system? Int J Radiat Oncol Biol Phys 2016; 96: 934–6. doi: 10.1016/j.ijrobp.2016.08.037 [DOI] [PubMed] [Google Scholar]
  • 31.Durante M, Paganetti H. Nuclear physics in particle therapy: a review. Rep Prog Phys 2016; 79: 096702. doi: 10.1088/0034-4885/79/9/096702 [DOI] [PubMed] [Google Scholar]
  • 32.Ling TC, Slater JM, Nookala P, Mifflin R, Grove R, Ly AM Grove R, Patyal B, Yang G, et al. Analysis of intensity-modulated radiation therapy (IMRT), proton and 3D conformal radiotherapy (3D-CRT) for reducing perioperative cardiopulmonary complications in esophageal cancer patients. Cancers 2014; 6: 2356–68. doi: 10.3390/cancers6042356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.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. doi: 10.1038/s41419-018-1095-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Venkatesulu BP, Mallick S, Lin SH, Krishnan S. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018; 123(January): 42–51. doi: 10.1016/j.critrevonc.2018.01.003 [DOI] [PubMed] [Google Scholar]
  • 35.Yovino S, Kleinberg L, Grossman SA, Narayanan M, Ford E. The etiology of treatment-related lymphopenia in patients with malignant gliomas: modeling radiation dose to circulating lymphocytes explains clinical observations and suggests methods of modifying the impact of radiation on immune cells. Cancer Invest 2013; 31: 140–4. doi: 10.3109/07357907.2012.762780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lobachevsky PN, Ventura J, Giannakandropoulou L, Forrester H, Palazzolo JS, Haynes NM, et al. A functional immune system is required for the systemic genotoxic effects of localized irradiation. Int J Radiat Oncol Biol Phys 2019; 103: 1184–93. doi: 10.1016/j.ijrobp.2018.11.066 [DOI] [PubMed] [Google Scholar]
  • 37.Crittenden MR, Zebertavage L, Kramer G, Bambina S, Friedman D, Troesch V, et al. Tumor cure by radiation therapy and checkpoint inhibitors depends on pre-existing immunity . Sci Rep 2018; 8: 7012. doi: 10.1038/s41598-018-25482-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ritter S, Durante M. Heavy-Ion induced chromosomal aberrations: a review. Mutat Res 2010; 701: 38–46. doi: 10.1016/j.mrgentox.2010.04.007 [DOI] [PubMed] [Google Scholar]
  • 39.d'Alesio V, Pacelli R, Durante M, Canale Cama G, Cella L, Gialanella G, et al. Lymph nodes in the irradiated field influence the yield of radiation-induced chromosomal aberrations in lymphocytes from breast cancer patients. Int J Radiat Oncol Biol Phys 2003; 57: 732–8. doi: 10.1016/S0360-3016(03)00664-3 [DOI] [PubMed] [Google Scholar]
  • 40.Durante M, Yamada S, Ando K, Furusawa Y, Kawata T, Majima H, et al. Measurements of the equivalent whole-body dose during radiation therapy by cytogenetic methods. Phys Med Biol 1999; 44: 1289–98. doi: 10.1088/0031-9155/44/5/314 [DOI] [PubMed] [Google Scholar]
  • 41.Durante M, Yamada S, Ando K, Furusawa Y, Kawata T, Majima H, et al. X-Rays vs. carbon-ion tumor therapy: cytogenetic damage in lymphocytes. Int J Radiat Oncol Biol Phys 2000; 47: 793–8. doi: 10.1016/S0360-3016(00)00455-7 [DOI] [PubMed] [Google Scholar]
  • 42.Hartel C, Nikoghosyan A, Durante M, Sommer S, Nasonova E, Fournier C, et al. Chromosomal aberrations in peripheral blood lymphocytes of prostate cancer patients treated with IMRT and carbon ions. Radiother Oncol 2010; 95: 73–8. doi: 10.1016/j.radonc.2009.08.031 [DOI] [PubMed] [Google Scholar]
  • 43.Pignalosa D, Lee R, Hartel C, Sommer S, Nikoghosyan A, Debus J, et al. Chromosome inversions in lymphocytes of prostate cancer patients treated with x-rays and carbon ions. Radiother Oncol 2013; 109: 256–61. doi: 10.1016/j.radonc.2013.09.021 [DOI] [PubMed] [Google Scholar]
  • 44.Fang P, Shiraishi Y, Verma V, Jiang W, Song J, Hobbs BP, et al. Lymphocyte-Sparing effect of proton therapy in patients with esophageal cancer treated with definitive chemoradiation. Int J Part Ther 2017; 4: 23–32. doi: 10.14338/IJPT-17-00033.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Davuluri R, Jiang W, Fang P, Xu C, Komaki R, Gomez DR, et al. Lymphocyte nadir and esophageal cancer survival outcomes after chemoradiation therapy. Int J Radiat Oncol Biol Phys 2017; 99: 128–35. doi: 10.1016/j.ijrobp.2017.05.037 [DOI] [PubMed] [Google Scholar]
  • 46.Zhang C, Palashati H, Tan Q, Ku W, Miao Y, Xiong H, et al. Immediate and substantial evolution of T-cell repertoire in peripheral blood and tumor microenvironment of patients with esophageal squamous cell carcinoma treated with preoperative chemotherapy. Carcinogenesis 2018; 39: 1389–98. doi: 10.1093/carcin/bgy116 [DOI] [PubMed] [Google Scholar]
  • 47.Proietti E, Moschella F, Capone I, Belardelli F. Exploitation of the propulsive force of chemotherapy for improving the response to cancer immunotherapy. Mol Oncol 2012; 6: 1–14. doi: 10.1016/j.molonc.2011.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Durante M. New challenges in high-energy particle radiobiology. Br J Radiol 2014; 87: 20130626. doi: 10.1259/bjr.20130626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Asaithamby A, Hu B, Chen DJ. Unrepaired clustered DNA lesions induce chromosome breakage in human cells. Proc Natl Acad Sci U S A 2011; 108: 8293–8. doi: 10.1073/pnas.1016045108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Löbrich M, Jeggo PA. The impact of a negligent G2/M checkpoint on genomic instability and cancer induction. Nat Rev Cancer 2007; 7: 861–9. doi: 10.1038/nrc2248 [DOI] [PubMed] [Google Scholar]
  • 51.Sage E, Harrison L. Clustered DNA lesion repair in eukaryotes: relevance to mutagenesis and cell survival. Mutat Res 2011; 711(1–2): 123–33. doi: 10.1016/j.mrfmmm.2010.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bukowska B, Karwowski BT. The Clustered DNA Lesions - Types, Pathways of Repair and Relevance to Human Health. Curr Med Chem 2018; 25: 2722–35. doi: 10.2174/0929867325666180226110502 [DOI] [PubMed] [Google Scholar]
  • 53.Sato H, Niimi A, Yasuhara T, Permata TBM, Hagiwara Y, Isono M, et al. Dna double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat Commun 2017; 8. doi: 10.1038/s41467-017-01883-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang W, Chapman NM, Zhang B, Li M, Fan M, Laribee RN, et al. Upregulation of PD-L1 via HMGB1-Activated IRF3 and NF-κB contributes to UV radiation-induced immune suppression. Cancer Res 2019; 79: 2909–22. doi: 10.1158/0008-5472.CAN-18-3134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Averbeck NB, Ringel O, Herrlitz M, Jakob B, Durante M, Taucher-Scholz G. Dna end resection is needed for the repair of complex lesions in G1-phase human cells. Cell Cycle 2014; 13: 2509–16. doi: 10.4161/15384101.2015.941743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Shevtsov M, Sato H, Multhoff G, Shibata A. Novel approaches to improve the efficacy of Immuno-Radiotherapy. Front Oncol 2019; 9: 156. doi: 10.3389/fonc.2019.00156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Paludan SR, Reinert LS, Hornung V. Dna-Stimulated cell death: implications for host defence, inflammatory diseases and cancer. Nat Rev Immunol 2019; 19: 141–53. doi: 10.1038/s41577-018-0117-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol 2011; 12: 385–92. doi: 10.1038/nrm3115 [DOI] [PubMed] [Google Scholar]
  • 59.Lotze MT, Tracey KJ. High-Mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 2005; 5: 331–42. doi: 10.1038/nri1594 [DOI] [PubMed] [Google Scholar]
  • 60.Ebner DK, Tinganelli W, Helm A, Bisio A, Yamada S, Kamada T, et al. The immunoregulatory potential of particle radiation in cancer therapy. Front Immunol 2017; 8: 99. doi: 10.3389/fimmu.2017.00099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gameiro SR, Malamas AS, Bernstein MB, Tsang KY, Vassantachart A, Sahoo N, 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: 120–30. doi: 10.1016/j.ijrobp.2016.02.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Onishi M, Okonogi N, Oike T, Yoshimoto Y, Sato H, Suzuki Y, et al. 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: 541–6. doi: 10.1093/jrr/rry049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Suzuki Y, Mimura K, Yoshimoto Y, Watanabe M, Ohkubo Y, Izawa S, et al. Immunogenic tumor cell death induced by chemoradiotherapy in patients with esophageal squamous cell carcinoma. Cancer Res 2012; 72: 3967–76. doi: 10.1158/0008-5472.CAN-12-0851 [DOI] [PubMed] [Google Scholar]
  • 64.Corrales L, Matson V, Flood B, Spranger S, Gajewski TF. Innate immune signaling and regulation in cancer immunotherapy. Cell Res 2017; 27: 96–108. doi: 10.1038/cr.2016.149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mackenzie KJ, Carroll P, Martin C-A, Murina O, Fluteau A, Simpson DJ, et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 2017; 548: 461–5. doi: 10.1038/nature23449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017; 548: 466–70. doi: 10.1038/nature23470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol 2016; 17: 1142–9. doi: 10.1038/ni.3558 [DOI] [PubMed] [Google Scholar]
  • 68.Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol 2015; 15: 405–14. doi: 10.1038/nri3845 [DOI] [PubMed] [Google Scholar]
  • 69.Galluzzi L, Vanpouille-Box C, Bakhoum SF, Demaria S. Snapshot: cGAS-STING signaling. Cell 2018; 173: 276–276.e1. doi: 10.1016/j.cell.2018.03.015 [DOI] [PubMed] [Google Scholar]
  • 70.Vanpouille-Box C, Demaria S, Formenti SC, Galluzzi L. Cytosolic DNA sensing in organismal tumor control. Cancer Cell 2018; 34: 361–78. doi: 10.1016/j.ccell.2018.05.013 [DOI] [PubMed] [Google Scholar]
  • 71.Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. Dna exonuclease TREX1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017; 8: 15618. doi: 10.1038/ncomms15618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vanpouille-Box C, Formenti SC, Demaria S. Trex1 dictates the immune fate of irradiated cancer cells. Oncoimmunology 2017; 6: e1339857. doi: 10.1080/2162402X.2017.1339857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Durante M, Formenti SC. Radiation-Induced chromosomal aberrations and immunotherapy: micronuclei, cytosolic DNA, and Interferon-Production pathway. Front Oncol 2018; 8: 192. doi: 10.3389/fonc.2018.00192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.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. doi: 10.14338/IJPT-15-00023.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ando K, Fujita H, Hosoi A, Ma L, Wakatsuki M, Seino K-I, et al. Intravenous dendritic cell administration enhances suppression of lung metastasis induced by carbon-ion irradiation. J Radiat Res 2017; 58: 446–55. doi: 10.1093/jrr/rrx005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sato K, Nitta N, Aoki I, Imai T, Shimokawa T. Repeated photon and C-ion irradiations in vivo have different impact on alteration of tumor characteristics. Sci Rep 2018; 8: 1458. doi: 10.1038/s41598-018-19422-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ebner DK, Tinganelli W, Helm A, Bisio A, Simoniello P, Natale F, et al. Generating and grading the abscopal effect: proposal for comprehensive evaluation of combination immunoradiotherapy in mouse models. Transl Cancer Res 2017; 6(S5): S892–S899. doi: 10.21037/tcr.2017.06.01 [DOI] [Google Scholar]
  • 78.Durante M. Proton beam therapy in Europe: more centres need more research. Br J Cancer 2019; ; 120: 777–8. doi: 10.1038/s41416-018-0329-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Paganetti H. Relative biological effectiveness (RBE) values for proton beam therapy. variations as a function of biological endpoint, dose, and linear energy transfer. Phys Med Biol 2014; 59: R419–R472. doi: 10.1088/0031-9155/59/22/R419 [DOI] [PubMed] [Google Scholar]

Articles from The British Journal of Radiology are provided here courtesy of Oxford University Press

RESOURCES