You are on page 1of 7

BJR © 2020 The Authors.

Published by the British Institute of Radiology


https://​doi.​org/​10.​1259/​bjr.​20190224
Received: Revised: Accepted:
27 February 2019 10 June 2019 14 July 2019

Cite this article as:


Durante M, Formenti S. Harnessing radiation to improve immunotherapy: better with particles?. Br J Radiol 2020; 93: 20190224.

Proton Therapy special feature: Review Article

Harnessing radiation to improve immunotherapy: better


with particles?
1,2
Marco Durante, PhD and 3Silvia Formenti, MD
1
GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Department, Darmstadt, Germany
2
Technische Universität Darmstadt, Institut für Festkörperphysik, Darmstadt, Germany
3
Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA

Address correspondence to: Professor Marco Durante


E-mail: ​M.​Durante@​gsi.​de

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 meta-
static patients. Charged particle therapy (using protons or heavier ions) is considered one of the most advanced radi-
otherapy 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 In recent years the introduction of checkpoint inhibitors


Radiotherapy is an effective and largely used treatment has proven a breakthrough strategy, capable to induce
to achieve local control of solid tumors.1 Conventional powerful tumor rejections and improved survival in meta-
radiotherapy use X-­rays generated by electron accelerators static patients.7 Excellent results have been obtained with
(linacs), but high-­ energy charged particles produced by immune checkpoint inhibitors such as anti-­CTLA4 and
larger cyclotrons or synchrotrons improve precision of dose anti-­PD1 antibodies in malignant melanoma.8 However,
delivery and enable to spare more normal tissue compared to severe immune-­related side-­effects complicate the use of
photons.2 Around 170,000 patients have been treated world- immunotherapy and limits its use in cancer patients.9
wide with protons, and 25,000 with C-­ions (source: PTCOG,
www.​ptcog.​ch). This remains a small fraction compared to It is widely acknowledged that the combination of immu-
the millions of patients treated with conventional X-­rays. The notherapy with local therapies can further improve the
main reason for this difference is the high cost of the particle survival in most solid cancers.10 Because radiation has the
accelerators compared to conventional linacs.3 Currently, the potential to activate an anti tumor immune response,11 it
cost effectiveness of particle therapy remains under scrutiny.4 is an optimal candidate for combinations with immuno-
therapy.12–18 Animal experiments have shown that the
Even with high local control rates achievable with particle superior activity of radiation and dual immune checkpoint
therapy, this advantage fails to translate in improved cancer blockade is mediated by non-­redundant immune mecha-
survival in most cancers, because of distant metastasis. nisms in cancer.19 Following initial positive results of this
Local radiotherapy can trigger an immune response that combination,20–22 hundreds of trials have been launched
can immunize the host, leading to immune destruction to test safety and efficacy of radiation and immunotherapy
of distant, unirradiated metastasis.5 This phenomenon, in different tumors types, including lung cancer,23 the
described as an abscopal effect, is rare and the mechanisms first cause of cancer-­ related death in United States for
are unclear.6 both males and females.24 The PACIFIC trial has shown
BJR Durante and Formenti

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.

significant improvements in disease free survival25 and overall immunity.36 Consistently, blocking immune response in mice
survival26 in stage III non-­ small-­cell lung cancer (NSCLC) prevents tumor control when radiation therapy is combined with
patients treated with Durvalumab after chemoradiotherapy. checkpoint immunotherapy.37
Prospective randomized trials in stage IV patients are still
missing and will be essential to quantify the benefit of combined Damage to peripheral blood lymphocytes can be measured during
radiotherapy and immunotherapy in lung cancer. In the pilot/ radiotherapy using the chromosomal aberration assay (Figure 2).
feasibility trial NCT02221739 at Weill Cornell Medicine, a When lymphocytes are irradiated in vitro, charged particles induce
30% disease control was achieved in stage IV NSCLC patients more chromosomal aberrations than X-­rays at the same dose
refractory to anti CTLA-4 alone or in combination with chemo- level, and the relative biological effectiveness (RBE) increases by
therapy by combining Ipilimumab with focal hypofractionated increasing the particle linear energy transfer (LET).38 However,
radiotherapy of a single metastasis.27 Several strategies are under in vivo, the damage mainly depends on the size of the irradiated
study to further improve these results, including modifying the volume (or integral dose), and on the presence of lymph nodes in
dose per fraction28 and the number of metastasis irradiated.29 the field.39 For sites such as the esophagus, an increased field size
leads to a proportional increase of the lymph nodes exposed. In
Considering the success and promise of the combination of fact, the chromosomal aberrations detected in circulating lympho-
X-­rays with checkpoint inhibitors, the question is whether cytes in esophageal cancer patients are proportional to the radio-
particle therapy can present additional advantages, and result therapy target volume.40 As a result of the reduced normal tissue
in better outcomes.30 This question is relevant to the future of volume irradiated with particles (Figure 1), less chromosomal aber-
particle therapy, in view of its higher cost. We will discuss here rations were measured in patients treated with C-­ions than with
the potential physical and biological advantages of particle X-­rays for esophageal cancer,.41 Similar results were reported for
therapy in combination with immunotherapy. patients treated with C-­ions or IMRT for prostate cancer.42,43

Physical advantages Damaged lymphocytes eventually die and can lead to lymph-
Particle therapy is based on the different depth-­dose distribution openia. The absolute lymphocyte count decreases during the
of charged particles compared to photons.31 Using protons or course of radiotherapy. In sites like the esophagus, it can be
heavier ions, much more normal tissue can be spared in virtu- expected that the use of protons reduce s the risk of severe lymph-
ally every tumor site, while preserving dose conformality on the openia.44 In fact, for the decrease in lymphocyte counts during
tumor (see e.g. Figure 1 for esophageal cancer32). Sparing of the the radiotherapy course is less pronounced using protons45 or
normal tissue, and in particular reducing the exposure of circu- C-­ions41 than with X-­rays (Figure 3). Even if the data in Figure 3
lating T-­lymphocytes and other immune cells, present advan- come from different centers and different chemotherapy drugs,
tages to immunotherapy. Non-­ proliferating peripheral blood the tendency to spare more effectively lymphocytes using parti-
lymphocytes are very radiosensitive33 and during radiotherapy cles is supported for esophageal cancer patients. These results
lymphopenia may occur, which is often correlated to a nega- suggest that the physical characteristics of charged particle
tive prognosis.34 In fact, both radiotherapy and chemotherapy lead to sparing of immune cells that can then be engaged in the
damage circulating immune cells,35 compromising the host’s immune response triggered by immunotherapy.

2 of 7 birpublications.org/bjr Br J Radiol;93:20190224
Charged particles and immunotherapy BJR

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.

While a reduced lymphopenia is certainly an advantage for patients receiving pre-­ operative chemotherapy, a significant
the patients, whether lymphocyte sparing boosts the immune increases in T-­cell receptor diversity was observed in peripheral
response remains to be demonstrated. In esophageal cancer 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
Figure 3.  Median values of lymphocyte count in esophageal
cancer patients during the course of radiotherapy. Data for
enlarged immune cell numbers, then relaxing to normality.47
protons (1.8 Gy RBE/fraction) and IMRT (1.8 Gy/fraction) are Whether this transient excess of immune cells supports immuno-
from ref.,45 data for 3DCRT (1.6–2.0 Gy/fraction) and C-­ions therapy or not remains to be clarified, and is certainly important
(2.7–3.6 Gy RBE/fraction) from ref.41 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 radia-
tion.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

3 of 7 birpublications.org/bjr Br J Radiol;93:20190224
BJR Durante and Formenti

Figure 4.  An illustration of immune-­ mediated effects of Figure 5.  Concentration of HGMB1 in the medium following
ionizing radiation. The green arrow points to the release of irradiation of HeLa cells with sham (0 Gy) radiation (black),
HGMB1, that interacts with the toll-­like receptor TLR4 activat- 4 Gy C-­ions in the plateau region of the Bragg curve, 13 keV/
ing the dendritic cell maturation. Cartoon from ref.,60 repro- μm (red) and 2 Gy C-­ions in the spread-­out-­Bragg-­peak, 70
duced with permission. keV/μm (green). The two doses were chosen to achieve the
same 10% survival in irradiated HeLa cells. Data from ref.62

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 activa-
tion, enabling cross-­presentation of radiation induced neoanti-
gens 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 activa-
recently shown in melanoma cells exposed to UV radiation.54 tion may be an essential pathway for a successful combination
Activation of different DNA damage response pathways at of radiation and immunotherapy.69,70 Increased cytosolic DNA
high-­LET, such as resection,55 may have different effects on the is observed after irradiation of human cancer cells, but at high
expression of immune receptors. It presently not known whether doses per fraction (>10 Gy) cells activate the DNA exonuclease
the radiation-­induced upregulation of PD-­L1 will actually trans- Trex1, that degrades cytosolic DNA thus blocking the STING
late into response to checkpoint inhibitors.56 activation.71 Type-­I interferon activation induced by radiation
therefore reflects a balance between sufficient dsDNA induc-
DNA damage eventually leads to cell death through different tion to stimulate cGAS/STING and Trex1 activation.72 How this
pathways (such as apoptosis, necrosis, mitotic catastrophe or pathway is affected by charged particles remains unknown, and
senescence),57,58 and to the consequent release of small mole- experiments are needed to clarify whether the production of
cules such as ATP, calreticulin, and HMGB159 that can trigger smaller DNA fragments by densely ionizing radiation can lead to
the immune response (Figure 4).60 In vitro studies with different an enhanced response.73
human tumor cell lines have shown that proton radiation induces
calreticulin cell-­surface expression, increasing sensitivity to Preliminary animal studies have shown that charged particles
cytotoxic T-­lymphocyte killing of tumor cells.61 Increased extra- effectively induce abscopal responses for instance by reducing
cellular concentration of HMGB1 has been measured after irra- lung metastasis after irradiation of the primary tumor.74–76
diation of human cancer cells with heavy ions,62 and the release However, these results remain inconclusive and a direct
is positively correlated to the particle LET (Figure 5). HMGB1 is comparison of X-­rays, protons, and C-­ions is warranted.77
upregulated in the serum of esophageal cancer patients following
chemoradiotherapy,63 and these results are therefore suggestive Conclusions
of an increased efficiency of densely ionizing radiation compared The potential advantages in using protons or heavy ions in combi-
to X-­rays. nation with immunotherapy are based on both their physical and
biological properties, warranting experimental verification. Since
In addition to the damage to nuclear DNA, recent evidence points many pre-­clinical and clinical trials are ongoing, it is expected that
to the sensing of cytoplasmic DNA as a key factor in eliciting within a few years the role of charged particles for a combined use

4 of 7 birpublications.org/bjr Br J Radiol;93:20190224
Charged particles and immunotherapy BJR

with modern immunotherapy of cancer will be defined. Several of radioimmunotherapy trials, this research is perhaps the most
new proton therapy centers are under construction in Europe, important to decide the future of particle therapy.
and many of these centers will dedicate “beamtime” to radio-
biology research.78 Often these research efforts are directed to Acknowledgments
measurements of proton RBE,79 but one of the key questions to be This work was partly supported by EU Horizon2020 grant
answered is whether particles are more effective than X-­rays when 73,0983 (INSPIRE). The research in radioimmunotherapy at GSI
used in combination with immunotherapy.30 Considering the high is in the frame of FAIR Phase-0 supported by the GSI Helmholtz-
cost of the proton or heavy ion therapy facilities and the success zentrum für Schwerionenforschung in Darmstadt (Germany).

References
1. Baumann M, Krause M, Overgaard J, Debus 1845–55. doi: https://​doi.​org/​10.​1158/​1078-​ mechanisms in cancer. Nature 2015;
J, Bentzen SM, Daartz J, et al. Radiation 0432.​CCR-​16-​0049 520: 373–7. doi: https://​doi.​org/​10.​1038/​
oncology in the era of precision medicine. 11. Formenti SC, Demaria S. Systemic effects of nature14292
Nat Rev Cancer 2016; 16: 234–49. doi: local radiotherapy. Lancet Oncol 2009; 10: 20. Postow MA, Callahan MK, Barker
https://​doi.​org/​10.​1038/​nrc.​2016.​18 718–26. doi: https://​doi.​org/​10.​1016/​S1470-​ CA, Yamada Y, Yuan J, Kitano S, et al.
2. Durante M, Orecchia R, Loeffler JS. 2045(​09)​70082-8 Immunologic correlates of the abscopal effect
Charged-­Particle therapy in cancer: clinical 12. Ngwa W, Irabor OC, Schoenfeld JD, in a patient with melanoma. N Engl J Med
uses and future perspectives. Nat Rev Clin Hesser J, Demaria S, Formenti SC. Using 2012; 366: 925–31. doi: https://​doi.​org/​10.​
Oncol 2017; 14: 483–95. doi: https://​doi.​org/​ immunotherapy to boost the abscopal 1056/​NEJMoa1112824
10.​1038/​nrclinonc.​2017.​30 effect. Nat Rev Cancer 2018; 18: 313–22. doi: 21. Hiniker SM, Chen DS, Knox SJ. Abscopal
3. Bortfeld TR, Loeffler JS. Three ways to make https://​doi.​org/​10.​1038/​nrc.​2018.6 effect in a patient with melanoma. N Engl J
proton therapy affordable. Nature 2017; 549: 13. Demaria S, Golden EB, Formenti SC. Med 2012; 366: 2035–6. doi: https://​doi.​org/​
451–3. doi: https://​doi.​org/​10.​1038/​549451a Role of local radiation therapy in cancer 10.​1056/​NEJMc1203984
4. Mitin T, Zietman AL. Promise and pitfalls immunotherapy. JAMA Oncol 2015; 1: 1325– 22. Golden EB, Chhabra A, Chachoua A,
of heavy-­particle therapy. JCO 2014; 32: 32. doi: https://​doi.​org/​10.​1001/​jamaoncol.​ Adams S, Donach M, Fenton-­Kerimian M,
2855–63. doi: https://​doi.​org/​10.​1200/​JCO.​ 2015.​2756 et al. Local radiotherapy and granulocyte-­
2014.​55.​1945 14. Formenti SC, Demaria S. Combining macrophage colony-­stimulating factor to
5. Demaria S, Bhardwaj N, McBride WH, radiotherapy and cancer immunotherapy: a generate abscopal responses in patients
Formenti SC. Combining radiotherapy and paradigm shift. J Natl Cancer Inst 2013; 105: with metastatic solid tumours: a proof-­
immunotherapy: a revived partnership. Int J 256–65. doi: https://​doi.​org/​10.​1093/​jnci/​ of-­principle trial. Lancet Oncol 2015; 16:
Radiat Oncol Biol Phys 2005; 63: 655–66. doi: djs629 795–803. doi: https://​doi.​org/​10.​1016/​S1470-​
https://​doi.​org/​10.​1016/​j.​ijrobp.​2005.​06.​032 15. Weichselbaum RR, Liang H, Deng L, Fu 2045(​15)​00054-6
6. Abuodeh Y, Venkat P, Kim S. Systematic Y-­X. Radiotherapy and immunotherapy: a 23. Ko EC, Raben D, Formenti SC. The
review of case reports on the abscopal effect. beneficial liaison? Nat Rev Clin Oncol 2017; integration of radiotherapy with
Curr Probl Cancer 2016; 40: 25–37. doi: 14: 365–79. doi: https://​doi.​org/​10.​1038/​ immunotherapy for the treatment of non-­
https://​doi.​org/​10.​1016/​j.​currproblcancer.​ nrclinonc.​2016.​211 small cell lung cancer. Clin Cancer Res 2018;
2015.​10.​001 16. Durante M, Reppingen N, Held KD. 24: 5792–806. doi: https://​doi.​org/​10.​1158/​
7. Kirkwood JM, Butterfield LH, Tarhini Immunologically augmented cancer 1078-​0432.​CCR-​17-​3620
AA, Zarour H, Kalinski P, Ferrone S. treatment using modern radiotherapy. Trends 24. Siegel RL, Miller KD, Jemal A. Cancer
Immunotherapy of cancer in 2012. CA Mol Med 2013; 19: 565–82. doi: https://​doi.​ statistics, 2019. CA Cancer J Clin 2019; 69:
Cancer J Clin 2012; 62: 309–35. doi: https://​ org/​10.​1016/​j.​molmed.​2013.​05.​007 7–34. doi: https://​doi.​org/​10.​3322/​caac.​21551
doi.​org/​10.​3322/​caac.​20132 17. Van Limbergen EJ, De Ruysscher DK, 25. Antonia SJ, Villegas A, Daniel D, Vicente
8. Robert C, Schachter J, Long GV, Arance A, Olivo Pimentel V, Marcus D, Berbee M, D, Murakami S, Hui R, et al. Durvalumab
Grob JJ, Mortier L, et al. Pembrolizumab Hoeben A, et al. Combining radiotherapy after chemoradiotherapy in stage III non-­
versus ipilimumab in advanced melanoma. N with immunotherapy: the past, the present small-­cell lung cancer. N Engl J Med 2017;
Engl J Med 2015; 372: 2521–32. doi: https://​ and the future. Br J Radiol 2017; 90: 377: 1919–29. doi: https://​doi.​org/​10.​1056/​
doi.​org/​10.​1056/​NEJMoa1503093 20170157. doi: https://​doi.​org/​10.​1259/​bjr.​ NEJMoa1709937
9. Postow MA, Sidlow R, Hellmann MD, 20170157 26. Antonia SJ, Villegas A, Daniel D, Vicente D,
Longo DL. Immune-­Related adverse 18. Herrera FG, Bourhis J, Coukos G. Murakami S, Hui R, et al. Overall survival
events associated with immune checkpoint Radiotherapy combination opportunities with Durvalumab after chemoradiotherapy
blockade. N Engl J Med(eds 2018; 378: Leveraging immunity for the next oncology in stage III NSCLC. N Engl J Med 2018;
158–68. doi: https://​doi.​org/​10.​1056/​ practice. CA Cancer J Clin 2017; 67: 65–85. 379: 2342–50. doi: https://​doi.​org/​10.​1056/​
NEJMra1703481 doi: https://​doi.​org/​10.​3322/​caac.​21358 NEJMoa1809697
10. Whiteside TL, Demaria S, Rodriguez-­Ruiz 19. Twyman-­Saint Victor C, Rech AJ, Maity 27. Formenti SC, Rudqvist N-­P, Golden E,
ME, Zarour HM, Melero I. Emerging A, Rengan R, Pauken KE, Stelekati E, Cooper B, Wennerberg E, Lhuillier C, et al.
opportunities and challenges in cancer et al. Radiation and dual checkpoint Radiotherapy induces responses of lung
immunotherapy. Clin Cancer Res 2016; 22: blockade activate non-­redundant immune cancer to CTLA-4 blockade. Nat Med 2018;

5 of 7 birpublications.org/bjr Br J Radiol;93:20190224
BJR Durante and Formenti

24: 1845–51. doi: https://​doi.​org/​10.​1038/​ doi: https://​doi.​org/​10.​1016/​j.​ijrobp.​2018.​11.​ evolution of T-­cell repertoire in peripheral
s41591-​018-​0232-2 066 blood and tumor microenvironment of
28. Formenti SC. Optimizing dose per fraction: 37. Crittenden MR, Zebertavage L, Kramer patients with esophageal squamous cell
a new chapter in the story of the Abscopal G, Bambina S, Friedman D, Troesch V, carcinoma treated with preoperative
effect? Int J Radiat Oncol Biol Phys 2017; 99: et al. Tumor cure by radiation therapy and chemotherapy. Carcinogenesis 2018; 39:
677–9. doi: https://​doi.​org/​10.​1016/​j.​ijrobp.​ checkpoint inhibitors depends on pre-­ 1389–98. doi: https://​doi.​org/​10.​1093/​carcin/​
2017.​07.​028 existing immunity . Sci Rep 2018; 8: 7012. bgy116
29. Brooks ED, Chang JY. Time to abandon doi: https://​doi.​org/​10.​1038/​s41598-​018-​ 47. Proietti E, Moschella F, Capone I, Belardelli
single-­site irradiation for inducing abscopal 25482-w F. Exploitation of the propulsive force of
effects. Nat Rev Clin Oncol 2019; 16: 123–35. 38. Ritter S, Durante M. Heavy-­Ion induced chemotherapy for improving the response to
doi: https://​doi.​org/​10.​1038/​s41571-​018-​ chromosomal aberrations: a review. Mutat cancer immunotherapy. Mol Oncol 2012; 6:
0119-7 Res 2010; 701: 38–46. doi: https://​doi.​org/​10.​ 1–14. doi: https://​doi.​org/​10.​1016/​j.​molonc.​
30. Durante M, Brenner DJ, Formenti SC. Does 1016/​j.​mrgentox.​2010.​04.​007 2011.​11.​005
heavy ion therapy work through the immune 39. d'Alesio V, Pacelli R, Durante M, 48. Durante M. New challenges in high-­energy
system? Int J Radiat Oncol Biol Phys 2016; 96: Canale Cama G, Cella L, Gialanella G, et al. particle radiobiology. Br J Radiol 2014; 87:
934–6. doi: https://​doi.​org/​10.​1016/​j.​ijrobp.​ Lymph nodes in the irradiated field influence 20130626. doi: https://​doi.​org/​10.​1259/​bjr.​
2016.​08.​037 the yield of radiation-­induced chromosomal 20130626
31. Durante M, Paganetti H. Nuclear physics aberrations in lymphocytes from breast 49. Asaithamby A, Hu B, Chen DJ. Unrepaired
in particle therapy: a review. Rep Prog Phys cancer patients. Int J Radiat Oncol Biol Phys clustered DNA lesions induce chromosome
2016; 79: 096702. doi: https://​doi.​org/​10.​ 2003; 57: 732–8. doi: https://​doi.​org/​10.​1016/​ breakage in human cells. Proc Natl Acad Sci
1088/​0034-​4885/​79/​9/​096702 S0360-​3016(​03)​00664-3 U S A 2011; 108: 8293–8. doi: https://​doi.​org/​
32. Ling TC, Slater JM, Nookala P, Mifflin R, 40. Durante M, Yamada S, Ando K, Furusawa 10.​1073/​pnas.​1016045108
Grove R, Ly AMGrove R, Patyal B, Yang Y, Kawata T, Majima H, et al. Measurements 50. Löbrich M, Jeggo PA. The impact of a
G, et al. Analysis of intensity-­modulated of the equivalent whole-­body dose during negligent G2/M checkpoint on genomic
radiation therapy (IMRT), proton and 3D radiation therapy by cytogenetic methods. instability and cancer induction. Nat Rev
conformal radiotherapy (3D-­CRT) for Phys Med Biol 1999; 44: 1289–98. doi: https://​ Cancer 2007; 7: 861–9. doi: https://​doi.​org/​
reducing perioperative cardiopulmonary doi.​org/​10.​1088/​0031-​9155/​44/​5/​314 10.​1038/​nrc2248
complications in esophageal cancer patients. 41. Durante M, Yamada S, Ando K, Furusawa 51. Sage E, Harrison L. Clustered DNA
Cancers 2014; 6: 2356–68. doi: https://​doi.​ Y, Kawata T, Majima H, et al. X-­Rays vs. lesion repair in eukaryotes: relevance to
org/​10.​3390/​cancers6042356 carbon-­ion tumor therapy: cytogenetic mutagenesis and cell survival. Mutat Res
33. Heylmann D, Badura J, Becker H, Fahrer J, damage in lymphocytes. Int J Radiat Oncol 2011; 711(1–2): 123–33. doi: https://​doi.​org/​
Kaina B. Sensitivity of CD3/CD28-­stimulated Biol Phys 2000; 47: 793–8. doi: https://​doi.​ 10.​1016/​j.​mrfmmm.​2010.​12.​010
versus non-­stimulated lymphocytes to org/​10.​1016/​S0360-​3016(​00)​00455-7 52. Bukowska B, Karwowski BT. The Clustered
ionizing radiation and genotoxic anticancer 42. Hartel C, Nikoghosyan A, Durante M, DNA Lesions - Types, Pathways of Repair
drugs: key role of ATM in the differential Sommer S, Nasonova E, Fournier C, et al. and Relevance to Human Health. Curr Med
radiation response. Cell Death Dis 2018; 9: Chromosomal aberrations in peripheral Chem 2018; 25: 2722–35. doi: https://​doi.​org/​
1053. doi: https://​doi.​org/​10.​1038/​s41419-​ blood lymphocytes of prostate cancer 10.​2174/​0929​8673​2566​6180​2261​10502
018-​1095-7 patients treated with IMRT and carbon ions. 53. Sato H, Niimi A, Yasuhara T, Permata TBM,
34. Venkatesulu BP, Mallick S, Lin SH, Krishnan Radiother Oncol 2010; 95: 73–8. doi: https://​ Hagiwara Y, Isono M, et al. Dna double-­
S. A systematic review of the influence of doi.​org/​10.​1016/​j.​radonc.​2009.​08.​031 strand break repair pathway regulates PD-­L1
radiation-­induced lymphopenia on survival 43. Pignalosa D, Lee R, Hartel C, Sommer S, expression in cancer cells. Nat Commun
outcomes in solid tumors. Crit Rev Oncol Nikoghosyan A, Debus J, et al. Chromosome 2017; 8. doi: https://​doi.​org/​10.​1038/​s41467-​
Hematol 2018; 123(January): 42–51. doi: inversions in lymphocytes of prostate cancer 017-​01883-9
https://​doi.​org/​10.​1016/​j.​critrevonc.​2018.​01.​ patients treated with x-­rays and carbon ions. 54. Wang W, Chapman NM, Zhang B, Li M,
003 Radiother Oncol 2013; 109: 256–61. doi: Fan M, Laribee RN, et al. Upregulation of
35. Yovino S, Kleinberg L, Grossman SA, https://​doi.​org/​10.​1016/​j.​radonc.​2013.​09.​021 PD-­L1 via HMGB1-­Activated IRF3 and
Narayanan M, Ford E. The etiology of 44. Fang P, Shiraishi Y, Verma V, Jiang W, Song NF-κB contributes to UV radiation-­induced
treatment-­related lymphopenia in patients J, Hobbs BP, et al. Lymphocyte-­Sparing immune suppression. Cancer Res 2019; 79:
with malignant gliomas: modeling radiation effect of proton therapy in patients with 2909–22. doi: https://​doi.​org/​10.​1158/​0008-​
dose to circulating lymphocytes explains esophageal cancer treated with definitive 5472.​CAN-​18-​3134
clinical observations and suggests methods chemoradiation. Int J Part Ther 2017; 4: 55. Averbeck NB, Ringel O, Herrlitz M, Jakob
of modifying the impact of radiation on 23–32. doi: https://​doi.​org/​10.​14338/​IJPT-​17-​ B, Durante M, Taucher-­Scholz G. Dna end
immune cells. Cancer Invest 2013; 31: 140–4. 00033.1 resection is needed for the repair of complex
doi: https://​doi.​org/​10.​3109/​07357907.​2012.​ 45. Davuluri R, Jiang W, Fang P, Xu C, Komaki lesions in G1-­phase human cells. Cell Cycle
762780 R, Gomez DR, et al. Lymphocyte nadir and 2014; 13: 2509–16. doi: https://​doi.​org/​10.​
36. Lobachevsky PN, Ventura J, esophageal cancer survival outcomes after 4161/​15384101.​2015.​941743
Giannakandropoulou L, Forrester H, chemoradiation therapy. Int J Radiat Oncol 56. Shevtsov M, Sato H, Multhoff G, Shibata A.
Palazzolo JS, Haynes NM, et al. A functional Biol Phys 2017; 99: 128–35. doi: https://​doi.​ Novel approaches to improve the efficacy of
immune system is required for the systemic org/​10.​1016/​j.​ijrobp.​2017.​05.​037 Immuno-­Radiotherapy. Front Oncol 2019; 9:
genotoxic effects of localized irradiation. Int 46. Zhang C, Palashati H, Tan Q, Ku W, Miao 156. doi: https://​doi.​org/​10.​3389/​fonc.​2019.​
J Radiat Oncol Biol Phys 2019; 103: 1184–93. Y, Xiong H, et al. Immediate and substantial 00156

6 of 7 birpublications.org/bjr Br J Radiol;93:20190224
Charged particles and immunotherapy BJR

57. Paludan SR, Reinert LS, Hornung V. Dna-­ 64. Corrales L, Matson V, Flood B, Spranger S, e1339857. doi: https://​doi.​org/​10.​1080/​
Stimulated cell death: implications for host Gajewski TF. Innate immune signaling and 2162402X.​2017.​1339857
defence, inflammatory diseases and cancer. regulation in cancer immunotherapy. Cell 73. Durante M, Formenti SC. Radiation-­
Nat Rev Immunol 2019; 19: 141–53. doi: Res 2017; 27: 96–108. doi: https://​doi.​org/​10.​ Induced chromosomal aberrations and
https://​doi.​org/​10.​1038/​s41577-​018-​0117-0 1038/​cr.​2016.​149 immunotherapy: micronuclei, cytosolic
58. Vitale I, Galluzzi L, Castedo M, Kroemer 65. Mackenzie KJ, Carroll P, Martin C-­A, Murina DNA, and Interferon-­Production pathway.
G. Mitotic catastrophe: a mechanism for O, Fluteau A, Simpson DJ, et al. cGAS Front Oncol 2018; 8: 192. doi: https://​doi.​org/​
avoiding genomic instability. Nat Rev Mol surveillance of micronuclei links genome 10.​3389/​fonc.​2018.​00192
Cell Biol 2011; 12: 385–92. doi: https://​doi.​ instability to innate immunity. Nature 2017; 74. Shimokawa T, Ma L, Ando K, Sato K, Imai
org/​10.​1038/​nrm3115 548: 461–5. doi: https://​doi.​org/​10.​1038/​ T. The future of combining Carbon-­Ion
59. Lotze MT, Tracey KJ. High-­Mobility group nature23449 radiotherapy with immunotherapy: evidence
box 1 protein (HMGB1): nuclear weapon 66. Harding SM, Benci JL, Irianto J, Discher DE, and progress in mouse models. Int J Part
in the immune arsenal. Nat Rev Immunol Minn AJ, Greenberg RA. Mitotic progression Ther 2016; 3: 61–70. doi: https://​doi.​org/​10.​
2005; 5: 331–42. doi: https://​doi.​org/​10.​1038/​ following DNA damage enables pattern 14338/​IJPT-​15-​00023.1
nri1594 recognition within micronuclei. Nature 2017; 75. Ando K, Fujita H, Hosoi A, Ma L,
60. Ebner DK, Tinganelli W, Helm A, Bisio 548: 466–70. doi: https://​doi.​org/​10.​1038/​ Wakatsuki M, Seino K-­I, et al. Intravenous
A, Yamada S, Kamada T, et al. The nature23470 dendritic cell administration enhances
immunoregulatory potential of particle 67. Chen Q, Sun L, Chen ZJ. Regulation and suppression of lung metastasis induced by
radiation in cancer therapy. Front Immunol function of the cGAS-­STING pathway of carbon-­ion irradiation. J Radiat Res 2017;
2017; 8: 99. doi: https://​doi.​org/​10.​3389/​ cytosolic DNA sensing. Nat Immunol 2016; 58: 446–55. doi: https://​doi.​org/​10.​1093/​jrr/​
fimmu.​2017.​00099 17: 1142–9. doi: https://​doi.​org/​10.​1038/​ni.​ rrx005
61. Gameiro SR, Malamas AS, Bernstein MB, 3558 76. Sato K, Nitta N, Aoki I, Imai T, Shimokawa
Tsang KY, Vassantachart A, Sahoo N, et al. 68. Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, T. Repeated photon and C-­ion irradiations
Tumor cells surviving exposure to proton Kroemer G. Type I interferons in anticancer in vivo have different impact on alteration of
or photon radiation share a common immunity. Nat Rev Immunol 2015; 15: tumor characteristics. Sci Rep 2018; 8: 1458.
immunogenic modulation signature, 405–14. doi: https://​doi.​org/​10.​1038/​nri3845 doi: https://​doi.​org/​10.​1038/​s41598-​018-​
rendering them more sensitive to T cell-­ 69. Galluzzi L, Vanpouille-­Box C, Bakhoum 19422-x
mediated killing. Int J Radiat Oncol Biol Phys SF, Demaria S. Snapshot: cGAS-­STING 77. Ebner DK, Tinganelli W, Helm A, Bisio A,
2016; 95: 120–30. doi: https://​doi.​org/​10.​ signaling. Cell 2018; 173: 276–276.e1. doi: Simoniello P, Natale F, et al. Generating and
1016/​j.​ijrobp.​2016.​02.​022 https://​doi.​org/​10.​1016/​j.​cell.​2018.​03.​015 grading the abscopal effect: proposal for
62. Onishi M, Okonogi N, Oike T, Yoshimoto 70. Vanpouille-­Box C, Demaria S, Formenti comprehensive evaluation of combination
Y, Sato H, Suzuki Y, et al. High linear SC, Galluzzi L. Cytosolic DNA sensing in immunoradiotherapy in mouse models.
energy transfer carbon-­ion irradiation organismal tumor control. Cancer Cell 2018; Transl Cancer Res 2017; 6(S5): S892–S899.
increases the release of the immune 34: 361–78. doi: https://​doi.​org/​10.​1016/​j.​ doi: https://​doi.​org/​10.​21037/​tcr.​2017.​06.​01
mediator high mobility group box 1 from ccell.​2018.​05.​013 78. Durante M. Proton beam therapy in Europe:
human cancer cells. J Radiat Res 2018; 59: 71. Vanpouille-­Box C, Alard A, Aryankalayil more centres need more research. Br J Cancer
541–6. doi: https://​doi.​org/​10.​1093/​jrr/​ MJ, Sarfraz Y, Diamond JM, Schneider 2019; ; 120: 777–8. doi: https://​doi.​org/​10.​
rry049 RJ, et al. Dna exonuclease TREX1 1038/​s41416-​018-​0329-x
63. Suzuki Y, Mimura K, Yoshimoto Y, regulates radiotherapy-­induced tumour 79. Paganetti H. Relative biological effectiveness
Watanabe M, Ohkubo Y, Izawa S, et al. immunogenicity. Nat Commun 2017; (RBE) values for proton beam therapy.
Immunogenic tumor cell death induced 8: 15618. doi: https://​doi.​org/​10.​1038/​ variations as a function of biological
by chemoradiotherapy in patients with ncomms15618 endpoint, dose, and linear energy transfer.
esophageal squamous cell carcinoma. Cancer 72. Vanpouille-­Box C, Formenti SC, Demaria S. Phys Med Biol 2014; 59: R419–R472. doi:
Res 2012; 72: 3967–76. doi: https://​doi.​org/​ Trex1 dictates the immune fate of irradiated https://​doi.​org/​10.​1088/​0031-​9155/​59/​22/​
10.​1158/​0008-​5472.​CAN-​12-​0851 cancer cells. Oncoimmunology 2017; 6: R419

7 of 7 birpublications.org/bjr Br J Radiol;93:20190224

You might also like