Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 7/2017

Open Access 30.03.2017 | Review

Cancer immunotherapy: how low-level ionizing radiation can play a key role

verfasst von: Marek K. Janiak, Marta Wincenciak, Aneta Cheda, Ewa M. Nowosielska, Edward J. Calabrese

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 7/2017

Abstract

The cancer immunoediting hypothesis assumes that the immune system guards the host against the incipient cancer, but also “edits” the immunogenicity of surviving neoplastic cells and supports remodeling of tumor microenvironment towards an immunosuppressive and pro-neoplastic state. Local irradiation of tumors during standard radiotherapy, by killing neoplastic cells and generating inflammation, stimulates anti-cancer immunity and/or partially reverses cancer-promoting immunosuppression. These effects are induced by moderate (0.1–2.0 Gy) or high (>2 Gy) doses of ionizing radiation which can also harm normal tissues, impede immune functions, and increase the risk of secondary neoplasms. In contrast, such complications do not occur with exposures to low doses (≤0.1 Gy for acute irradiation or ≤0.1 mGy/min dose rate for chronic exposures) of low-LET ionizing radiation. Furthermore, considerable evidence indicates that such low-level radiation (LLR) exposures retard the development of neoplasms in humans and experimental animals. Here, we review immunosuppressive mechanisms induced by growing tumors as well as immunomodulatory effects of LLR evidently or likely associated with cancer-inhibiting outcomes of such exposures. We also offer suggestions how LLR may restore and/or stimulate effective anti-tumor immunity during the more advanced stages of carcinogenesis. We postulate that, based on epidemiological and experimental data amassed over the last few decades, whole- or half-body irradiations with LLR should be systematically examined for its potential to be a viable immunotherapeutic treatment option for patients with systemic cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00262-017-1993-z) contains supplementary material, which is available to authorized users.
Abstract entitled “Cancer immunoedition modified by low-level ionizing radiation” was submitted to the International Conference on Medical Physics, August 03–05, 2015 Birmingham, UK; abstract is available at the following link: https://​www.​omicsonline.​org/​2155-9619/​2155-9619.​S1.​003-015.​pdf.
Abkürzungen
ADCC
Antibody-dependent cellular cytotoxicity
DAMP
Damage-associated molecular pattern
Gy
Gray (the SI unit of absorbed dose defined as the absorption of 1 J of the radiation energy per 1 kg of matter)
HBI
Half-body irradiation
HMGB1
High-mobility group box 1 protein
IR
Ionizing radiation
LET
Linear energy transfer
LLR
Low-level radiation
LNT
Linear, no threshold
M1, M2
Macrophage phenotypes 1 and 2
MC
Mast cell
mGy
Milligray (0.001 Gy)
N1, N2
Neutrophil phenotypes 1 and 2
NHL
Non-Hodgkin’s lymphoma
NKG2DL
Ligand for the natural killer group 2D receptor
NKT
Natural killer T lymphocyte
NOD
Nucleotide-binding oligomerization domain
PGE2
Prostaglandin E2
RT
Radiotherapy
Th
Helper T lymphocyte
Treg
Regulatory T lymphocyte
VEGF
Vascular endothelial growth factor
WBI
Whole-body irradiation

Introduction

The immune system is a crucial player in the organism’s control over the development of neoplasms (reviewed in [1]). After years of controversies, the early concept of cancer immunological surveillance [2, 3], whereby specifically stimulated (adaptive) immunity wards off proliferation of neoplastically transformed cells, has now been incorporated into the modern cancer immunoediting process. During the three phases of this process, the anti-neoplastic immune functions and immunogenicity of cancer cells are being gradually “edited”, so that the immune system protects the host against the development of a malignancy during the initial “elimination” phase, but later, during the following “equilibrium” and, especially, “escape” phases, morphs into an active supporter of cancer progression. Consequently, the emerging tumor not only evades immune recognition and destruction, but also actively contributes to remodeling of its microenvironment towards the immunosuppressive and pro-neoplastic state [410].
The improved understanding of the relationship between a growing tumor and the immune system has shed new light on the recently acknowledged complex interactions of ionizing radiation (IR) with cancer-related immunity. This, in turn, has led to the development of novel radiotherapeutic schemes based on the notion that local exposures at moderate (between 0.1 and 2.0 Gy absorbed during acute exposures) or even high doses (over 2.0 Gy) of radiation can, especially in combination with standard immunotherapy, stimulate various anti-neoplastic immune reactions, and/or reverse their suppressive state. These effects are thought to result from the radiation-induced immunogenic types of cell death, local inflammation, and tissue injury, all leading to the emergence of “danger signals” which prompt activities of the non-specific (innate) immune system; extensive recapitulation of the immunomodulatory effects of local radiotherapy (RT) has recently been summarized in a number of excellent reviews [1120]. However, even moderate radiotherapeutic doses are potentially harmful to the surrounding normal tissues, which can cause immunosuppression and/or induce secondary cancers [2123]. Such complications are highly unlikely after exposures to low doses (≤0.1 Gy absorbed within a short time or ≤0.1 mGy/min dose rate applied during a protracted exposure) of low linear energy transfer (LET) IR, referred to in this paper as low-level radiation (LLR). Indeed, the effects of exposures to LLR, including modulation of the immune functions, can qualitatively and quantitatively differ from those induced by moderate-to-high doses of low-LET radiation [2429].
The present paper indentifies and evaluates epidemiological as well as animal studies which indicate that exposures to LLR can inhibit or retard the development of primary and metastatic cancers [27, 3091]. This evaluation will include an assessment of possible mechanisms by which such protective effects may be mediated including: LLR-induced scavenging of reactive chemical intermediates, stimulation of the repair of the DNA damage, mitigation of inflammation, triggering of selective apoptosis or senescence of aberrant cells, and the up-regulation of both the innate and adaptive arms of the anti-cancer immune system [25, 9295]. Since enhancing anti-neoplastic immunity may be an important mechanism of the cancer-inhibitory effects of LLR [93101], clinical trials of whole- or half-body irradiations (WBI or HBI) with LLR are also evaluated [102106].
This paper will also assess how LLR can affect and modify advanced phases of cancer development resulting in a reversal of suppressed immune functions and/or restoration of the susceptibility of cancer cells to the assaults by immune effectors. However, in contrast to the extensively reviewed relations between moderate- and high-dose RT and the response of the immune system, recapitulations of the similar effects of LLR in the context of their clinical exploitation are virtually nonexistent. The present paper will complement and extend a recent review of the vast pre-clinical evidence of the LLR-induced protective/adaptive response in normal but not neoplastic tissues, which provides arguments for the trials of the LLR-based therapy of cancer [29].

Immunosuppressive tumor microenvironment

The concept that in vertebrates, elements of the immune system specifically recognize and eliminate incipient neoplastic cells and protect thereby against the development of overt malignancy dates back to late 1950s [2, 3]. In accordance with this “cancer immunosurveillance” hypothesis, it was demonstrated that both immuno-compromised human patients and experimental animals are at increased risk of developing various neoplasms (reviewed in [107]). However, investigations by Stutman showed that chemically induced sarcomas or adenomas do not develop more often in athymic, T-cell-deficient, nude mice than in their wide-type, immunocompetent counterparts [108]. This observation seriously challenged the cancer immunosurveillance model and almost led to its abandonment [7]. Yet, evidence accumulated in recent years has helped to explain what was wrong with the original cancer immunosurveillance hypothesis and why some neoplasms progress to their clinical stage. Thus, it was found that innate immunity initially senses the presence of transformed cells and exercises the first line of anti-cancer defense. Soon after the activation, elements of the innate immune system promote induction of adaptive (specific) anti-tumor responses. However, owing to genetic and epigenetic changes in the developing neoplastic cells, tumors may become “invisible” to immune effectors through loss or aberrant expression of the MHC class I antigens (reviewed in [109, 110]) or of other molecules on cancer cells involved in triggering of the innate and/or adaptive immune responses [111, 112]. For example, a change in hydrophobicity of tumor cells may lead to suppressed expression of the “damage-associated molecular pattern” (DAMP) molecules necessary to alert the innate immune system to a “danger” incurred by the presence of aberrant cells [113]. Notably, even the “danger signals”, such as high-mobility group box 1 protein (HMGB1), can actually support cancer growth through stimulation of myeloid-derived suppressor cells (MDSCs) [114] or nurse-like cells [115] that create conditions favorable for cancer progression. Furthermore, tumor-associated specific antigens may assume forms similar to those expressed on normal cells and evade recognition as “non-self” by the immune system (reviewed in [116]).
Developing tumors create microenvironments that not only support neoplastic growth and metastasis, but also significantly reduce the effectiveness and corrupt the functions of both the innate and adaptive arms of anti-cancer immunity [10]. Among the immunosuppressive components of tumor microenvironments are various soluble factors such as IL-10, TGF-β, vascular endothelial growth factor (VEGF), prostaglandin E2 (PGE2), HMGB1, indoleamine-2,3-dioxygenase (IDO), as well as soluble forms of phosphatidylserine, Fas receptors, and MHC class I-related chain A proteins (reviewed in [117, 118]). Another recently recognized immunosuppressive mechanism involves the activation of the so-called immune checkpoints whose function is to prevent overstimulation of the immune system (reviewed in [119, 120]). The two most important immune checkpoint co-inhibitory molecules likely to play a role in induction and maintenance of the immunosuppressive state within tumors are members of the immunoglobulin gene superfamily, the cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) whose expression on T helper cells suppresses the activity of cytolytic CD8+ T lymphocytes, and the programmed death 1 (PD-1; CD279) receptor primarily expressed on tumor-infiltrating lymphocytes (TILs) and monocytes which, upon combining with its respective ligands (PDL-1 and PDL-2), negatively regulates the anti-neoplastic function of T cells [121, 122]; in addition, the PD-1:PDL-1 interaction may promote the development and function of regulatory T (Treg) cells [123].
Active immunosuppression is also exerted by many non-specific and specific cellular effectors residing in or attracted to neoplastic tissue. Many different cells capable of inhibiting anti-cancer immunity and promoting cancer growth have now been identified. These include Treg lymphocytes [125, 126], MDSCs [127130], macrophages [128, 131133], natural killer T (NKT) [134136], Th17 [137139] and B lymphocytes [140143], but also neutrophils [131, 144147], dendritic cells (DCs) [148151], mast cells (MCs) [152], and mesenchymal stem cells [153155].
It has been finally well established that persistent activation of pro-inflammatory immunity facilitates cellular transformation and promotes tumor advancement. Unlike acute transient inflammatory responses which attract and activate elements of the innate immune system, chronic inflammation not only supports cancer progression, but also prevents the host from mounting effective immune defenses against it [129, 156162]. An intermediate role in this process of the inflammation-driven type 2 immune response is played by MDSCs which are attracted to inflammatory sites and facilitate tumor growth [163, 164]. Chronic inflammation, as a powerful driver of carcinogenesis, is associated with aberrant signaling mediated by the nucleotide-binding oligomerization domain (NOD)-like receptors expressed on DCs, macrophages, and lymphocytes [165, 166]. Critical immunosuppressive mechanisms operating in the tumor microenvironment during the advanced stages of carcinogenesis are outlined in Fig. 1.

Anti-neoplastic and immunomodulatory effects of LLR

Overview

The development and progression of cancer in both humans and laboratory animals can be suppressed or prevented by exposures to LLR. The results of about 40 epidemiological studies published since 1987 have demonstrated decreased or unaltered cancer incidence or mortality rates in human populations exposed to LLR during medical diagnostic tests and therapy, in the course of professional activities, or as residents of geographical areas and homes with elevated levels of natural background radiation (evidence presented in Supplementary Table 1). Likewise, between 1996 and 2014, at least 27 reports were published from controlled experiments carried out in mice, rats, and dogs, as well as in cultured cells demonstrating that single, multiple, or chronic irradiations with LLR exert anti-neoplastic activities and markedly inhibit the growth and/or advancement of spontaneous or induced tumors (evidence presented in Supplementary Table 2). In general, the results of both epidemiological and experimental studies indicate or suggest that, in the case of short-term exposures at a high-dose rate, the upper threshold for the control of tumor growth is around 0.1 Gy [25, 6163, 71, 167169]. As evidenced by the results of experimental studies conducted in the in vivo and in vitro systems, one of the most important underlying mechanisms of such tumor-inhibitory effects is up-regulation of both the innate and adaptive immunity. Numerous reports published between 1988 and 2014 indicate that exposures to LLR are potent stimulators of various anti-neoplastic functions of the immune system, including inhibition of inflammation and/or up-regulation of anti-inflammatory cytokines (evidence presented in Supplementary Table 3 and reviewed in [74, 80, 94, 170, 171]).

Specific studies demonstrating anti-tumor effects by LLR

There are also a number of reports dating back to early 1980s which demonstrate association of the LLR-induced up-regulation of anti-neoplastic immunity with inhibition of cancer development:
1.
In 1982, Robert Anderson and collaborators [172] were among the first to report retardation of the growth of transplanted tumors in A/J mice following WBI with X-rays at doses ranging from 0.005 to 0.025 Gy immediately prior to s.c. inoculation of Sarcoma I cells. The evidence clearly suggested the involvement of “a very radiosensitive T cell with suppressor activity”.
 
2.
In 1994, Kharazi et al. showed that chronic low-dose WBI with γ-rays (0.04 Gy per exposure, three times per week for 4 weeks) when combined with caloric restriction enhanced the regression of mammary tumors spontaneously developing in female C3H/He mice. These tumors were massively infiltrated with cytotoxic CD8+ T cells. Such tumor regression did not occur in mice subjected to caloric restriction alone [173].
 
3.
As reported in 1999 by Hashimoto et al., a single WBI at 0.2 Gy of γ-rays of WKHA rats injected with hepatoma cells led to a significant reduction in the number of lung and lymph node metastases accompanied by the markedly stimulated influx of CD8+ lymphocytes into the spleen and the tumor site along with the enhanced expression of mRNAs for IFN-γ and TNF-α and down-regulation of mRNA for TGF-β; no mRNAs for IL-4, IL-6, and IL-10, the Th2-type cytokines that inhibit the anti-tumor Th1 responses, were detected in these tissues [69].
 
4.
The studies by Yu et al. showed that a single exposure of male Kunming mice (a strain similar to C57BL/6 mice) to 0.075 Gy X-rays 6 h before implantation of S180 sarcoma cells significantly inhibited tumor growth accompanied by the influx of TILs as well as enhanced necrosis and down-regulation of the expression of receptors for VEGF in the neoplastic tissue [73, 74].
 
5.
Continuous irradiation of C57BL/6 mice with γ-rays at 1.2 mGy/h for 258 days (up to 7.2 Gy total dose) did not induce thymic lymphomas, whereas the same total dose absorbed during four acute exposures to X-rays at 1.8 Gy resulted in the appearance of the lymphomas in 90% of these animals; in the continuously irradiated mice, the numbers of CD4+ T cells and antibody-producing B cells were significantly enhanced in the spleen [75].
 
6.
Continuous exposure to γ-rays of the lymphoma-prone SJL/J mice at 100 mGy/y dose rate slightly prolonged life span of the animals and the effect was accompanied by the significant increase in the percentages of CD49+ NK cells and decreased percentages of CD4+ and CD8+ lymphocytes in the spleen [174]. When spleens of rats with a diethylnitrosamine-induced liver cancer were irradiated at 0.15 Gy from the 6 MeV β-beam accelerator at 100 mGy/min dose rate, the percentage of CD4+CD25+ Treg cells in the blood significantly decreased and the levels of Foxp3, IL-10, TGF-β, and CTLA-4 were down-regulated in the spleen and the tumor; these changes were accompanied by the suppressed tumor growth [175].
 
7.
Experimental combinations of low-level WBI with the conventional (intermediate- or high-dose) local RT also yielded promising results: using murine tumor models of B16 melanoma and Lewis lung carcinoma, Liu and collaborators demonstrated that when fractionated local X-ray irradiations of the tumors at 2 Gy/fraction were several times substituted for WBI at 0.075 Gy, the cancer control (as judged by the reduced tumor mass and pulmonary metastases as well as by the increased survival of the hosts) was significantly improved compared to local RT alone; this effect was accompanied by up-regulation of the activities of the splenic NK and cytotoxic T lymphocytes which secreted elevated amounts of IFN-γ and TNF-α [77, 78].
 

Our strategies showing anti-tumor effects by LLR

In a series of our own experiments carried out in the relatively radiosensitive BALB/c mice and the relatively radioresistant C57BL/6 mice, both single and multiple WBI with X-rays at total doses ranging from 0.05 to 0.2 Gy reproducibly suppressed development of the induced neoplastic colonies in the lungs. Since the mice were whole-body irradiated before inoculation of the syngeneic tumor cells, the obvious suggestion was that the low-level X-ray exposures stimulated systemic innate anti-neoplastic reactions. Although we were not able to directly estimate the activities of immune cells in the lungs, a significant stimulation of the cytotoxic activities of NK cells and LPS- and IFN-γ-stimulated macrophages obtained from the spleen and peritoneal cavity, respectively, was detected in the X-ray-exposed mice from both strains. Interestingly, no elevation of the activities of these cells was detected after their in vitro irradiation at the same doses of X-rays indicating that enhancing of the NK- and macrophage-mediated cytolytic functions by LLR depends on the presence of factors occurring in in vivo but not the in vitro conditions [8190, 176179].

Clinical trials

The above-described epidemiological and experimental observations of anti-neoplastic and immunomodulatory effects of LLR exposures provide grounds for clinical trials with WBI or HBI of oncological patients [101, 180]. Even before the aforementioned evidence gained significance, a few LLR-based therapy trials had been performed. In 1965, Holder reported on positive therapeutic effects of the low-level total-body irradiation of patients with multiple myeloma [181]. In 1975, Kazem described curative effects of WBI (0.15 Gy of γ-rays daily for the first 5 days and thereafter at 0.1–0.15 Gy every other day or at longer intervals to the total doses of 2.0–2.65 Gy applied over 5–12 weeks) of patients with disseminated stage III lymphomas [182]. Likewise, Chaffey et al. obtained complete remissions in 32 out of 40 patients with advanced lymphocytic lymphoma after repeated WBI (0.15 Gy twice a week to a total dose of 1.5 Gy) as an initial and only primary therapy [102]. Very promising results of low-level total-body exposures to γ-rays of patients with non-Hodgkin’s lymphoma (NHL) were also reported by Qasim [183] and Choi et al. [103]. In one of the later trials, 24 out of 26 patients with stage IV low grade NHL were in complete remission after two courses of low-dose, total-body irradiation at 0.75 Gy given in five fractions; when the initially pathological lymph node areas of these patients were 1 month later treated with the conventional RT (total dose of 40 Gy applied in 20 fractions), the disease remitted in yet another patient [184]. Similarly, Safwat et al. who used low-level total-body exposures (0.1–0.25 Gy several times a week to the total dose of 1.5–2.0 Gy) obtained complete remissions in 11 out of 35 patients and 2-year progression-free survival in 12 patients with relapsed and/or chemo-resistant NHL; in 14 patients, a significant increase in the percentage of CD4+ T cells in the blood was noted [105]. In addition, as demonstrated by Sakamoto et al., low-dose HBI with X-rays (0.1–0.15 Gy two times a week for 5 weeks) combined with local RT (2 Gy five times a week for 6 weeks) resulted in the 5-year survival of 84% of patients with stage I and II NHL as compared to 65% survival of patients treated solely with local RT (the difference significant at p < 0.05); in these patients, percentages of peripheral blood CD4+ T helper lymphocytes were significantly elevated [100].
While more clinical trials employing WBI or HBI with LLR are needed, they are hampered by radiation safety regulations based on the linear, no threshold (LNT) model of the dose–effect relationship assuming that any absorbed dose of radiation causes a finite increase in cancer risk. There is a growing consensus that the LNT hypothesis lacks a solid experimental foundation and is based largely on ideology rather than science [25, 169, 185193]. Hopefully, the many recent appeals from radiobiologists, physicians, and health physicists to various regulatory bodies and authorities to base the radiation protection system on scientific data indicating that there are quantitative and qualitative differences between the effects of low doses delivered at low dose rates and high doses delivered at high-dose rates [171, 187, 188, 190, 192, 194, 195] will lead to a revision of current radiation protection regulations, so that WBI with LLR can be tested in clinical trials.

Suggested effects of LLR on cancer immunoediting process

As reviewed above, both acute and chronic exposures to LLR stimulate various anti-neoplastic immune reactions that are stifled or corrupted within the tumor microenvironment, especially during the later stages of carcinogenesis. Based on evidence indicating that tumor-inhibiting effects of LLR have been observed in both humans and experimental animals exposed in many different ways to single, multiple, and chronic irradiation with LLR, it may be argued that many, if not all, of the above-reviewed tumor-promoting immune mechanisms are likely to be blocked and/or reversed by such exposures (Fig. 2). Indeed, data indicating that LLR exposures may reverse the tumor-associated immune suppression has recently begun to emerge, even though many underlying LLR-induced mechanisms remain to be clarified. Based on the current evidence it may be postulated that, in addition to the direct activation of NK lymphocytes [83, 196, 197] and possibly other anti-tumor cytotoxic cells, LLR exposures enhance the “visibility” and/or susceptibility of cancer cells to immune assaults through stimulation of the expression by neoplastic and immune cells of molecules and ligands (e.g., CD2, B7, CD28, NKG2D) necessary for triggering of cytotoxic reactions [198200] and/or turning on “danger signals” in the neoplastic tissue [201, 202]. Furthermore, low-level radiation exposures are likely to alleviate or reverse the tumor-associated immune degeneracy through elimination or inhibition of the multiple cells, cytokines, and other factors associated with immunosuppressive loops induced by the tumor [175, 203207]. This could result in: (a) shifting of the immune response in favor of the anti-neoplastic phenotypes such as Th1 in the case of CD4+ T cells [97, 208], M1 in the case of macrophages [209, 210], and N1 in the case of neutrophils [211], (b) targeting the Treg-Th17 and Th17-DC interactions conducive to tumor regression [212214], (c) activation of the Toll-like receptor-mediated signaling in phagocytes and antigen-presenting cells [215217], (d) attenuation of the chronic inflammation pertinent to cancer initiation, promotion, and progression [94, 95, 170, 218, 219], and/or (e) down-regulation of the immune checkpoint molecules such as the CTLA-4, PD-1, and/or PD-L1 on T cells [198, 220222]. Indeed, one of the recent reports indicates that hypofractionated γ-ray irradiation of tumors induced in C57BL/6 mice combined with blockade of the PD-1 checkpoint stimulated accumulation of TILs associated with complete eradication of very large neoplasms [222]. In addition, there are numerous non-immune mechanisms triggered by LLR that positively affect normal, but not malignant cells [29]. These include: (a) increased cell proliferation, (b) stimulation of anti-oxidant reactions associated with the reduction of tissue injury, (c) improved repair of the DNA damage, and (d) metabolic shift from oxidative phosphorylation to aerobic glycolysis resulting in increased radioresistance of healthy tissues. Such outcomes are of primary importance for the combination of the LLR-based immunotherapy with classic forms of cancer therapy (i.e., high-dose RT and chemotherapy) that are lethal to normal cells and tissues and promote the formation of reactive oxygen species and inflammation. It is expected that other LLR-triggered reactions and mechanisms will be detected providing additional grounds for the use of the truly low-level exposures to IR in the treatment of cancer and, possibly, other diseases.

Conclusion and prospects

Cancer immunotherapy has matured from the application of several therapeutic agents, including tumor cell- and dendritic cell-based vaccines, anti-cytokine antibodies, checkpoint inhibitors, and genetically engineered T cells and stem cells, which collectively act to reverse immune suppression in the tumor environment and/or immune resistance of tumor cells (reviewed in [208]). There are also clinical trials combining such agents with local irradiation of tumors at moderate doses (i.e., >0.5–1.0 Gy per fraction) currently used in RT [16]. The recently acknowledged capacity of locally applied moderate or high (radiotherapeutic) doses of radiation to induce immunogenic death of cancer cells and local inflammatory reactions associated with stimulation of dendritic cells and enhancing the suppressed anti-cancer immunity has been employed as an adjuvant to improve the efficacy of existing immunotherapy protocols (reviewed in [1119, 21]). However, such exposures can also cause persistent inflammation and multiple cell death in normal tissues, impede various immune and other physiological functions, and increase the risk of secondary primary cancers. In contrast, LLR exposures do not kill or impair and actually support functions of normal cells and tissues, selectively eliminate precancerous and transformed cells, attenuate rather than induce chronic inflammation, stimulate various anti-neoplastic reactions of the immune system, and are not associated with the development of secondary malignancies [21, 29, 94, 95, 170]. Finally, as indicated by the above-reviewed results of experimental and epidemiological studies as well as several clinical trials, WBI or HBI with LLR are not likely to induce any untoward side effects and can thus be used in treatment of patients with systemic or metastatic cancer.
It is, therefore, time to employ whole- or half-body exposures to LLR (alone or as an adjuvant to conventional therapeutics) to restore the efficacy of systemic anti-cancer functions of the immune system, the most potent guardian against neoplasia. This approach is expected to mediate improved clinical responses in cancer patients, as well as protect normal tissues from the well-known adverse effects associated with standard chemo- and radiotherapy used in contemporary cancer therapeutics.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
3.
Zurück zum Zitat Thomas L (1959) Discussion in: Lawrence HS (ed) Cellular and humoral aspects of the hypersensitive states. Hoeber-Harper, New York, pp 529–532 Thomas L (1959) Discussion in: Lawrence HS (ed) Cellular and humoral aspects of the hypersensitive states. Hoeber-Harper, New York, pp 529–532
11.
Zurück zum Zitat Rödel F, Frey B, Gaipl U, Keilholz L, Fournier C, Manda K et al (2012) Modulation of inflammatory immune reactions by low-dose ionizing radiation: Molecular mechanisms and clinical applications. Curr Medic Chem 19:1741–1750. doi:10.2174/092986712800099866 CrossRef Rödel F, Frey B, Gaipl U, Keilholz L, Fournier C, Manda K et al (2012) Modulation of inflammatory immune reactions by low-dose ionizing radiation: Molecular mechanisms and clinical applications. Curr Medic Chem 19:1741–1750. doi:10.​2174/​0929867128000998​66 CrossRef
16.
Zurück zum Zitat Draghiciu O, Walczak M, Hoogeboom BN, Franken KL, Melief KJ, Nijman HW, Daemen T (2014) Therapeutic immunization and local low-dose tumour irradiation, a reinforcing combination. Int J Cancer 134:859–872. doi:10.1002/ijc.28418 PubMedCrossRef Draghiciu O, Walczak M, Hoogeboom BN, Franken KL, Melief KJ, Nijman HW, Daemen T (2014) Therapeutic immunization and local low-dose tumour irradiation, a reinforcing combination. Int J Cancer 134:859–872. doi:10.​1002/​ijc.​28418 PubMedCrossRef
17.
Zurück zum Zitat Frey B, Rubner Y, Kulzer L, Werthmöller N, Weiss EM, Fietkau R, Gaipl US (2014) Antitumour immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol Immunother 63:29–36. doi:10.1007/s00262-013-1474-y PubMedCrossRef Frey B, Rubner Y, Kulzer L, Werthmöller N, Weiss EM, Fietkau R, Gaipl US (2014) Antitumour immune responses induced by ionizing irradiation and further immune stimulation. Cancer Immunol Immunother 63:29–36. doi:10.​1007/​s00262-013-1474-y PubMedCrossRef
22.
Zurück zum Zitat Gudowska I, Ardenfors O, Toma-Dasu I, Dasu A (2014) Radiation burden from secondary doses to patients undergoing radiation therapy with photons and light ions and radiation doses from imaging modalities. Radiat Prot Dosim 161:357–362. doi:10.1093/rpd/nct335 CrossRef Gudowska I, Ardenfors O, Toma-Dasu I, Dasu A (2014) Radiation burden from secondary doses to patients undergoing radiation therapy with photons and light ions and radiation doses from imaging modalities. Radiat Prot Dosim 161:357–362. doi:10.​1093/​rpd/​nct335 CrossRef
24.
Zurück zum Zitat Albrecht H, Durbin-Johnson B, Yunis R, Kalanetra KM, Wu S, Chen R et al (2012) Transcriptional response of ex vivo human skin to ionizing radiation: comparison between low- and high-dose effects. Radiat Res 177:69–83. doi:10.1667/RR2524.1 PubMedCrossRef Albrecht H, Durbin-Johnson B, Yunis R, Kalanetra KM, Wu S, Chen R et al (2012) Transcriptional response of ex vivo human skin to ionizing radiation: comparison between low- and high-dose effects. Radiat Res 177:69–83. doi:10.​1667/​RR2524.​1 PubMedCrossRef
25.
Zurück zum Zitat Feinendegen LE, Pollycove M, Neumann RD (2013) Hormesis by low dose radiation effects: Low-dose cancer risk modeling must recognize up-regulation of protection In: Baum RP (ed) Therapeutic nuclear medicine. Springer, Berlin, pp 789–805 doi:10.1007/174_2012_686 Feinendegen LE, Pollycove M, Neumann RD (2013) Hormesis by low dose radiation effects: Low-dose cancer risk modeling must recognize up-regulation of protection In: Baum RP (ed) Therapeutic nuclear medicine. Springer, Berlin, pp 789–805 doi:10.​1007/​174_​2012_​686
26.
Zurück zum Zitat Yu H, Liu N, Wang H, Shang Q, Jiang P, Zhang Y (2013) Different responses of tumor and normal cells to low-dose radiation. Contemp Oncol (Pozn) 17:356–362. doi:10.5114/wo.2013.35289 Yu H, Liu N, Wang H, Shang Q, Jiang P, Zhang Y (2013) Different responses of tumor and normal cells to low-dose radiation. Contemp Oncol (Pozn) 17:356–362. doi:10.​5114/​wo.​2013.​35289
29.
Zurück zum Zitat Yang G, Li W, Jiang H, Liang X, Zhao Y, Yu D et al (2016) Low-dose radiation may be a novel approach to enhance the effectiveness of cancer therapeutics. Int J Cancer 139:2157–2168. doi:10.1002/ijc.30235 PubMedCrossRef Yang G, Li W, Jiang H, Liang X, Zhao Y, Yu D et al (2016) Low-dose radiation may be a novel approach to enhance the effectiveness of cancer therapeutics. Int J Cancer 139:2157–2168. doi:10.​1002/​ijc.​30235 PubMedCrossRef
30.
Zurück zum Zitat Rowland RE (1970) Dose and damage in long term radium cases. In Cloutier RJ, Edwards CL, Snyder WS (eds), Medical radionuclides: radiation dose and effects: Proceedings of a Symposium held at the Oak Ridge Associated Universities, Dec 8–11, 1969. Publ.: US Atomic Energy Comission, Division of Technical Information (available from Clearinghouse for Federal Scientific and Technical Information, Springfield, VA), pp 369–386 Rowland RE (1970) Dose and damage in long term radium cases. In Cloutier RJ, Edwards CL, Snyder WS (eds), Medical radionuclides: radiation dose and effects: Proceedings of a Symposium held at the Oak Ridge Associated Universities, Dec 8–11, 1969. Publ.: US Atomic Energy Comission, Division of Technical Information (available from Clearinghouse for Federal Scientific and Technical Information, Springfield, VA), pp 369–386
32.
Zurück zum Zitat Nambi KS, Soman SD (1990) Further observations on environmental radiation and cancer in India. Health Phys 59:339–344PubMed Nambi KS, Soman SD (1990) Further observations on environmental radiation and cancer in India. Health Phys 59:339–344PubMed
33.
Zurück zum Zitat Wei LX, Zha YR, Tao ZF, He WH, Chen DQ, Yuan YL (1990) Epidemiological investigation of radiological effects in high background radiation areas of Yangjiang, China. J Radiat Res 31:119–136. doi:10.1269/jrr.31.119 PubMedCrossRef Wei LX, Zha YR, Tao ZF, He WH, Chen DQ, Yuan YL (1990) Epidemiological investigation of radiological effects in high background radiation areas of Yangjiang, China. J Radiat Res 31:119–136. doi:10.​1269/​jrr.​31.​119 PubMedCrossRef
38.
42.
Zurück zum Zitat Cohen BL (2008) The linear no-threshold theory of radiation carcinogenesis should be rejected. JPandS 13:70–76 Cohen BL (2008) The linear no-threshold theory of radiation carcinogenesis should be rejected. JPandS 13:70–76
44.
Zurück zum Zitat Miller AB, Howe GR, Sherman GJ, Lindsay JP, Yaffe MJ, Dinner PJ et al (1989) Mortality from breast cancer after irradiation during fluoroscopic examinations in patients being treated for tuberculosis. New Eng J Med 321:1285–1289. doi:10.1056/NEJM198911093211902 PubMedCrossRef Miller AB, Howe GR, Sherman GJ, Lindsay JP, Yaffe MJ, Dinner PJ et al (1989) Mortality from breast cancer after irradiation during fluoroscopic examinations in patients being treated for tuberculosis. New Eng J Med 321:1285–1289. doi:10.​1056/​NEJM198911093211​902 PubMedCrossRef
48.
Zurück zum Zitat Mohan AK, Hauptmann M, Freedman DM, Ron E, Matanoski GM, Lubin JH et al (2003) Cancer and other causes of mortality among radiologic technologists in the United States. Int J Cancer 103:259–267. doi:10.1002/ijc.10811 PubMedCrossRef Mohan AK, Hauptmann M, Freedman DM, Ron E, Matanoski GM, Lubin JH et al (2003) Cancer and other causes of mortality among radiologic technologists in the United States. Int J Cancer 103:259–267. doi:10.​1002/​ijc.​10811 PubMedCrossRef
55.
Zurück zum Zitat Hwang SL, Guo HR, Hsieh WA, Hwang JS, Lee SD, Tang JL et al (2006) Cancer risks in a population with prolonged low dose-rate ɣ -radiation exposure in radiocontaminated buildings, 1983–2002. Int J Radiat Biol 82:849–858. doi:10.1080/09553000601085980 PubMedCrossRef Hwang SL, Guo HR, Hsieh WA, Hwang JS, Lee SD, Tang JL et al (2006) Cancer risks in a population with prolonged low dose-rate ɣ -radiation exposure in radiocontaminated buildings, 1983–2002. Int J Radiat Biol 82:849–858. doi:10.​1080/​0955300060108598​0 PubMedCrossRef
56.
Zurück zum Zitat Boice JD Jr, Mumma MT, Blot WJ (2007) Cancer and noncancer mortality in populations living near uranium and vanadium mining and milling operations in Montrose County, Colorado, 1950–2000. Radiat Res 167:711–726. doi:10.1667/RR0839.1 PubMedCrossRef Boice JD Jr, Mumma MT, Blot WJ (2007) Cancer and noncancer mortality in populations living near uranium and vanadium mining and milling operations in Montrose County, Colorado, 1950–2000. Radiat Res 167:711–726. doi:10.​1667/​RR0839.​1 PubMedCrossRef
57.
Zurück zum Zitat Cardis E, Vrijheid M, Blettner M, Gilbert E, Hakama M, Hill C et al (2007) The 15-Country collaborative study of cancer risk among radiation workers in the nuclear industry: estimates of radiation-related cancer risks. Radiat Res 167:396–416. doi:10.1667/RR0553.1 PubMedCrossRef Cardis E, Vrijheid M, Blettner M, Gilbert E, Hakama M, Hill C et al (2007) The 15-Country collaborative study of cancer risk among radiation workers in the nuclear industry: estimates of radiation-related cancer risks. Radiat Res 167:396–416. doi:10.​1667/​RR0553.​1 PubMedCrossRef
58.
Zurück zum Zitat Minister of Public Works and Government Services Canada, Canadian Nuclear Safety Commission (CNSC) (2011) Verifying Canadian nuclear energy worker radiation risk: A reanalysis of cancer mortality in Canadian nuclear energy workers (1957–1994) Summary report (2011). ISBN 978-1-100-17760-1 Minister of Public Works and Government Services Canada, Canadian Nuclear Safety Commission (CNSC) (2011) Verifying Canadian nuclear energy worker radiation risk: A reanalysis of cancer mortality in Canadian nuclear energy workers (1957–1994) Summary report (2011). ISBN 978-1-100-17760-1
61.
Zurück zum Zitat UNSCEAR (2008) Report (2011): Sources and Effects of Ionizing Radiation United Nations Scientific Committee on the Effects of Atomic Radiation Vol II: Effects, Scientific Annex D, United Nations, New York, pp 45–219 UNSCEAR (2008) Report (2011): Sources and Effects of Ionizing Radiation United Nations Scientific Committee on the Effects of Atomic Radiation Vol II: Effects, Scientific Annex D, United Nations, New York, pp 45–219
63.
Zurück zum Zitat Ivanov VK, Tsyb AF (2013) Thyroid cancer: lessons of Chernobyl and prognosis for Fukushima. Vestn Ross Akad Med Nauk (5):38–44. Ivanov VK, Tsyb AF (2013) Thyroid cancer: lessons of Chernobyl and prognosis for Fukushima. Vestn Ross Akad Med Nauk (5):38–44.
65.
Zurück zum Zitat Tubiana M, Diallo I, Chavaudra J, Lefkopoulos D, Bourhis J, Girinsky T et al (2011) A new method of assessing the dose-carcinogenic effect relationship in patients exposed to ionizing radiation A concise presentation of preliminary data. Health Phys 100:296–299. doi:10.1097/HP.0b013e31820a1b35 PubMedCrossRef Tubiana M, Diallo I, Chavaudra J, Lefkopoulos D, Bourhis J, Girinsky T et al (2011) A new method of assessing the dose-carcinogenic effect relationship in patients exposed to ionizing radiation A concise presentation of preliminary data. Health Phys 100:296–299. doi:10.​1097/​HP.​0b013e31820a1b35​ PubMedCrossRef
68.
Zurück zum Zitat Hashimoto S (1997) Effects of low-dose total body irradiation (TBI) on tumor-bearing rats. Nihon Igaku Hoshasen Gakkai Zasshi 57:418–424PubMed Hashimoto S (1997) Effects of low-dose total body irradiation (TBI) on tumor-bearing rats. Nihon Igaku Hoshasen Gakkai Zasshi 57:418–424PubMed
69.
Zurück zum Zitat Hashimoto S, Shirato H, Hosokawa M, Nishioka T, Kuramitsu Y, Matushita K et al (1999) The suppression of metastases and the change in host immune response after low-dose total-body irradiation in tumour-bearing rats. Radiat Res 151:717–724. doi:10.1016/S0360-3016(98)80333-7 PubMedCrossRef Hashimoto S, Shirato H, Hosokawa M, Nishioka T, Kuramitsu Y, Matushita K et al (1999) The suppression of metastases and the change in host immune response after low-dose total-body irradiation in tumour-bearing rats. Radiat Res 151:717–724. doi:10.​1016/​S0360-3016(98)80333-7 PubMedCrossRef
70.
Zurück zum Zitat Mitchel RE, Jackson JS, McCann RA, Boreham DR (1999) The adaptive response modifies latency for radiation-induced myeloid leukemia in CBA/H mice. Radiat Res 152:273–279. doi:10.2307/3580327 PubMedCrossRef Mitchel RE, Jackson JS, McCann RA, Boreham DR (1999) The adaptive response modifies latency for radiation-induced myeloid leukemia in CBA/H mice. Radiat Res 152:273–279. doi:10.​2307/​3580327 PubMedCrossRef
73.
Zurück zum Zitat Yu HS, Song AQ, Lu YD, Qiu WS, Shen FZ (2004) Effects of low-dose radiation on tumour growth, erythrocyte immune function and SOD activity in tumour-bearing mice. Chin Med J (Engl) 117:1036–1039 Yu HS, Song AQ, Lu YD, Qiu WS, Shen FZ (2004) Effects of low-dose radiation on tumour growth, erythrocyte immune function and SOD activity in tumour-bearing mice. Chin Med J (Engl) 117:1036–1039
75.
Zurück zum Zitat Ina Y, Tanooka H, Yamada T, Sakai K (2005) Suppression of thymic lymphoma induction by life-long low-dose-rate irradiation accompanied by immune activation in C57BL/6 mice. Radiat Res 163:153–158. doi:10.1667/RR3289 PubMedCrossRef Ina Y, Tanooka H, Yamada T, Sakai K (2005) Suppression of thymic lymphoma induction by life-long low-dose-rate irradiation accompanied by immune activation in C57BL/6 mice. Radiat Res 163:153–158. doi:10.​1667/​RR3289 PubMedCrossRef
76.
Zurück zum Zitat Ishii K, Hosoi Y, Yamada S, Ono T, Sakamoto K (1996) Decreased incidence of thymic lymphoma in AKR mice as a result of chronic, fractionated low-dose total-body X irradiation. Radiat Res 146:582–585. doi:10.2307/3579560 PubMedCrossRef Ishii K, Hosoi Y, Yamada S, Ono T, Sakamoto K (1996) Decreased incidence of thymic lymphoma in AKR mice as a result of chronic, fractionated low-dose total-body X irradiation. Radiat Res 146:582–585. doi:10.​2307/​3579560 PubMedCrossRef
79.
Zurück zum Zitat Ogura K, Magae J, Kawakami Y, Koana T (2009) Reduction in mutation frequency by very low-dose gamma irradiation of Drosophila melanogaster germ cells. Radiat Res 171:1–8. doi:10.1667/RR1288.1 PubMedCrossRef Ogura K, Magae J, Kawakami Y, Koana T (2009) Reduction in mutation frequency by very low-dose gamma irradiation of Drosophila melanogaster germ cells. Radiat Res 171:1–8. doi:10.​1667/​RR1288.​1 PubMedCrossRef
81.
Zurück zum Zitat Cheda A, Wrembel-Wargocka J, Lisiak E, Marciniak M, Nowosielska EM, Janiak MK (2004) Inhibition of the development of pulmonary tumour nodules and stimulation of the activity of NK cells and macrophages in mice by single low doses of low-LET radiation. Int J Low Radiat 1:171–179. doi:10.1504/IJLR.2004.003868 CrossRef Cheda A, Wrembel-Wargocka J, Lisiak E, Marciniak M, Nowosielska EM, Janiak MK (2004) Inhibition of the development of pulmonary tumour nodules and stimulation of the activity of NK cells and macrophages in mice by single low doses of low-LET radiation. Int J Low Radiat 1:171–179. doi:10.​1504/​IJLR.​2004.​003868 CrossRef
82.
Zurück zum Zitat Cheda A, Wrembel-Wargocka J, Lisiak E, Nowosielska EM, Marciniak M, Janiak MK (2004) Single low doses of X rays inhibit the development of experimental tumour metastases and trigger the activities of NK cells in mice. Radiat Res 161:335–340. doi:10.1667/RR3123 PubMedCrossRef Cheda A, Wrembel-Wargocka J, Lisiak E, Nowosielska EM, Marciniak M, Janiak MK (2004) Single low doses of X rays inhibit the development of experimental tumour metastases and trigger the activities of NK cells in mice. Radiat Res 161:335–340. doi:10.​1667/​RR3123 PubMedCrossRef
83.
Zurück zum Zitat Cheda A, Wrembel-Wargocka J, Nowosielska EM, Janiak MK (2006) Immune mechanism of the retarded growth of tumor nodules in mice exposed to single low-level irradiations with X-rays. Centr. Eur J Immunol 31:44–50 Cheda A, Wrembel-Wargocka J, Nowosielska EM, Janiak MK (2006) Immune mechanism of the retarded growth of tumor nodules in mice exposed to single low-level irradiations with X-rays. Centr. Eur J Immunol 31:44–50
84.
Zurück zum Zitat Janiak MK, Wrembel-Wargocka J, Cheda A, Nowosielska EM, Lisiak E, Bilski M (2006) Modulation of anti-tumour functions of NK cells and macrophages after single low-level exposures of mice to X-rays. Int J Low Radiat 3:178–191. doi:10.1504/IJLR.2006.012017 CrossRef Janiak MK, Wrembel-Wargocka J, Cheda A, Nowosielska EM, Lisiak E, Bilski M (2006) Modulation of anti-tumour functions of NK cells and macrophages after single low-level exposures of mice to X-rays. Int J Low Radiat 3:178–191. doi:10.​1504/​IJLR.​2006.​012017 CrossRef
85.
Zurück zum Zitat Nowosielska EM, Wrembel-Wargocka J, Cheda A, Lisiak E, Janiak MK (2005) Low-level exposures to ionising radiation modulate the anti-tumour activity of murine NK cells. Nukleonika 50(suppl 2):21–24 Nowosielska EM, Wrembel-Wargocka J, Cheda A, Lisiak E, Janiak MK (2005) Low-level exposures to ionising radiation modulate the anti-tumour activity of murine NK cells. Nukleonika 50(suppl 2):21–24
86.
Zurück zum Zitat Nowosielska EM, Wrembel-Wargocka J, Cheda A, Lisiak E, Janiak MK (2006) Enhanced cytotoxic activity of macrophages and suppressed tumour metastases in mice irradiated with low doses of X-rays. J Radiat Res 47:229–236. doi:10.1269/jrr.0572 PubMedCrossRef Nowosielska EM, Wrembel-Wargocka J, Cheda A, Lisiak E, Janiak MK (2006) Enhanced cytotoxic activity of macrophages and suppressed tumour metastases in mice irradiated with low doses of X-rays. J Radiat Res 47:229–236. doi:10.​1269/​jrr.​0572 PubMedCrossRef
87.
Zurück zum Zitat Nowosielska EM, Cheda A, Wrembel-Wargocka J, Janiak MK (2008) Modulation of the growth of pulmonary tumour colonies in mice after single or fractionated low-level irradiations with X-rays. Nukleonika 53(suppl 1):s9–s15 Nowosielska EM, Cheda A, Wrembel-Wargocka J, Janiak MK (2008) Modulation of the growth of pulmonary tumour colonies in mice after single or fractionated low-level irradiations with X-rays. Nukleonika 53(suppl 1):s9–s15
95.
103.
104.
Zurück zum Zitat Cuttler JM, Pollycove M (2003) Can cancer be treated with low doses of radiation? JPandS 8:108–111. Cuttler JM, Pollycove M (2003) Can cancer be treated with low doses of radiation? JPandS 8:108–111.
105.
Zurück zum Zitat Safwat A, Bayoumy Y, El-Sharkawy N, Shaaban K, Mansour O, Kamel A (2003) The potential palliative role and possible immune modulatory effects of low-dose total body irradiation in relapsed or chemo-resistant non-Hodgkin’s lymphoma. Radiother Oncol 69:33–36. doi:10.1016/S0167-8140(03)00247-0 PubMedCrossRef Safwat A, Bayoumy Y, El-Sharkawy N, Shaaban K, Mansour O, Kamel A (2003) The potential palliative role and possible immune modulatory effects of low-dose total body irradiation in relapsed or chemo-resistant non-Hodgkin’s lymphoma. Radiother Oncol 69:33–36. doi:10.​1016/​S0167-8140(03)00247-0 PubMedCrossRef
106.
109.
Zurück zum Zitat Algarra I, García-Lora A, Cabrera T, Ruiz-Cabello F, Garrido F (2004) The selection of tumour variants with altered expression of classical and nonclassical MHC class I molecules: implications for tumour immune escape. Cancer Immunol Immunother 53:904–910. doi:10.1007/s00262-004-0517-9 PubMedCrossRef Algarra I, García-Lora A, Cabrera T, Ruiz-Cabello F, Garrido F (2004) The selection of tumour variants with altered expression of classical and nonclassical MHC class I molecules: implications for tumour immune escape. Cancer Immunol Immunother 53:904–910. doi:10.​1007/​s00262-004-0517-9 PubMedCrossRef
113.
Zurück zum Zitat Seong SY, Matzinger P (2004) Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol 4:469–478. doi:10.1038/nri1372 PubMedCrossRef Seong SY, Matzinger P (2004) Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol 4:469–478. doi:10.​1038/​nri1372 PubMedCrossRef
130.
Zurück zum Zitat Umansky V, Blattner C, Fleming V, Hu X, Gebhardt C, Altevogt P et al (2016) Myeloid-derived suppressor cells and tumor escape from immune surveillance. Semin Immunopathol. doi:10.1007/s00281-016-0597-6 PubMed Umansky V, Blattner C, Fleming V, Hu X, Gebhardt C, Altevogt P et al (2016) Myeloid-derived suppressor cells and tumor escape from immune surveillance. Semin Immunopathol. doi:10.​1007/​s00281-016-0597-6 PubMed
136.
142.
Zurück zum Zitat Zhang Y, Eliav Y, Shin SU, Schreiber TH, Podack ER, Tadmor T, Rosenblatt JD (2013) B lymphocyte inhibition of anti-tumor response depends on expansion of Treg but is independent of B-cell IL-10 secretion. Cancer Immunol Immunother 62:87–99. doi:10.1007/s00262-012-1313-6 PubMedCrossRef Zhang Y, Eliav Y, Shin SU, Schreiber TH, Podack ER, Tadmor T, Rosenblatt JD (2013) B lymphocyte inhibition of anti-tumor response depends on expansion of Treg but is independent of B-cell IL-10 secretion. Cancer Immunol Immunother 62:87–99. doi:10.​1007/​s00262-012-1313-6 PubMedCrossRef
143.
Zurück zum Zitat Zhang Y, Morgan R, Chen C, Cai Y, Clark E, Khan WN et al (2016) Mammary-tumor-educated B cells acquire LAP/TGF-β and PD-L1 expression and suppress anti-tumor immune responses. Int Immunol 28:423–423. doi:10.1093/intimm/dxw007 PubMedCrossRef Zhang Y, Morgan R, Chen C, Cai Y, Clark E, Khan WN et al (2016) Mammary-tumor-educated B cells acquire LAP/TGF-β and PD-L1 expression and suppress anti-tumor immune responses. Int Immunol 28:423–423. doi:10.​1093/​intimm/​dxw007 PubMedCrossRef
149.
Zurück zum Zitat Huang XM, Liu XS, Lin XK, Yu H, Sun JY, Liu XK et al (2014) Role of plasmacytoid dendritic cells and inducible costimulator-positive regulatory T cells in the immunosuppression microenvironment of gastric cancer. Cancer Sci 105:150–158. doi:10.1111/cas.12327 PubMedPubMedCentralCrossRef Huang XM, Liu XS, Lin XK, Yu H, Sun JY, Liu XK et al (2014) Role of plasmacytoid dendritic cells and inducible costimulator-positive regulatory T cells in the immunosuppression microenvironment of gastric cancer. Cancer Sci 105:150–158. doi:10.​1111/​cas.​12327 PubMedPubMedCentralCrossRef
154.
158.
160.
Zurück zum Zitat Pal S, Bhattacharjee A, Ali A, Mandal NC, Mandal SC, Pal M (2014) Chronic inflammation and cancer: potential chemoprevention through nuclear factor kappa B and p53 mutual antagonism. J Inflamm (Lond) 11:23. doi:10.1186/1476-9255-11-23 CrossRef Pal S, Bhattacharjee A, Ali A, Mandal NC, Mandal SC, Pal M (2014) Chronic inflammation and cancer: potential chemoprevention through nuclear factor kappa B and p53 mutual antagonism. J Inflamm (Lond) 11:23. doi:10.​1186/​1476-9255-11-23 CrossRef
161.
Zurück zum Zitat Khan S, Jain M, Mathur V, Feroz SM (2016) Chronic inflammation and cancer: paradigm on tumor progression, metastasis and therapeutic intervention. Gulf J Oncolog 1:86–93PubMed Khan S, Jain M, Mathur V, Feroz SM (2016) Chronic inflammation and cancer: paradigm on tumor progression, metastasis and therapeutic intervention. Gulf J Oncolog 1:86–93PubMed
162.
Zurück zum Zitat Romano M, DE Francesco F, Zarantonello L, Ruffolo C, Ferraro GA, Zanus G et al (2016) From inflammation to cancer in inflammatory bowel disease: molecular perspectives. Anticancer Res 36:1447–1460PubMed Romano M, DE Francesco F, Zarantonello L, Ruffolo C, Ferraro GA, Zanus G et al (2016) From inflammation to cancer in inflammatory bowel disease: molecular perspectives. Anticancer Res 36:1447–1460PubMed
168.
Zurück zum Zitat Mitchel RE, Jackson JS, Carlisle SM (2004) Upper dose threshold for radiation-induced adaptive response against cancer in high-dose-exposed, cancer-prone, radiation-sensitive Trp53 heterozygous mice. Radiat Res 162:20–30. doi:10.1667/RR3190 PubMedCrossRef Mitchel RE, Jackson JS, Carlisle SM (2004) Upper dose threshold for radiation-induced adaptive response against cancer in high-dose-exposed, cancer-prone, radiation-sensitive Trp53 heterozygous mice. Radiat Res 162:20–30. doi:10.​1667/​RR3190 PubMedCrossRef
169.
Zurück zum Zitat Taylor LS (1980) Some nonscientific influences on radiation protection standards and practice. The 1980 Sievert Lecture. Health Phys 39:851–874PubMed Taylor LS (1980) Some nonscientific influences on radiation protection standards and practice. The 1980 Sievert Lecture. Health Phys 39:851–874PubMed
170.
Zurück zum Zitat Zhang C, Jin S, Guo W, Li C, Li X, Rane MJ et al (2011) Attenuation of diabetes-induced cardiac inflammation and pathological remodeling by low-dose radiation. Radiat Res 175:307–321. doi:10.1667/RR1950.1 PubMedCrossRef Zhang C, Jin S, Guo W, Li C, Li X, Rane MJ et al (2011) Attenuation of diabetes-induced cardiac inflammation and pathological remodeling by low-dose radiation. Radiat Res 175:307–321. doi:10.​1667/​RR1950.​1 PubMedCrossRef
172.
Zurück zum Zitat Anderson RE, Tokuda S, Williams WL, Warner NL (1982) Radiation-induced augmentation of the response of A/J mice to SaI tumor cells. Am J Pathol 108:24–37PubMedPubMedCentral Anderson RE, Tokuda S, Williams WL, Warner NL (1982) Radiation-induced augmentation of the response of A/J mice to SaI tumor cells. Am J Pathol 108:24–37PubMedPubMedCentral
173.
Zurück zum Zitat Kharazi AI, James SJ, Taylor JM, Lubinski JM, Nakamura LT, Makinodan T (1994) Combined chronic low dose radiation-caloric restriction: a model for regression of spontaneous mammary tumor. Int J Radiat Oncol Biol Phys 28:641–647. doi:10.1016/0360-3016(94)90189-9 PubMedCrossRef Kharazi AI, James SJ, Taylor JM, Lubinski JM, Nakamura LT, Makinodan T (1994) Combined chronic low dose radiation-caloric restriction: a model for regression of spontaneous mammary tumor. Int J Radiat Oncol Biol Phys 28:641–647. doi:10.​1016/​0360-3016(94)90189-9 PubMedCrossRef
174.
Zurück zum Zitat Lacoste-Collin L, Jozan S, Cances-Lauwers V, Pipy B, Gasset G, Caratero C, Courtade-Saïdi M (2007) Effect of continuous irradiation with a very low dose of gamma rays on life span and the immune system in SJL mice prone to B-cell lymphoma. Radiat Res 168:725–732. doi:10.1667/RR1007.1 PubMedCrossRef Lacoste-Collin L, Jozan S, Cances-Lauwers V, Pipy B, Gasset G, Caratero C, Courtade-Saïdi M (2007) Effect of continuous irradiation with a very low dose of gamma rays on life span and the immune system in SJL mice prone to B-cell lymphoma. Radiat Res 168:725–732. doi:10.​1667/​RR1007.​1 PubMedCrossRef
176.
Zurück zum Zitat Cheda A, Wrembel-Wargocka J, Nowosielska EM, Janiak MK (2005) Stimulatory effects of a single low-level irradiations with X-rays on functions of murine peritoneal macrophages. Nukleonika 50(suppl 2):13–16. Cheda A, Wrembel-Wargocka J, Nowosielska EM, Janiak MK (2005) Stimulatory effects of a single low-level irradiations with X-rays on functions of murine peritoneal macrophages. Nukleonika 50(suppl 2):13–16.
177.
Zurück zum Zitat Cheda A, Nowosielska EM, Wrembel-Wargocka J, Janiak MK (2008) Production of cytokines by peritoneal macrophages and splenocytes after exposures of mice to low doses of X-rays. Radiat Environ Biophys 47:275–283. doi:10.1007/s00411-007-0147-7 PubMedCrossRef Cheda A, Nowosielska EM, Wrembel-Wargocka J, Janiak MK (2008) Production of cytokines by peritoneal macrophages and splenocytes after exposures of mice to low doses of X-rays. Radiat Environ Biophys 47:275–283. doi:10.​1007/​s00411-007-0147-7 PubMedCrossRef
178.
Zurück zum Zitat Cheda A, Nowosielska EM, Wrembel-Wargocka J, Janiak MK (2009) Single or fractionated irradiations of mice with low doses of X-rays stimulate innate immune mechanisms. Int J Low Radiat 6:325–342. doi:10.1504/IJLR.2009.029312 CrossRef Cheda A, Nowosielska EM, Wrembel-Wargocka J, Janiak MK (2009) Single or fractionated irradiations of mice with low doses of X-rays stimulate innate immune mechanisms. Int J Low Radiat 6:325–342. doi:10.​1504/​IJLR.​2009.​029312 CrossRef
179.
Zurück zum Zitat Nowosielska EM, Wrembel-Wargocka J, Cheda A, Janiak MK (2006) A single low-dose irradiation with X-rays stimulates NK cells and macrophages to release factors related to the cytotoxic functions of these cells. Centr Eur J Immunol 31:51–56 Nowosielska EM, Wrembel-Wargocka J, Cheda A, Janiak MK (2006) A single low-dose irradiation with X-rays stimulates NK cells and macrophages to release factors related to the cytotoxic functions of these cells. Centr Eur J Immunol 31:51–56
181.
182.
Zurück zum Zitat Kazem I (1975) Total body irradiation in the management of malignant lymphoma. Radiol Clin 44:457–463. Kazem I (1975) Total body irradiation in the management of malignant lymphoma. Radiol Clin 44:457–463.
183.
Zurück zum Zitat Qasim MM (1975) Total body irradiation in non-Hodgkin lymphoma. Strahlentherapie 149:364–367.PubMed Qasim MM (1975) Total body irradiation in non-Hodgkin lymphoma. Strahlentherapie 149:364–367.PubMed
184.
Zurück zum Zitat Richaud PM, Soubeyran P, Eghbali H, Chacon B, Marit G, Broustet A, Hoerni B (1998) Place of low-dose total body irradiation in the treatment of localized follicular non-Hodgkin’s lymphoma: results of a pilot study. Int J Radiat Oncol Biol Phys 40:387–390. doi:10.1016/S0360-3016(97)00722-0 PubMedCrossRef Richaud PM, Soubeyran P, Eghbali H, Chacon B, Marit G, Broustet A, Hoerni B (1998) Place of low-dose total body irradiation in the treatment of localized follicular non-Hodgkin’s lymphoma: results of a pilot study. Int J Radiat Oncol Biol Phys 40:387–390. doi:10.​1016/​S0360-3016(97)00722-0 PubMedCrossRef
185.
193.
Zurück zum Zitat Kesavan PC (2014) Linear, no threshold response at low doses of ionizing radiation: ideology, prejudice and science. Curr Sci India 107:46–53. Kesavan PC (2014) Linear, no threshold response at low doses of ionizing radiation: ideology, prejudice and science. Curr Sci India 107:46–53.
197.
199.
203.
Zurück zum Zitat Chen Y, Chen X, Li Y, Zhang H, Xie Y, Zhang X et al (2011) Early effects of low dose C ion or X-ray irradiation on peripheral blood lymphocytes of patients with alimentary tract cancer. Dose Response 9:356–368. doi:10.2203/dose-response.10-015.Chen PubMed Chen Y, Chen X, Li Y, Zhang H, Xie Y, Zhang X et al (2011) Early effects of low dose C ion or X-ray irradiation on peripheral blood lymphocytes of patients with alimentary tract cancer. Dose Response 9:356–368. doi:10.​2203/​dose-response.​10-015.​Chen PubMed
204.
Zurück zum Zitat Chen Y, Wang C, He M, Zhang H, Chen X (2014) Effect of low dose heavy ion irradiation on subset percentage and cytokines expression of peripheral blood lymphocytes in patients with pancreatic cancer. Zhonghua Zhong Liu Za Zhi 36:435–439PubMed Chen Y, Wang C, He M, Zhang H, Chen X (2014) Effect of low dose heavy ion irradiation on subset percentage and cytokines expression of peripheral blood lymphocytes in patients with pancreatic cancer. Zhonghua Zhong Liu Za Zhi 36:435–439PubMed
209.
Zurück zum Zitat Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N et al (2013) Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24:589–602. doi:10.1016/j.ccr.2013.09.014 PubMedCrossRef Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N et al (2013) Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24:589–602. doi:10.​1016/​j.​ccr.​2013.​09.​014 PubMedCrossRef
215.
Zurück zum Zitat Shan YX, Jin SZ, Liu XD, Liu Y, Liu SZ (2007) Ionizing radiation stimulates secretion of pro-inflammatory cytokines: dose-response relationship, mechanisms and implications. Radiat Environ Biophys 46:21–29. doi:10.1007/s00411-006-0076-x PubMedCrossRef Shan YX, Jin SZ, Liu XD, Liu Y, Liu SZ (2007) Ionizing radiation stimulates secretion of pro-inflammatory cytokines: dose-response relationship, mechanisms and implications. Radiat Environ Biophys 46:21–29. doi:10.​1007/​s00411-006-0076-x PubMedCrossRef
218.
Zurück zum Zitat Zhang C, Tan Y, Guo W, Li C, Ji S, Li X, Cai L (2009) Attenuation of diabetes-induced renal dysfunction by multiple exposures to low-dose radiation is associated with the suppression of systemic and renal inflammation. Am J Physiol Endocrinol Metab 297:E1366–E1377. doi:10.1152/ajpendo.00478 PubMedCrossRef Zhang C, Tan Y, Guo W, Li C, Ji S, Li X, Cai L (2009) Attenuation of diabetes-induced renal dysfunction by multiple exposures to low-dose radiation is associated with the suppression of systemic and renal inflammation. Am J Physiol Endocrinol Metab 297:E1366–E1377. doi:10.​1152/​ajpendo.​00478 PubMedCrossRef
219.
220.
Metadaten
Titel
Cancer immunotherapy: how low-level ionizing radiation can play a key role
verfasst von
Marek K. Janiak
Marta Wincenciak
Aneta Cheda
Ewa M. Nowosielska
Edward J. Calabrese
Publikationsdatum
30.03.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 7/2017
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-017-1993-z

Weitere Artikel der Ausgabe 7/2017

Cancer Immunology, Immunotherapy 7/2017 Zur Ausgabe

Update Onkologie

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