Next Article in Journal
Carbamazepine Restores Neuronal Signaling, Protein Synthesis, and Cognitive Function in a Mouse Model of Fragile X Syndrome
Next Article in Special Issue
The Abscopal Effect in the Era of Checkpoint Inhibitors
Previous Article in Journal
Acceleration of Carbon Fixation in Chilling-Sensitive Banana under Mild and Moderate Chilling Stresses
Previous Article in Special Issue
Overcoming Immune Evasion in Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review

by
Junko Takahashi
1,* and
Shinsuke Nagasawa
2
1
Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
2
Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(23), 9324; https://doi.org/10.3390/ijms21239324
Submission received: 30 September 2020 / Revised: 4 December 2020 / Accepted: 5 December 2020 / Published: 7 December 2020
(This article belongs to the Special Issue Immunotherapy of Melanoma: Challenges and Solutions)

Abstract

:
Recently, modern therapies involving immune checkpoint inhibitors, cytokines, and oncolytic virus have been developed. Because of the limited treatment effect of modern therapy alone, the immunostimulatory effect of radiotherapy attracted increasing attention. The combined use of radiotherapy and modern therapy has been examined clinically and non-clinically, and its effectiveness has been confirmed recently. Because melanomas have high immunogenicity, better therapeutic outcomes are desired when using immunotherapy. However, sufficient therapeutic effects have not yet been achieved. Thus far, radiotherapy has been used only for local control of tumors. Although extremely rare, radiotherapy has also been reported for systemic control, i.e., abscopal effect. This is thought to be due to an antitumor immune response. Therefore, we herein summarize past information on not only the mechanism of immune effects on radiotherapy but also biomarkers reported in case reports on abscopal effects. We also reviewed the animal model suitable for evaluating abscopal effects. These results pave the way for further basic research or clinical studies on new treatment methods for melanoma. Currently, palliative radiation is administered to patients with metastatic melanoma for local control. If it is feasible to provide both systemic and local control, the treatment benefit for the patients is very large.

Graphical Abstract

1. Introduction

According to information from the Sydney Melanoma Unit database, the initial presentation of recurrence in 873 melanoma patients with American Joint Committee on Cancer (AJCC) Stage I and II disease treated during 1960–2002 was as follows: local, 95 patients (10.9%); in-transit, 86 patients (9.9%); regional lymph nodes (LNs), 300 patients (34.4%); and distant, 392 patients (44.9%) [1]. The median survival time of 1,521 patients with AJCC stage IV melanoma treated during 1971–1993 was 7.5 months, and the estimated five-year survival rate was 6%. Melanoma patients could be divided into three distinct prognostic groups according to the initial site of metastases: cutaneous, nodal, or gastrointestinal metastases (median survival: 12.5 months; estimated five-year survival rate: 14%); pulmonary metastases (8.3 months; 4%); and metastases to the liver, brain, or bone (4.4 months; 3%). No significant difference was observed in the survival rate of patients with AJCC stage IV melanoma during the 22-year review period [2]. Therefore, cutaneous melanoma is reported to be a highly aggressive cancer with a strong propensity for metastases and is associated with a very poor prognosis. Therefore, in addition to topical treatment, treatment for systemic control and metastasis is required.
Modern immunotherapy together with targeted therapy has been effective at treating melanoma metastasis. Currently, immunotherapy together with immune checkpoint inhibitors (ICIs) such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed death-ligand 1 (PD-1), or programmed cell death 1 (PD-L1) inhibitor with specific monoclonal antibodies has been effective at treating advanced melanoma, lung cancer, renal cancer, and other types of cancers [3]. However, this treatment modality has three serious drawbacks: high cost, severe side effects, and effectiveness limited only to approximately 50% of patients [3]. Alternatively, radiotherapy (RT) is a topical approach for treating cancer. However, very rarely, a systemic therapeutic effect called “the abscopal effect” is observed. The abscopal effect includes tumor regression outside of the irradiated field and has been attributed to immunostimulatory effects [4]. Therefore, modern immunotherapy together with RT is considered effective at improving immunotherapy.
Many preclinical and clinical studies have investigated the combination of RT and ICIs (reviewed in [4,5,6,7,8,9,10]). Some studies have reported that the abscopal effect is induced by this combination (RT and ICIs), although there are still individual differences in the effect of the two therapies. Moreover, it seems unclear whether a sufficient outcome can be obtained with this combination. Chicas-Sett et al. listed four items as key aspects for combined RT and immunotherapy: (1) methods and condition of the RT technique for more immunogenicity, (2) ideal treatment sequence (concurrent RT with anti-PD-1/L1, or sequential RT after anti-CTLA-4), (3) whether multisite irradiation would be adequate rather than single-site irradiation, and (4) biomarkers that can guide patient selection [5]. The answer to these questions is yet to be obtained owing to a lack of understanding of the ICIs and the mechanism underlying the abscopal effects of RT. Therefore, herein, we present a review of the use of RT for the systemic and local control of melanoma. We also investigated the mechanism underlying the abscopal effect, case studies on the abscopal effect, and the candidate biomarkers used in those published case studies. Additionally, we reviewed suitable animal model wherein the abscopal effect was evaluated. This review provides information for planning basic research for developing new treatment methods or clinical studies to optimize systemic treatment for melanoma.

2. Review

2.1. Role of RT

RT is one of the three major treatment modalities used in the management of cancer patients. RT, when used either alone or in combination with surgery or chemotherapy, displays a wide range of antitumor effects [5]. An understanding of the basic principles of RT is essential in comprehending its role in the immune effects.

2.1.1. Mechanism of RT

RT aims toward topical control of the tumor. The biological mechanisms underlying the topical antitumor effect of RT have been well established for decades [11]. Briefly, RT induces DNA damage, interrupts cell cycle, and causes tumor cell death through apoptosis and necrosis [12]. When radiation energy is absorbed by the cells, it results in direct macromolecular damage, to some extent. Alternatively, when radiation energy is absorbed by water, it induces radiolysis of water and leads to the production of reactive oxygen species (ROS) such as hydroxyl (OH) radicals, thereby leading to DNA damage [13]. In addition, the bystander effect, which causes cellular damage, is transmitted to an adjacent cell through the communicating gap junctions, and soluble factors such as lipid peroxide products, inosine nucleotides, and cytokines are released from the irradiated cells [14,15,16]. Moreover, radiation-induced vascular fibrosis and occlusion cause nutrient depletion in the tumor [17,18]. Such topical effects are considered to be the main mechanism of RT with conventionally fractionated radiation. Technical advances in delivering RT, such as intensity-modulated, radiosurgery, proton therapy, and electron brachytherapy, enable the routine delivery of a higher radiation dose per fraction [19,20,21]. High doses of radiation, such as in radiosurgery, are thought to induce endothelial cell death, resulting in vascular damage and increased T cell priming in draining lymphoid tissues [13,22,23].
Traditionally, malignant melanoma is thought to be a radio-resistant tumor; however, substantial radiobiological and clinical evidence of cutaneous malignant melanoma is available to refute this notion. Local control has been improved with the use of adjuvant RT, wherein the primary site or regional lymphatics is irradiated in patients with high-risk clinical or pathological features [24]. RT plays an important role in the palliation of metastatic disease and as a treatment for malignant melanoma [25]. Mucosal melanoma is associated with a worse prognosis when compared to the cutaneous form, and the benefit of adjuvant RT has been controversial [6]. Palliative RT offers effective symptom control for focal disease due to cancer. During palliative RT in patients with advanced metastases, shrinkage of tumors outside the area of irradiation is rarely observed. This phenomenon was originally described as “abscopal effect” by Mole in 1953 [26]. The presumed mechanism of the abscopal effect has long been thought to be immune response, and it is difficult to prove the mechanism because the abscopal effect has been observed very infrequently.

2.1.2. Mechanism of Immune Effects on RT

Originally, RT has been considered to have rather an immunosuppressive effect. However, the abscopal effect is considered to be an immunostimulatory effect. The mechanism underlying the immunosuppressive or stimulating effect of radiation is very complex, although much remains unknown (reviewed in [4,5,10,11,13,27]).
The process of T cell priming is thought to be the main part of the immunostimulatory effect of RT. Radiation-induced T cell priming was caused by damage-associated molecular patterns (DAMPs), which are molecules that are secreted, released, or surface-exposed due to death, stress, or injury in cells. DAMPs such as surface-exposed calreticulin, which secrete ATP and passively release high-mobility group protein B1 (HMGB1), are vital for the immunogenic cell death of cancer cells [28]. Calreticulin triggers the phagocytosis of the irradiated tumor cells by dendritic cells and increases the lysis of the irradiated tumor cells by cytotoxic lymphocytes [29,30,31,32]. ATP is released from the irradiated tumor cells, and this release is dependent on the expression of the autophagy factor ATG5 [29,33]. HMGB1 triggers antigen presentation by dendritic cells and priming of antigen-specific T cells after RT in a TLR4-dependent manner [29,30,34]. Other signals involved in radiation-induced T cell priming are interferon (IFN) α/β and complement. IFNs, which are induced by RT, can directly activate lymphocytes including T cells [35,36,37]. The activated lymphocytes express a stimulator molecule of IFN genes (STING) and therefore result in the release of IFN-γ [38].
The effects of such signals on tumor cell in terms of T cell- or natural killer cell-mediated lysis by RT involve major histocompatibility complex class I (MHC-I) [39,40], natural killer cell receptor NKG2D ligands, tumor necrosis factor receptor superfamily (TNFRSF) members, immune checkpoint molecules, and others. NKG2D is bound to MHC class I-related chain A/B (MICB or MICA) on the tumor cell surface, which is upregulated in stem-like cancer cells preferentially [41,42,43,44]. Fas induction of tumor cells by RT increases susceptibility to T cell-mediated lysis and is associated with increased NK cell-mediated lysis [40,45,46]. Upregulation of PL-D1 is induced by RT alone or by chemoradiotherapy in various cancer cells such as B16-F10, GL261, A549, U2OS, H1299, DU145 [47,48,49]. DNA double-strand break-dependent PD-L1 upregulation is caused by ATM/ATR–Chk1/2 activation, followed by STAT-IRF1 activation [49,50]. Upregulation of PD-L1 expression in tumor cells is triggered by activation of the cGAS/STING pathway, followed by IFNα/β, or IFN-γ by CD8+ T cells, or the IL-6-mediated STAT-IRF1 pathway [50,51,52,53].
Cytokine, macrophage, and immunosuppressive leukocyte cause radiation-induced changes in the tumor microenvironment. IFN-α/β and IFN-γ favor tumor control [35,36,37,38], whereas TGF-β, IL-6, and CSF-1 favor tumor growth [54,55,56,57].
Adhesion molecules and chemokines are involved in radiation-induced leukocyte filtration. Vascular cell adhesion molecule 1 (VCAM-1) is mediated by Inos-positive macrophages and IFN-γ produced by hematopoietic cells [36,58]. STING-dependent induction of type I IFNs mediates the upregulation of CXCL 10 expression and subsequent infiltration of T-cell, macrophage, and dendritic cells (DC). RT-induced secretion of CXCL9, CXCL10, and CXCL16 attracts primed effector T cells to the tumor microenvironment [35,37,59,60]. CXCL12 induced by irradiation recruits CD11b+ myeloid cells, which mediate vasculogenesis, and recruits suppressive myeloid cells to the tumor microenvironment [61,62].

2.2. Case Reports of Abscopal Effects of RT

To understand the mechanism of the abscopal effect, it is necessary to understand the phenomena occurring in the clinical setting. Abscopal effects are very rare events that are observed when palliative RT is performed. Therefore, we performed a literature search on PubMed data published from 1989 to August 2020 using the following search terms: “abscopal” and “palliative” in Title/Abstract. Full articles were retrieved when the abstract was considered relevant and only papers published in English were considered. The bibliographies of retrieved papers and reviews were also sought to identify other relevant articles for inclusion. Case reports wherein the abscopal effect was observed after RT were considered eligible. The results are shown in Table 1. There were descriptions about biomarkers in white blood cells, serum, and tumors.

2.2.1. Biomarkers in Leukocytes

The abscopal effects in patients with malignant lymphomas were reported by Antoniades et al. That report presents the cases of two patients with clinical stage III non-Hodgkin’s lymphoma who exhibited marked reduction in the size of the abdominal lymph nodes following irradiation to the mantle [64]. Both of them showed a decrease in total leucocyte count after irradiation, with an increased ratio of segmented neutrophils and a decreased ratio of lymphocyte [64]. It is very important to take a closer look at the white blood cell status before and after the occurrence of the abscopal effect.
Golden et al. reported the first abscopal response to one of the hepatic metastases and ipilimumab in a treatment-refractory lung cancer patient treated with RT [76]. They observed that the absolute lymphocyte count (ALC) increased after RT and ipilimumab treatment. The absolute eosinophil count (AEC) also increased between the first two infusions of ipilimumab [76]. ALCs and AECs are two biomarkers associated with improved survival rates in ipilimumab-treated melanoma patients [99,100,101]. Additionally, post-treatment carcinoembryonic antigen (CEA) levels, a non-specific tumor marker, demonstrated a dramatic drop to normal levels after a peak of 119.6 ng/mL. The pathologic evaluation of a persistent supraclavicular LN showed increased CD8+ cell count and FoxP3+ cell count and a high ratio of CD8+/FoxP3+ cells.

2.2.2. Biomarkers in the Serum

Ohba et al. reported the case of a 76-year-old Japanese man with hepatocellular carcinoma that regressed after RT for thoracic vertebral bone metastasis. They performed serial measurements of serum concentrations of IL-1β, IL-2, IL-4, IL-6, HGF, and tumor necrosis factor-α (TNF-α) before and after RT using sera stored at −80 °C. Serum levels of TNF-α increased and reached 102 pg/mL after RT. They inferred the following: the findings suggest that such abscopal effect-related regression may be associated with host immune response, involving cytokines such as TNF-α [66].
Takaya et al. reported the case of a 69-year-old woman with advanced uterine cervical carcinoma with toruliform para-aortic LN metastases that showed an abscopal effect due to RT (effect outside of the irradiated field). The patient received RT without chemotherapy only for the primary pelvic lesions. After treatment, not only did the cervical tumor in the irradiated field disappear, but the toruliform para-aortic LN swelling outside the irradiated field also spontaneously disappeared. The patient’s laboratory data before irradiation showed normal values except for elevated levels of serum squamous cell carcinoma (SCC) antigen, which increased to 73.5 ng/mL (normal range: 0–1.5 ng/mL) The serum level of the SCC antigen after irradiation had also decreased to the normal range (0.6 ng/mL) [68]. Whether or not tumor antigen levels correlate with systemic response will not be known without accumulating more data.
Postow et al. reported a case of abscopal effect in a patient with NY-ESO-1-positive melanoma treated with ipilimumab and RT [74]. NY-ESO-1 is a molecule belonging to the CTAg (Cancer/testis antigens) family. NY-ESO-1 is a cancer antigen expressed in 30–40% of patients with advanced melanoma, but it is not present in normal adult tissues except testicular germ cells and placenta [102]. NY-ESO-1 expression in a pulmonary nodule removed before ipilimumab treatment was confirmed by immunohistochemical analysis. In serum samples collected before the first ipilimumab treatment and before and after RT, titers of antibody against the NY-ESO-1 protein increased with disease progression, and when ipilimumab therapy was administered, the titers diminished with disease response after RT. This behavior of NY-ESO-1 may reflect the activation of immunity by RT. The authors also monitored the levels of CD4+ Inducible co-stimulator (ICOS)-high in peripheral blood mononuclear cells. ICOS is a marker of activated T cells. An increase in CD4+ ICOS-high cells is associated with clinical benefit from ipilimumab [103]. The number of CD4+ ICOS-high cells increased during ipilimumab induction but decreased before RT. After RT, there was a second increase in the levels.
Stamell et al. reported a case of primary melanoma lesion on the skin of the scalp. With this treatment, serum analysis revealed an increase in the level of autoantibodies against melanoma antigen A3 (MAGEA3) and response to cancer antigen PAS domain-containing 1 (PASD1), indicating a systemic antitumor immune response. Anti-MAGEA3 antibodies were found upon serological testing, and there was an association between the abscopal effect and a systemic antitumor immune response [77].

2.2.3. Biomarkers in Tumors

Joe et al. reported the case of the abscopal effect in squamous carcinoma of the anal canal, with metastases to the pelvic LN, liver, and bone [79]. After palliative RT to the pelvis with sensitizing chemotherapy but without immunotherapy, complete response was observed not only in the primary tumor but also in the bone and multiple liver metastases at 4 months after treatment. SCC antigen levels were elevated at 4.7 µg/L (normal <1.5 µg/L) before RT and decreased at 1.5 µg/L at 5 weeks after RT. The patient received chemotherapy but not immunotherapy. The patient received chemotherapy but not immunotherapy. Immunohistochemical staining of the tumor using immune markers such as PD1, PDL1, CD163, CD3, and CD8 showed heterogeneity depending on the regions within the tumor. Lymphocytes, including CD8+ and CD4+ T cells, were thickly infiltrated in some regions, which suggests an abundance of the immune response.
Most of the intra-tumor-infiltrating lymphocytes (TILs) co-expressed PD1, but lymphocytes present at the boundary between the stroma and the epithelium did not co-express PD1. PDL1 expression was also observed in the tumor and stromal macrophages [79].
Brenneman reported the case of a 67-year-old female with inoperable metastatic unclassified round cell retroperitoneal sarcomas (RPS) treated with palliative proton RT only to the primary tumor. After completion of RT, the patient demonstrated complete regression of all un-irradiated metastases, and near-complete response of the primary lesion without additional therapy. They evaluated CD4 and CD8 of TILs before and after irradiation. The study on TILs revealed that the patient’s pretreatment of primary tumor showed CD4/CD8 infiltration, which suggests immunogenicity. The non-treated metastatic lesion after RT also showed CD4/CD8 infiltration and the CD4/CD8 TIL ratio was similar to that in the pretreated primary tumor [87].
Yaguchi et al. reported the case of a patient with rapidly progressive systemic metastasis and the recurrence of pulmonary pleomorphic carcinoma early after surgery. Bone metastasis was treated with palliative RT, followed by the administration of an ICI, nivolumab, and a marked effect was noted after only three cycles, achieving a near-complete response. IHC analysis before irradiation showed that the tumor cells strongly expressed PD-L1 in the resected lung [92].
D’Andrea and Reddy reported a case of abscopal response in a 42-year-old female patient with brain metastatic melanoma [93]. BRAF mutation and expression of excision repair cross-complementation group 1 (ERCC1), O6-methylguanine-DNA-methyltransferase (MGMT), and class III β-tubulin (TUBB3) were observed on the initial biopsy of the chest lesions before RT [93].
Another study reported the case of a 40-year-old woman who was diagnosed with renal cell carcinoma (RCC) at 3 months after RT, and the levels of C-reactive protein (CRP), hemoglobin (Hb), and platelet (Plt values) were found to be normalized [98]. Several prognostic markers have been reported in RCCs. High CRP, low Hb, and thrombocythemia were also associated with poorer prognosis in RCCs. Histological analysis of the primary RCC lesion was performed by staining with hematoxylin and eosin, anti-CD8 antibody, anti-HLA class 1 antibody, and anti-PD-L1 antibody. Histological re-examination showed the heterogeneity of the primary RCC lesions in this case [98].
Before the development of immunotherapy, the abscopal effects varied considerably from one individual to another and was very infrequent. For this reason, in literature, there are few descriptions of biomarkers for understanding the mechanism, biomarkers for predicting good results of RT, and biomarkers for evaluating therapeutic effects. However, in the future, it will be possible to store samples and evaluate them retrospectively for serum components and tumors. In addition, systematic measurement in future clinical trials will advance the understanding of the immunostimulatory mechanism of RT.

2.3. Animal Model for Evaluating the Abscopal Effect

We performed a literature search on PubMed data published from 1989 to August 2020 using the following search terms when found in the title/abstract: “abscopal” and “model” and “mouse.” Full articles were retrieved when the abstract was considered relevant. The bibliographies of retrieved papers and reviews were also sought to identify other relevant articles to be included. Papers were considered eligible when the experimental protocol for evaluating the abscopal effect of RT was well defined. The results are shown in Table 2. Here, we have confirmed the usefulness of these models, not those target to examined using these animal models.
Usually, to evaluate the occurrence of distant metastasis through the abscopal effect, tumor cells were inoculated subcutaneously on the left and right sides, or up and down. Irradiation at any one of the tumors with X-rays showed the local control of the tumor, and the non-irradiated tumor showed the abscopal effect [55,104,106,107,109,112,114,117,118].
As their purpose was to investigate distant metastases of the cancer, the same cell line was inoculated in the left and right sides. However, Dewan et al. used two cell lines [107]. BALB/c and C57BL/6 mice were injected s.c. with 1 × 105 TSA (mouse mammary carcinoma cell line) and 5 × 105 MCA38 (mouse colon carcinoma) cells, respectively, on the right flank on day 0 (primary tumor) and in the left flank on day 2 (secondary tumor). The mice received a single radiation dose of 20 Gy, in three fractions of 8 Gy, or in five fractions of 6 Gy in the right flank continuously for the first time from the 12th day. CTLA-4–blocking mAb 9H10 or vehicle (PBS) was administered i.p. at a dose of 200 μg/mouse (10 mg/kg) on days 14, 17, and 20. The frequency of CD8+ T cells showing tumor-specific IFN-gamma production was proportional to the inhibition of the secondary tumor [107].
C57BL/6 mice were injected s.c. with 2 × 105 B16-F10 cells (mouse melanoma cell line) in the right flank (primary tumor) and 1 × 104 B16-F10 cells in the left flank. The mice received a single radiation dose of 5, 10, or 20 Gy on the ninth day after inoculation. The tumors were then treated with five intratumoral administration of Newcastle disease virus at 106 PFU/dose every 2 days and three i.p. injections of anti-PD1 (200 μg) every 4 days. Tumor volumes were measured until the humane endpoint was 1,000 mm3. The result suggests that the abscopal effect is driven primarily by the virus, and radiation adds superior local control while not hampering the development of systemic immunity [115].
Tumors to observe the abscopal effect usually inoculate fewer cells than tumor for irradiation, or delay inoculation a few days. This is because if the tumor grows too large, even if the abscopal effect occurs, the effect is small and difficult to observe. Six-week-old mice have been used in many experiments. However, Markovsky et al. used 12-week-old mice in order to allow to mature the immune response [112].

2.3.1. Model of Metastasis

Pfannenstiel et al. [113] used a brain metastasis mice model for irradiating brain tumors with X-rays and observing body tumors. C57BL/6 mice were injected with 1 × 104 D4M cells (mouse melanoma cell line) on the right frontal lobes and s.c. with 1.5 × 105 D4M cells on the right flank (primary tumor). The mice received a fractionated irradiation (2 Gy × 4) on the 15th day after inoculation. For antibody treatment, anti-PD-1 antibody or an IgG control was injected at 150 μg/dose i.p. starting five days before irradiation and continuing every fifth day for the entire duration of the experiment. Combination treatment produced a stronger systemic antitumor immune response than either treatment alone [113].
Xia et al. used the mouse model of metastatic osteosarcoma to the brain. They tried to explore the ability of local radiation and anti-PD-1 blockade to induce beneficial antitumor immune responses against distant, un-irradiated brain metastatic tumors by irradiation to flank tumors [111].
For ethical reasons, it is not possible to resect LNs from patients during the course of RT for metastatic LNs (MLNs) or carry out histopathological examination over time. Thus, the effect of radiation on the histopathology of MLNs has remained unclear. Kikuchi et al. succeeded in irradiating individual MLNs through a hole in a lead shield, using mice with swollen LNs. They used MXH10/Mo/lpr mice whose immune system is functional except for the signaling pathway related to Fas. A 60-μL aliquot of tumor cells (3.3 × 105 cells/mL) was manually injected into the subiliac LNs (SiLN) to produce metastasis in the ipsilateral proper axillary LN (PALN). The PALN has LNs of comparable size to those found in humans, approximately 10 mm in size, which permits shielding of sites other than the target LN with a lead plate [116].
Yasuda et al. used a BALB/c mouse model of simultaneous subcutaneous tumor and liver metastasis of Colon26. Colon26 cells were implanted subcutaneously (s.c) in the left flanks of BALB/c mice. The liver metastasis was produced by intra-splenic injection of colon 26 cells at 3 weeks after s.c. inoculation. They showed that topical administration of IL-2 not only enhances shrinkage of the irradiated tumor itself but can also suppress the development of distant metastasis of tumors located outside the RT field, possibly through the induction of a systemic T cell response [108].

2.3.2. Verification of the Abscopal Effect Using Immunodeficient Mice

Based on classical radiobiology, the tumor response of RT is caused by radiation-induced DNA damage produced within the primary radiation field. However, RT also affects the tumor microenvironment and alters the balance of inflammatory signals in the tumor [112].
Markovsky et al. investigated 67NR murine orthotopic breast tumors in both immunocompetent and nude mice. They examined the effect of hemi vs. full-tumor irradiation. The expectation is that a hemi-irradiated tumor would undergo no more than 50% cell killing and this was confirmed in nude mice. However, hemi-irradiation resulted in several (five of 15) tumor cures in immunocompetent BALB/c mice. These results showed that the tumor response of RT is due to radiation-induced DNA damage produced within the primary radiation field. They experimentally verified that ICAM, FTZ720 (a compound that inhibits T-cell egress from LNs), CD8, etc. are immune mediators [112].
Blanquicett et al. examined the effect of X-ray irradiation alone and in combination with capecitabine and/or celecoxib using nude mice bearing BxPC-3 pancreatic in both irradiated and lead-shielded contralateral BxPC-3 human pancreatic xenografts. As a result, they showed that treatment with X-ray irradiation in combination with capecitabine and/or celecoxib suppressed not only the tumors present in the irradiation field but also the tumors outside the field of irradiation [106].
Strigari et al. investigated whether the abscopal effect induced by RT is able to sterilize non-irradiated tumor cells. Athymic female nude mice bearing wild-type (wt)-p53 or p53-null HCT116 human colon cancer xenografts were irradiated at a dose of 10 or 20 Gy (IR groups), delivered using a 10-MeV electron beam. All directly irradiated tumors, showed a dose-dependent delayed and reduced regrowth, independent of the p53 status. Importantly, a significant effect on tumor growth inhibition was also demonstrated in a non-irradiated tumor of wt-p53 tumors in the 20 Gy-irradiation group, but no significant difference was observed in the NIR p53-null tumors, independent of the dose delivered. These results suggest that the interplay between the delivered dose and p53 status might help sterilize out-of-field tumor cells [109].
The results of Blanquicett et al. and Strigari et al. indicate that not only the immune system by T cells but also another mechanism occurs in the abscopal effect.

2.3.3. Other Models

Experiments have also been conducted to observe the regression of cancer by irradiating normal tissue with X-rays instead of cancer. The Lewis lung carcinoma (LLC) cell line was implanted in the midline dorsum of C57BL/6 mice. At day 10 in post-implantation animals with tumors, irradiation was initiated on day 10 and 10 Gy was delivered for 5 consecutive days for the leg. The tumors in the mice that received radiation treatment to the leg grew at a significantly slower rate than those in the mice of the non-irradiated group [105].
Aravindan et al. investigated the effects of non-targeted distant organs by irradiation on normal tissues [110]. They examined the orchestration of NF-κB signaling after ischemia-reperfusion (IR) in the heart, a non-targeted distant organ, tissues of C57/BL6 mice exposed, limiting to lower abdomen 1-cm diameter, to single-dose IR (2 or 10 Gy) or fractionated IR (2 Gy per day for 5 days). As result, some genes showed dose- and fractionation-independent upregulation. Immunohistochemistry revealed a robust increase in p65 and cMyc expression in the distant heart after single-dose and fractionated irradiation [110].
Rationally designing the treatment for understanding the mechanism of the abscopal effect may have a great impact on the treatment of metastatic disease.

3. Discussion

3.1. Immunogenicity of Melanoma Cells Is Immunostimulated Not Only by Immunotherapy But Also by RT

Melanoma has been regarded as a malignant tumor with high immunogenicity. The presence of TILs, defined as a polymorphic group composed mainly of effector T lymphocytes, Tregs, NK cells, dendritic cells, and macrophages, is a well-described feature as a favorable prognostic factor in melanoma [119,120]. The immunogenic property of melanoma caused improvement of the median overall survival and provided hope to many melanoma patients in terms of using inhibition of immune checkpoints [121]. However, because of primary (intrinsic) and secondary (acquired) resistance to ICIs, not all patients derive benefit from ICI treatment. Patients experience immune-related adverse events such as colitis, hypothyroidism, hepatitis, hypophysitis, hyperthyroidism, and pneumonitis, which are significantly escalated when combined with anti-CTLA-4 and anti-PD-1 therapy [122].
Historically, the immune effect of RT was considered to be suppressive. However, in the light of recent research, it has been shown that its interaction with the immune system is much more complex [123]. With the progress of immunotherapy, the immunostimulatory mechanism of RT is drawing attention. The largest clinical case showing the immunostimulatory effect of radiation is the abscopal effect, which, although very infrequently, has manifested as an actual therapeutic effect, and cases have been published with radiation alone. Similar to immune reactions, the abscopal effect requires priming of immune cells against tumor antigens [124]. The abscopal effects of RT is enhanced when combined with ICI drugs such as ipilimumab, pembrolizumab, etc., which induces the systemic antitumor immune response [125]. Furthermore, taking advantage of the immunogenicity of RT, its combined use with treatment methods other than the inhibition of immune checkpoints, such as the combined use with cytokine treatment, such as IL-2, IL-15, GM-CSF, IFN-α, TNF-α, and IL-12, has been studied, and clinical effects have been obtained by the combination of some of them with RT [27].

3.2. Enhanced RT Induced Immunity with Other Modern Therapies

To improve the therapeutic effect of RT using the immunogenicity of radiation, the combined treatment of RT and oncolytic viruses, hyperthermia, photodynamic therapy, etc. is being studied. Oncolytic viruses preferentially replicate in tumors, compared to that in normal tissue and promote ICD and induction of host systemic antitumor immunity. Research indicated that the combination of viral therapy and RT has synergistic antitumor effects both in vitro and in vivo [126]. In addition, oncolytic viruses are studied as a combination treatment with ICI. Anti-murine PD1 antibody showed enhanced antitumor effects with the combination with Herpes simplex virus type 1 (HSV-1). HSV-1 is the only oncolytic immunotherapy treatment that has received approval from the Food and Drug Approval, and have suggested to lead immunological memory [127]. Hyperthermia application in combination with RT and/or chemotherapy may not only improve local tumor control but also lead to systemic and immune mediated antitumor responses according to recent research [128]. 5-aminolevulinic acid (5-ALA) mediated photodynamic therapy, an established approach for topical cancers, can induce an effective antitumor immune response [129]. Zhang et al. demonstrated that dendritic cell stimulated by 5-ALA mediated photodynamic therapy can induce immune responses against cancers. Similar to RT, photodynamic treatment, which is thought to be a topical treatment approach, also affects the systemic immune system [129].
Immune activations of carbon ion or proton beams, which are the modern RT technologies, have also been investigated. Response to proton irradiation, a considerable immune response was showed by gene expression profile analysis of breast cancer xenograft model [130]. Using the cell surface translocation of calreticulin (ecto-CRT) as an “eat me” signal for phagocytosis of dying cells in vitro, proton and photon increased ecto-CRT exposure with dose escalation up to 10 Gy, while carbon-ion increased most ecto-CRT exposure at 4 Gy rather than 10 or 2 Gy [131]. A new RT method, “radiodynamic therapy (RDT),” which produces a larger amount and type of ROS in the tumor by a physicochemical reaction of protoporphyrin IX with radiation [132], also showed higher immunostimulatory effect than RT [133,134]. In this way, research on the immunostimulatory effect of modern RT-induced immunity are being advanced.

3.3. Evaluation of the Abscopal Effect Using Nude Mice

The study that reported abscopal effects in nude mice are to be highlighted [106,109]. Nude mice are partially immune-deficient; hence, if the abscopal effect occurs in producing mature T-cell-dependent manner, then the abscopal effect should not occur. In nude mice, T cell precursors do not have a defect, and some functional mature T cells can be found especially in adult animals. A nude mouse is a strain with a genetic mutation that causes a deteriorated or absent thymus, resulting in an inhibited immune system due to a greatly reduced number of T cells. However, their response to T-independent antigens is normal, having an increased response of macrophage and NK cells [135]. Thus, the abscopal effect observed in nude mice seems to suggest that other immune mechanisms different from T cell priming may play a role. This aspect seems to be very important in considering the effectiveness or ineffectiveness of the combination of immunotherapy and RT and their factors.

3.4. Biomarker of Immune Response

Research on the biomarkers for immune response during treatment is ongoing. Some predictive biomarkers including protein, DNA and mRNA, in melanoma immunotherapy, such as PDL1(protein), TMB (tumor mutational burden, DNA), B2m (β- microglobulin 2 gene, DNA, mRNA), associated with the tumor microenvironment and associated with the whole organism with melanoma, were candidates (reviewed in [3]).
However, there are only a few studies, and there are still many unknown factors. Three categories of biomarkers may be needed to further sophisticate to conventional melanoma treatments, (1) to understand the mechanism, (2) predict the therapeutic effect, and (3) evaluate the therapeutic effectiveness.
Abscopal case reports of RT showed that information could be obtained from the serum, white blood cells, and tumors. However, as can be seen from the case report, there is too little information on clinical biomarkers. When conducting prospective clinical trials, it is desirable to obtain this information as systematically as possible [64,68,74,76,77,79,87,92,93,98]. Because the activation of the abscopal effect requires immune preparation, tumor antigen of serum, cytokines of serum, and monitoring the status of leukocyte in the blood and tumors seem essential.
Animal models are very useful for assessing the systemic effects of RT and for evaluating the effects of the abscopal effect. In addition, to investigate which biomarker should be focused on, research involving preclinical studies using the abscopal animal models is required. For these purposes, the vertically or left and right transplantation model of the body [55,104,106,107,109,112,114,115,117,118], the brain metastasis model [111,113], the lung metastasis model [104], the LN metastasis model [116], the liver metastasis model [108], etc. will be very useful. In addition, the use of nude mice makes it possible to separate T cells effect from other factors.
However, there are only a few studies, and there are still many unknown factors; therefore, further investigations are needed. The vast amount of basic research as well as clinical studies focused on the immune effect mechanisms of RT, immunotherapy, and combined therapy provides hope for melanoma treatment in the future.

4. Conclusions

Palliative RT is provided to patients with metastatic melanoma for local control. However, as shown through the abscopal effect, RT has the potential for not only local control but also systemic control.
In recent years, modern therapies such as ICIs, cytokines, and oncolytic virus treatment have been developed. Furthermore, the immunostimulatory effect of X-rays has attracted increasing attention, and research on the combined use with ICIs and cytokines is progressing clinically and non-clinically, and its effectiveness is being verified. Currently, there is not enough accumulated knowledge for immunoregulation, although research and development of therapeutic methods that output systemic control is desired.

Author Contributions

Conceptualization, J.T.; writing–original draft preparation, J.T.; writing–review and editing, J.T. and S.N. Both authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI, Grant Numbers 18H02705, 19K22609, and 20K08003.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CTLA-4Cytotoxic T-lymphocyte-associated protein 4
ICAM-1Intercellular adhesion molecule 1
ICIImmune checkpoint inhibitor
DAMPsDamage-associated molecular patterns
HMGB1High-mobility group protein B1
IFNInterferon
LNLymph node
MDSCsMyeloid-derived suppressor cells
MICA/BMajor histocompatibility complex class I-related chain A/B
NK cellsNatural killer cells
PD-1Programmed cell death 1
PD-L1Programmed death-ligand 1
ROSReactive oxygen species
RTRadiotherapy
STINGStimulator of IFN genes
TILsTumor-infiltrating lymphocytes
TNFRSFTumor necrosis factor receptor superfamily
TregsRegulatory T lymphocytes
VCAM-1Vascular cell adhesion molecule 1

References

  1. Francken, A.B.; Accortt, N.A.; Shaw, H.M.; Wiener, M.; Soong, S.J.; Hoekstra, H.J.; Thompson, J.F. Prognosis and determinants of outcome following locoregional or distant recurrence in patients with cutaneous melanoma. Ann. Surg. Oncol. 2008, 15, 1476–1484. [Google Scholar] [CrossRef] [PubMed]
  2. Barth, A.; Wanek, L.A.; Morton, D.L. Prognostic factors in 1521 melanoma patients with distant metastases. J. Am. Coll Surg. 1995, 181, 193–201. [Google Scholar] [PubMed]
  3. Olbryt, M.; Rajczykowski, M.; Widłak, W. Biological Factors behind Melanoma Response to Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2020, 21, 4071. [Google Scholar] [CrossRef] [PubMed]
  4. Walle, T.; Martinez Monge, R.; Cerwenka, A.; Ajona, D.; Melero, I.; Lecanda, F. Radiation effects on antitumor immune responses: Current perspectives and challenges. Ther. Adv. Med. Oncol. 2018, 18, 1758834017742575. [Google Scholar] [CrossRef] [PubMed]
  5. Chicas-Sett, R.; Zafra-Martin, J.; Morales-Orue, I.; Castilla-Martinez, J.; Berenguer-Frances, M.A.; Gonzalez-Rodriguez, E.; Rodriguez-Abreu, D.; Couñago, F. Immunoradiotherapy as An Effective Therapeutic Strategy in Lung Cancer: From Palliative Care to Curative Intent. Cancers 2020, 12, 2178. [Google Scholar] [CrossRef]
  6. Rogers, S.J.; Puric, E.; Eberle, B.; Datta, N.R.; Bodis, S.B. Radiotherapy for Melanoma: More than DNA Damage. Dermatol. Res. Pract. 2019, 2019, 9435389. [Google Scholar] [CrossRef] [Green Version]
  7. Walshaw, R.C.; Honeychurch, J.; Illidge, T.M. Stereotactic ablative radiotherapy and immunotherapy combinations: Turning the future into systemic therapy? Br. J. Radiol. 2016, 89, 20160472. [Google Scholar] [CrossRef] [Green Version]
  8. Schoenfeld, J.D.; Mahadevan, A.; Floyd, S.R.; Dyer, M.A.; Catalano, P.J.; Alexander, B.M.; McDermott, D.F.; Kaplan, I.D. Ipilmumab and cranial radiation in metastatic melanoma patients: A case series and review. J. Immunother. Cancer. 2015, 3, 50. [Google Scholar] [CrossRef] [Green Version]
  9. Seyedin, S.N.; Schoenhals, J.E.; Lee, D.A.; Cortez, M.A.; Wang, X.; Niknam, S.; Tang, C.; Hong, D.S.; Naing, A.; Sharma, P.; et al. Strategies for combining immunotherapy with radiation for anticancer therapy. Immunotherapy 2015, 7, 967–980. [Google Scholar] [CrossRef] [Green Version]
  10. Vacchelli, E.; Vitale, I.; Tartour, E.; Eggermont, A.; Sautès-Fridman, C.; Galon, J.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial Watch: Anticancer radioimmunotherapy. Oncoimmunology. 2013, 2, e25595. [Google Scholar] [CrossRef] [Green Version]
  11. Connell, P.P.; Hellman, S. Advances in radiotherapy and implications for the next century: A historical perspective. Cancer Res. 2009, 69, 383–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.-W. Cancer and radiation therapy: Current advances and future directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wani, S.Q.; Dar, I.A.; Khan, T.; Lone, M.M.; Afroz, F. Radiation Therapy and its Effects beyond the Primary Target: An Abscopal Effect. Cureus 2019, 19, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mothersill, C.; Seymour, C. Radiation-induced bystander effects: Past history and future directions. Radiat. Res. 2001, 155, 759–767. [Google Scholar] [CrossRef]
  15. Prise, K.M.; O’Sullivan, J.M. Radiation-induced bystander signalling in cancer therapy. Nat. Rev. Cancer. 2009, 9, 351–360. [Google Scholar] [CrossRef] [PubMed]
  16. Mothersill, C.; Seymour, C. Medium from irradiated human epithelial cells but not human fibroblasts reduces the clonogenic survival of unirradiated cells. Int. J. Radiat. Biol. 1997, 71, 421–427. [Google Scholar]
  17. Finger, P.T. Radiation therapy for choroidal melanoma. Surv. Ophthalmol. 1997, 42, 215–232. [Google Scholar] [CrossRef]
  18. Nag, S.; Quivey, J.M.; Earle, J.D.; Followill, D.; Fontanesi, J.; Finger, P.T. American Brachytherapy Society. The American Brachytherapy Society recommendations for brachytherapy of uveal melanomas. Int. J. Radiat. Oncol. Biol. Phys. 2003, 56, 544–555. [Google Scholar] [CrossRef]
  19. Scaringi, C.; Agolli, L.; Minniti, G. Technical Advances in Radiation Therapy for Brain Tumors. Anticancer. Res. 2018, 38, 6041–6045. [Google Scholar] [CrossRef] [Green Version]
  20. Podder, T.K.; Fredman, E.T.; Ellis, R.J. Advances in Radiotherapy for Prostate Cancer Treatment. Adv. Exp. Med. Biol. 2018, 1096, 31–47. [Google Scholar]
  21. Paunesku, T.; Woloschak, G.E. Future Directions of Intraoperative Radiation Therapy: A Brief Review. Front. Oncol. 2017, 7, 300. [Google Scholar] [CrossRef] [Green Version]
  22. Kolesnick, R.; Fuks, Z. Radiation and ceramide-induced apoptosis. Oncogene 2003, 22, 5897–5906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ahmad, S.S.; Duke, S.; Jena, R.; Williams, M.V.; Burnet, N.G. Advances in radiotherapy. BMJ 2012, 345, e7765. [Google Scholar] [CrossRef]
  24. Shuff, J.H.; Siker, M.L.; Daly, M.D.; Schultz, C.J. Role of radiation therapy in cutaneous melanoma. Clin. Plast. Surg. 2010, 37, 147–160. [Google Scholar] [CrossRef] [PubMed]
  25. Fenig, E.; Eidelevich, E.; Njuguna, E.; Katz, A.; Gutman, H.; Sulkes, A.; Schechter, J. Role of radiation therapy in the management of cutaneous malignant melanoma. Am. J. Clin. Oncol. 1999, 22, 184–186. [Google Scholar] [CrossRef] [PubMed]
  26. Mole, R.H. Whole body irradiation; radiobiology or medicine? Br. J. Radiol. 1953, 26, 234–241. [Google Scholar] [CrossRef] [PubMed]
  27. Palata, O.; Hradilova Podzimkova, N.; Nedvedova, E.; Umprecht, A.; Sadilkova, L.; Palova Jelinkova, L.; Spisek, R.; Adkins, I. Radiotherapy in Combination With Cytokine Treatment. Front. Oncol. 2019, 9, 367. [Google Scholar] [CrossRef]
  28. Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef]
  29. Golden, E.B.; Frances, D.; Pellicciotta, I.; Demaria, S.; Helen Barcellos-Hoff, M.; Formenti, S.C. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014, 3, e28518. [Google Scholar] [CrossRef] [Green Version]
  30. Ko, A.; Kanehisa, A.; Martins, I.; Senovilla, L.; Chargari, C.; Dugue, D.; Mariño, G.; Kepp, O.; Michaud, M.; Perfettini, J.L.; et al. Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ. 2014, 21, 92–99. [Google Scholar] [CrossRef]
  31. Gameiro, S.R.; Jammeh, M.L.; Wattenberg, M.M.; Tsang, K.Y.; Ferrone, S.; Hodge, J.W. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 2014, 5, 403–416. [Google Scholar] [CrossRef] [Green Version]
  32. Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.L.; Castedo, M.; Mignot, G.; Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 2007, 13, 54–61. [Google Scholar] [CrossRef]
  33. Elliott, M.R.; Chekeni, F.B.; Trampont, P.C.; Lazarowski, E.R.; Kadl, A.; Walk, S.F.; Park, D.; Woodson, R.I.; Ostankovich, M.; Sharma, P.; et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 2009, 461, 282–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef] [PubMed]
  35. Burnette, B.C.; Liang, H.; Lee, Y.; Chlewicki, L.; Khodarev, N.N.; Weichselbaum, R.R.; Fu, Y.X.; Auh, S.L. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011, 71, 2488–2496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lugade, A.A.; Sorensen, E.W.; Gerber, S.A.; Moran, J.P.; Frelinger, J.G.; Lord, E.M. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J. Immunol. 2008, 180, 3132–3139. [Google Scholar] [CrossRef]
  37. Lim, J.Y.; Gerber, S.A.; Murphy, S.P.; Lord, E.M. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol. Immunother. 2014, 63, 259–271. [Google Scholar] [CrossRef] [Green Version]
  38. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
  39. Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A.; Chakraborty, M.; Wansley, E.K.; Camphausen, K.; Luiten, R.M.; de Ru, A.H.; Neijssen, J.; et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef]
  40. Garnett, C.T.; Palena, C.; Chakraborty, M.; Tsang, K.Y.; Schlom, J.; Hodge, J.W. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2005, 64, 7985–7994. [Google Scholar] [CrossRef]
  41. Gasser, S.; Orsulic, S.; Brown, E.J.; Raulet, D.H. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005, 436, 1186–1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Xu, X.; Rao, G.S.; Groh, V.; Spies, T.; Gattuso, P.; Kaufman, H.L.; Plate, J.; Prinz, R.A. Major histocompatibility complex class I-related chain A/B (MICA/B) expression in tumor tissue and serum of pancreatic cancer: Role of uric acid accumulation in gemcitabine-induced MICA/B expression. BMC Cancer 2011, 11, 194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bedel, R.; Thiery-Vuillemin, A.; Grandclement, C.; Balland, J.; Remy-Martin, J.P.; Kantelip, B.; Pallandre, J.R.; Pivot, X.; Ferrand, C.; Tiberghien, P.; et al. Novel role for STAT3 in transcriptional regulation of NK immune cell targeting receptor MICA on cancer cells. Cancer Res. 2011, 71, 1615–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Steinle, A.; Li, P.; Morris, D.L.; Groh, V.; Lanier, L.L.; Strong, R.K.; Spies, T. Interactions of human NKG2D with its ligands MICA, MICB, and homologs of the mouse RAE-1 protein family. Immunogenetics 2001, 53, 279–287. [Google Scholar] [CrossRef]
  45. Ames, E.; Canter, R.J.; Grossenbacher, S.K.; Mac, S.; Smith, R.C.; Monjazeb, A.M.; Chen, M.; Murphy, W.J. Enhanced targeting of stem-like solid tumor cells with radiation and natural killer cells. Oncoimmunology 2015, 4, e1036212. [Google Scholar] [CrossRef] [Green Version]
  46. Suzuki, Y.; Mimura, K.; Yoshimoto, Y.; Watanabe, M.; Ohkubo, Y.; Izawa, S.; Murata, K.; Fujii, H.; Nakano, T.; Kono, K. Immunogenic tumor cell death induced by chemoradiotherapy in patients with esophageal squamous cell carcinoma. Cancer Res. 2012, 72, 3967–3976. [Google Scholar] [CrossRef] [Green Version]
  47. Gong, X.; Li, X.; Jiang, T.; Xie, H.; Zhu, Z.; Zhou, F.; Zhou, C. Combined Radiotherapy and Anti-PD-L1 Antibody Synergistically Enhances Antitumor Effect in Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 1085–1097. [Google Scholar] [CrossRef] [Green Version]
  48. Derer, A.; Spiljar, M.; Bäumler, M.; Hecht, M.; Fietkau, R.; Frey, B.; Gaipl, U.S. Chemoradiation Increases PD-L1 Expression in Certain Melanoma and Glioblastoma Cells. Front. Immunol. 2016, 7, 610. [Google Scholar] [CrossRef] [Green Version]
  49. Sato, H.; Niimi, A.; Yasuhara, T.; Permata, T.; Hagiwara, Y.; Isono, M.; Nuryadi, E.; Sekine, R.; Oike, T.; Kakoti, S.; et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 2017, 8, 1751. [Google Scholar] [CrossRef]
  50. Sato, H.; Jeggo, P.A.; Shibata, A. Regulation of programmed death-ligand 1 expression in response to DNA damage in cancer cells: Implications for precision medicine. Cancer Sci. 2019, 110, 3415–3423. [Google Scholar] [CrossRef] [Green Version]
  51. Verbrugge, I.; Hagekyriakou, J.; Sharp, L.L.; Galli, M.; West, A.; McLaughlin, N.M.; Duret, H.; Yagita, H.; Johnstone, R.W.; Smyth, M.J.; et al. Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies. Cancer Res. 2012, 72, 3163–3174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Deng, L.; Liang, H.; Burnette, B.; Beckett, M.; Darga, T.; Weichselbaum, R.R.; Fu, Y.X. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Investig. 2014, 124, 687–695. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, M.F.; Chen, P.T.; Chen, W.C.; Lu, M.S.; Lin, P.Y.; Lee, K.D. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget 2016, 7, 7913–7924. [Google Scholar] [CrossRef]
  54. Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef] [PubMed]
  55. Vanpouille-Box, C.; Diamond, J.M.; Pilones, K.A.; Zavadil, J.; Babb, J.S.; Formenti, S.C.; Barcellos-Hoff, M.H.; Demaria, S. TGFβ Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res. 2015, 75, 2232–2242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Xu, J.; Escamilla, J.; Mok, S.; David, J.; Priceman, S.; West, B.; Bollag, G.; McBride, W.; Wu, L. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013, 73, 2782–2794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Matsuoka, Y.; Nakayama, H.; Yoshida, R.; Hirosue, A.; Nagata, M.; Tanaka, T.; Kawahara, K.; Sakata, J.; Arita, H.; Nakashima, H.; et al. IL-6 controls resistance to radiation by suppressing oxidative stress via the Nrf2-antioxidant pathway in oral squamous cell carcinoma. Br. J. Cancer 2016, 115, 1234–1244. [Google Scholar] [CrossRef] [Green Version]
  58. Klug, F.; Prakash, H.; Huber, P.E.; Seibel, T.; Bender, N.; Halama, N.; Pfirschke, C.; Voss, R.H.; Timke, C.; Umansky, L.; et al. Low-dose irradiation programs macrophage differentiation to an iNOS⁺/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013, 24, 589–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef] [PubMed]
  60. Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef]
  61. Kozin, S.V.; Kamoun, W.S.; Huang, Y.; Dawson, M.R.; Jain, R.K.; Duda, D.G. Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res. 2010, 70, 5679–5685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kioi, M.; Vogel, H.; Schultz, G.; Hoffman, R.M.; Harsh, G.R.; Brown, J.M. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Investig. 2010, 120, 694–705. [Google Scholar] [CrossRef] [PubMed]
  63. Ehlers, G.; Fridman, M. Abscopal effect of radiation in papillary adenocarcinoma. Br. J. Radiol. 1973, 46, 220–222. [Google Scholar] [CrossRef] [PubMed]
  64. Antoniades, J.; Brady, L.W.; Lightfoot, D.A. Lymphangiographic demonstration of the abscopal effect in patients with malignant lymphomas. Int. J. Radiat. Oncol. Biol. Phys. 1977, 2, 141–147. [Google Scholar] [CrossRef]
  65. Rees, G.J. Abscopal regression in lymphoma: A mechanism in common with total body irradiation? Clin. Radiol. 1981, 32, 475–480. [Google Scholar] [CrossRef]
  66. Ohba, K.; Omagari, K.; Nakamura, T.; Ikuno, N.; Saeki, S.; Matsuo, I.; Kinoshita, H.; Masuda, J.; Hazama, H.; Sakamoto, I.; et al. Abscopal regression of hepatocellular carcinoma after radiotherapy for bone metastasis. Gut 1998, 43, 575–577. [Google Scholar] [CrossRef] [PubMed]
  67. Wersäll, P.J.; Blomgren, H.; Pisa, P.; Lax, I.; Kälkner, K.M.; Svedman, C. Regression of non-irradiated metastases after extracranial stereotactic radiotherapy in metastatic renal cell carcinoma. Acta Oncol. 2006, 45, 493–497. [Google Scholar] [CrossRef]
  68. Takaya, M.; Niibe, Y.; Tsunoda, S.; Jobo, T.; Imai, M.; Kotani, S.; Unno, N.; Hayakawa, K. Abscopal effect of radiation on toruliform para-aortic lymph node metastases of advanced uterine cervical carcinoma--a case report. Anticancer Res. 2007, 27, 499–503. [Google Scholar]
  69. Lakshmanagowda, P.B.; Viswanath, L.; Thimmaiah, N.; Dasappa, L.; Supe, S.S.; Kallur, P. Abscopal effect in a patient with chronic lymphocytic leukemia during radiation therapy: A case report. Cases J. 2009, 2, 204. [Google Scholar] [CrossRef] [Green Version]
  70. Okuma, K.; Yamashita, H.; Niibe, Y.; Hayakawa, K.; Nakagawa, K. Abscopal effect of radiation on lung metastases of hepatocellular carcinoma: A case report. J. Med. Case Rep. 2011, 5, 111. [Google Scholar] [CrossRef] [Green Version]
  71. Cotter, S.E.; Dunn, G.P.; Collins, K.M.; Sahni, D.; Zukotynski, K.A.; Hansen, J.L.; O’Farrell, D.A.; Ng, A.K.; Devlin, P.M.; Wang, L.C. Abscopal effect in a patient with metastatic Merkel cell carcinoma following radiation therapy: Potential role of induced antitumor immunity. Arch. Dermatol. 2011, 147, 870–872. [Google Scholar] [CrossRef]
  72. Tubin, S.; Casamassima, F.; Menichelli, C.; Pastore, G.; Fanelli, A.; Crisci, R. A case report on metastatic thyroid carcinoma: Radiation-induced bystander or abscopal effect? J. Cancer Sci. Ther. 2012, 4, 408–411. [Google Scholar] [CrossRef] [Green Version]
  73. Ishiyama, H.; Teh, B.S.; Ren, H.; Chiang, S.; Tann, A.; Blanco, A.I.; Paulino, A.C.; Amato, R. Spontaneous regression of thoracic metastases while progression of brain metastases after stereotactic radiosurgery and stereotactic body radiotherapy for metastatic renal cell carcinoma: Abscopal effect prevented by the blood-brain barrier? Clin. Genitourin. Cancer 2012, 10, 196–198. [Google Scholar] [CrossRef]
  74. Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [Green Version]
  75. Siva, S.; Callahan, J.; MacManus, M.P.; Martin, O.; Hicks, R.J.; Ball, D.L. Abscopal [corrected] effects after conventional and stereotactic lung irradiation of non-small-cell lung cancer. J. Thorac. Oncol. 2013, 8, e71–e72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Golden, E.B.; Demaria, S.; Schiff, P.B.; Chachoua, A.; Formenti, S.C. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol. Res. 2013, 1, 365–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Stamell, E.F.; Wolchok, J.D.; Gnjatic, S.; Lee, N.Y.; Brownell, I. The abscopal effect associated with a systemic anti-melanoma immune response. Int. J. Radiat. Oncol. Biol. Phys. 2013, 85, 293–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Thallinger, C.; Prager, G.; Ringl, H.; Zielinski, C. Abscopal-Effekt in der Therapie des malignen Melanoms [Abscopal effect in the treatment of malignant melanoma]. Hautarzt 2015, 66, 545–548. [Google Scholar] [CrossRef]
  79. Joe, M.B.; Lum, J.J.; Watson, P.H.; Tonseth, R.P.; McGhie, J.P.; Truong, P.T. Radiation generates an abscopal response and complete resolution of metastatic squamous cell carcinoma of the anal canal: A case report. J. Gastrointest. Oncol. 2017, 8, E84–E89. [Google Scholar] [CrossRef] [Green Version]
  80. Sperduto, W.; King, D.M.; Watanabe, Y.; Lou, E.; Sperduto, P.W. Case Report of Extended Survival and Quality of Life in a Melanoma Patient with Multiple Brain Metastases and Review of Literature. Cureus 2017, 9, e1947. [Google Scholar] [CrossRef] [Green Version]
  81. Van Gysen, K.; Kneebone, A.; Eade, T.; Guminski, A.; Hruby, G. Advanced Renal Cell Cancer and Low-Dose Palliative Radiation Treatment: A Case of a Substantial and Sustained Treatment Response. Case Rep. Oncol. 2018, 11, 756–762. [Google Scholar] [CrossRef]
  82. Bruton Joe, M.; Truong, P.T. Abscopal Effect after Palliative Radiation Therapy for Metastatic Adenocarcinoma of the Esophagus. Cureus 2018, 10, e3089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chantharasamee, J.; Treetipsatit, J. Metastatic Melanoma of Uncertain Primary with 5-Year Durable Response after Conventional Therapy: A Case Report with Literature Review. Case Rep. Oncol. Med. 2018, 2018, 7289896. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, M.J.; Wu, S.; Daud, A.I.; Yu, S.S.; Yom, S.S. In-field and abscopal response after short-course radiation therapy in patients with metastatic Merkel cell carcinoma progressing on PD-1 checkpoint blockade: A case series. J. Immunother. Cancer 2018, 6, 43. [Google Scholar] [CrossRef] [PubMed]
  85. Tsui, J.M.; Mihalcioiu, C.; Cury, F.L. Abscopal Effect in a Stage IV Melanoma Patient who Progressed on Pembrolizumab. Cureus 2018, 10, e2238. [Google Scholar] [CrossRef] [Green Version]
  86. Bonilla, C.E.; Esguerra, J.; Mendoza Díaz, S.; Álvarez, A.; Morales, R.L. Abscopal Effect After Palliative Radiotherapy in a Patient with a Gastric Adenocarcinoma Disseminated to Retroperitoneal Space: Case Report from a Latin American Reference Center and Review of the Literature. Cureus 2019, 11, e6235. [Google Scholar] [CrossRef] [Green Version]
  87. Brenneman, R.J.; Sharifai, N.; Fischer-Valuck, B.; Hassanzadeh, C.; Guzelian, J.; Chrisinger, J.; Michalski, J.M.; Oppelt, P.; Baumann, B.C. Abscopal Effect Following Proton Beam Radiotherapy in a Patient With Inoperable Metastatic Retroperitoneal Sarcoma. Front. Oncol. 2019, 9, 922. [Google Scholar] [CrossRef]
  88. Shinde, A.; Novak, J.; Freeman, M.L.; Glaser, S.; Amini, A. Induction of the Abscopal Effect with Immunotherapy and Palliative Radiation in Metastatic Head and Neck Squamous Cell Carcinoma: A Case Report and Review of the Literature. Cureus 2019, 11, e4201. [Google Scholar] [CrossRef] [Green Version]
  89. Abbas, W.; Goel, V.; Verma, A.; Gupta, V.G.; Rao, R.R. Harnessing the Immunomodulatory Effects of Radiation in Urinary Bladder Cancer. Cureus 2019, 11, e4108. [Google Scholar] [CrossRef] [Green Version]
  90. Barsky, A.R.; Cengel, K.A.; Katz, S.I.; Sterman, D.H.; Simone, C.B., 2nd. First-ever Abscopal Effect after Palliative Radiotherapy and Immuno-gene Therapy for Malignant Pleural Mesothelioma. Cureus 2019, 11, e4102. [Google Scholar] [CrossRef] [Green Version]
  91. Kim, J.O.; Kim, C.A. Abscopal Resolution of a Hepatic Metastasis in a Patient with Metastatic Cholangiocarcinoma Following Radical Stereotactic Body Radiotherapy to a Synchronous Early Stage Non-small Cell Lung Cancer. Cureus 2019, 11, e4082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yaguchi, D.; Ichikawa, M.; Ito, M.; Okamoto, S.; Kimura, H.; Watanabe, K. Dramatic response to nivolumab after local radiotherapy in pulmonary pleomorphic carcinoma with rapid progressive post-surgical recurrence. Thorac. Cancer. 2019, 10, 1263–1266. [Google Scholar] [CrossRef]
  93. D’Andrea, M.A.; Reddy, G.K. Extracranial systemic antitumor response through the abscopal effect induced by brain radiation in a patient with metastatic melanoma. Radiat. Oncol. J. 2019, 37, 302–308. [Google Scholar] [CrossRef] [PubMed]
  94. Matsushita, Y.; Nakamura, K.; Furuse, H.; Ichinohe, K.; Miyake, H. Marked response to nivolumab combined with external radiation therapy for metastatic renal cell carcinoma: Report of two cases. Int. Cancer. Conf. J. 2018, 8, 29–32. [Google Scholar] [CrossRef] [PubMed]
  95. Moran, A.; Azghadi, S.; Maverakis, E.M.; Christensen, S.; Dyer, B.A. Combined Immune Checkpoint Blockade and Stereotactic Ablative Radiotherapy Can Stimulate Response to Immunotherapy in Metastatic Melanoma: A Case Report. Cureus 2019, 11, e4038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Sohal, R.J.; Sohal, S.; Wazir, A.; Benjamin, S. Mucosal Melanoma: A Rare Entity and Review of the Literature. Cureus 2020, 12, e9483. [Google Scholar] [CrossRef]
  97. Ellerin, B.E.; Demandante, C.; Martins, J.T. Pure abscopal effect of radiotherapy in a salivary gland carcinoma: Case report, literature review, and a search for new approaches. Cancer Radiother. 2020, 24, 226–246. [Google Scholar] [CrossRef]
  98. Hori, K.; Hirohashi, Y.; Aoyagi, T.; Taniguchi, N.; Murakumo, M.; Miyata, H.; Torigoe, T.; Abe, T.; Shinohara, N.; Morita, K. Abscopal effect following nivolumab induction in a patient with metastatic renal cell carcinoma-unique pathological features of the primary specimen: A case report. Exp. Ther. Med. 2020, 19, 1903–1907. [Google Scholar] [CrossRef]
  99. Ku, G.Y.; Yuan, J.; Page, D.B.; Schroeder, S.E.; Panageas, K.S.; Carvajal, R.D.; Chapman, P.B.; Schwartz, G.K.; Allison, J.P.; Wolchok, J.D. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: Lymphocyte count after 2 doses correlates with survival. Cancer 2010, 116, 1767–1775. [Google Scholar] [CrossRef]
  100. Callahan, M.K.; Wolchok, J.D.; Allison, J.P. Anti-CTLA-4 antibody therapy: Immune monitoring during clinical development of a novel immunotherapy. Semin. Oncol. 2010, 37, 473–484. [Google Scholar] [CrossRef] [Green Version]
  101. Delyon, J.; Mateus, C.; Lefeuvre, D.; Lanoy, E.; Zitvogel, L.; Chaput, N.; Roy, S.; Eggermont, A.M.; Routier, E.; Robert, C. Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: An early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann. Oncol. 2013, 24, 1697–1703. [Google Scholar] [CrossRef] [PubMed]
  102. Jungbluth, A.A.; Chen, Y.T.; Stockert, E.; Busam, K.J.; Kolb, D.; Iversen, K.; Coplan, K.; Williamson, B.; Altorki, N.; Old, L.J. Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int. J. Cancer 2001, 92, 856–860. [Google Scholar] [CrossRef] [PubMed]
  103. Carthon, B.C.; Wolchok, J.D.; Yuan, J.; Kamat, A.; Ng Tang, D.S.; Sun, J.; Ku, G.; Troncoso, P.; Logothetis, C.J.; Allison, J.P.; et al. Preoperative CTLA-4 blockade: Tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 2010, 16, 2861–2871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Hodge, J.W.; Sharp, H.J.; Gameiro, S.R. Abscopal regression of antigen disparate tumors by antigen cascade after systemic tumor vaccination in combination with local tumor radiation. Cancer Biother. Radiopharm. 2012, 27, 12–22. [Google Scholar] [CrossRef] [Green Version]
  105. Camphausen, K.; Moses, M.A.; Ménard, C.; Sproull, M.; Beecken, W.D.; Folkman, J.; O’Reilly, M.S. Radiation abscopal antitumor effect is mediated through p53. Cancer Res. 2003, 63, 1990–1993. [Google Scholar]
  106. Blanquicett, C.; Saif, M.W.; Buchsbaum, D.J.; Eloubeidi, M.; Vickers, S.M.; Chhieng, D.C.; Carpenter, M.D.; Sellers, J.C.; Russo, S.; Diasio, R.B.; et al. Antitumor efficacy of capecitabine and celecoxib in irradiated and lead-shielded, contralateral human BxPC-3 pancreatic cancer xenografts: Clinical implications of abscopal effects. Clin. Cancer Res. 2005, 11, 8773–8781. [Google Scholar] [CrossRef] [Green Version]
  107. Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef]
  108. Yasuda, K.; Nirei, T.; Tsuno, N.H.; Nagawa, H.; Kitayama, J. Intratumoral injection of interleukin-2 augments the local and abscopal effects of radiotherapy in murine rectal cancer. Cancer Sci. 2011, 102, 1257–1263. [Google Scholar] [CrossRef]
  109. Strigari, L.; Mancuso, M.; Ubertini, V.; Soriani, A.; Giardullo, P.; Benassi, M.; D’Alessio, D.; Leonardi, S.; Soddu, S.; Bossi, G. Abscopal effect of radiation therapy: Interplay between radiation dose and p53 status. Int. J. Radiat. Biol. 2014, 90, 248–255. [Google Scholar] [CrossRef]
  110. Aravindan, S.; Natarajan, M.; Ramraj, S.K.; Pandian, V.; Khan, F.H.; Herman, T.S.; Aravindan, N. Abscopal effect of low-LET γ-radiation mediated through Rel protein signal transduction in a mouse model of nontargeted radiation response. Cancer Gene Ther. 2014, 21, 54–59. [Google Scholar] [CrossRef] [Green Version]
  111. Xia, L.; Wu, H.; Qian, W. Irradiation enhanced the effects of PD-1 blockade in brain metastatic osteosarcoma. J. Bone Oncol. 2018, 12, 61–64. [Google Scholar] [CrossRef] [PubMed]
  112. Markovsky, E.; Budhu, S.; Samstein, R.M.; Li, H.; Russell, J.; Zhang, Z.; Drill, E.; Bodden, C.; Chen, Q.; Powell, S.N.; et al. An Antitumor Immune Response Is Evoked by Partial-Volume Single-Dose Radiation in 2 Murine Models. Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 697–708. [Google Scholar] [CrossRef] [PubMed]
  113. Pfannenstiel, L.W.; McNeilly, C.; Xiang, C.; Kang, K.; Diaz-Montero, C.M.; Yu, J.S.; Gastman, B.R. Combination PD-1 blockade and irradiation of brain metastasis induces an effective abscopal effect in melanoma. Oncoimmunology 2018, 8, e1507669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dudzinski, S.O.; Cameron, B.D.; Wang, J.; Rathmell, J.C.; Giorgio, T.D.; Kirschner, A.N. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J. Immunother. Cancer 2019, 7, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Vijayakumar, G.; Palese, P.; Goff, P.H. Oncolytic Newcastle disease virus expressing a checkpoint inhibitor as a radioenhancing agent for murine melanoma. EBioMedicine 2019, 49, 96–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Kikuchi, R.; Sukhbaatar, A.; Sakamoto, M.; Mori, S.; Kodama, T. A model system for studying superselective radiotherapy of lymph node metastasis in mice with swollen lymph nodes. Clin. Transl. Radiat. Oncol. 2019, 20, 53–57. [Google Scholar] [CrossRef] [Green Version]
  117. Baba, K.; Nomura, M.; Ohashi, S.; Hiratsuka, T.; Nakai, Y.; Saito, T.; Kondo, Y.; Fukuyama, K.; Kikuchi, O.; Yamada, A.; et al. Experimental model for the irradiation-mediated abscopal effect and factors influencing this effect. Am. J. Cancer Res. 2020, 10, 440–453. [Google Scholar]
  118. Zhang, P.; Darmon, A.; Marill, J.; Mohamed Anesary, N.; Paris, S.; Zhang, P.; Darmon, A.; Marill, J.; Mohamed Anesary, N.; Paris, S. Radiotherapy-Activated Hafnium Oxide Nanoparticles Produce Abscopal Effect in a Mouse Colorectal Cancer Model. Int. J. Nanomed. 2020, 15, 3843–3850. [Google Scholar] [CrossRef]
  119. Antohe, M.; Nedelcu, R.I.; Nichita, L.; Popp, C.G.; Cioplea, M.; Brinzea, A.; Hodorogea, A.; Calinescu, A.; Balaban, M.; Ion, D.A.; et al. Tumor infiltrating lymphocytes: The regulator of melanoma evolution. Oncol. Lett. 2019, 17, 4155–4161. [Google Scholar] [CrossRef]
  120. Gata, V.A.; Lisencu, C.I.; Vlad, C.I.; Piciu, D.; Irimie, A.; Achimas-Cadariu, P. Tumor infiltrating lymphocytes as a prognostic factor in malignant melanoma. Review of the literature. J. BUON 2017, 22, 592–598. [Google Scholar]
  121. Ugurel, S.; Rohmel, J.; Ascierto, P.A.; Flaherty, K.T.; Grob, J.J.; Hauschild, A.; Larkin, J.; Long, G.V.; Lorigan, P.; McArthur, G.A.; et al. Survival of patients with advanced metastatic melanoma: The impact of novel therapies-update 2017. Eur. J. Cancer. 2017, 83, 247–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Da, L.J.; Teng, Y.J.; Wang, N.; Zaguirre, K.; Liu, Y.T.; Qi, Y.L.; Song, F.X. Organ-Specific Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitor Monotherapy Versus Combination Therapy in Cancer: A Meta-Analysis of Randomized Controlled Trials. Front. Pharm. 2020, 10, 1671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rodríguez-Ruiz, M.E.; Vanpouille-Box, C.; Melero, I.; Formenti, S.C.; Demaria, S. Immunological Mechanisms Responsible for Radiation-Induced Abscopal Effect. Trends Immunol. 2018, 39, 644–655. [Google Scholar] [CrossRef] [PubMed]
  124. Brix, N.; Tiefenthaller, A.; Anders, H.; Belka, C.; Lauber, K. Abscopal, immunological effects of radiotherapy: Narrowing the gap between clinical and preclinical experiences. Immunol. Rev. 2017, 280, 249–279. [Google Scholar] [CrossRef] [PubMed]
  125. Twyman-Saint Victor, C.; Rech, A.J.; Maity, A.; Rengan, R.; Pauken, K.E.; Stelekati, E.; Benci, J.L.; Xu, B.; Dada, H.; Odorizzi, P.M.; et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015, 520, 373–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Simbawa, E.; Al-Johani, N.; Al-Tuwairqi, S. Modeling the Spatiotemporal Dynamics of Oncolytic Viruses and Radiotherapy as a Treatment for Cancer. Comput. Math. Methods Med. 2020, 2020, 3642654. [Google Scholar] [CrossRef]
  127. Thomas, S.; Kuncheria, L.; Roulstone, V.; Kyula, J.N.; Mansfield, D.; Bommareddy, P.K.; Smith, H.; Kaufman, H.L.; Harrington, K.J.; Coffin, R.S. Development of a new fusion-enhanced oncolytic immunotherapy platform based on herpes simplex virus type 1. J. Immunother. Cancer 2019, 7, 214. [Google Scholar] [CrossRef] [Green Version]
  128. Werthmöller, N.; Frey, B.; Rückert, M.; Lotter, M.; Fietkau, R.; Gaipl, U.S. Combination of ionising radiation with hyperthermia increases the immunogenic potential of B16-F10 melanoma cells in vitro and in vivo. Int. J. Hyperther. 2016, 32, 23–30. [Google Scholar] [CrossRef] [Green Version]
  129. Zhang, H.; Wang, P.; Wang, X.; Shi, L.; Fan, Z.; Zhang, G.; Yang, D.; Bahavar, C.F.; Zhou, F.; Chen, W.R.; et al. Antitumor Effects of DC Vaccine With ALA-PDT-Induced Immunogenic Apoptotic Cells for Skin Squamous Cell Carcinoma in Mice. Technol. Cancer Res. Treat. 2018, 17, 1533033818785275. [Google Scholar] [CrossRef] [Green Version]
  130. Cammarata, F.P.; Forte, G.I.; Broggi, G.; Bravatà, V.; Minafra, L.; Pisciotta, P.; Calvaruso, M.; Tringali, R.; Tomasello, B.; Torrisi, F.; et al. Molecular Investigation on a Triple Negative Breast Cancer Xenograft Model Exposed to Proton Beams. Int. J. Mol. Sci. 2020, 21, 6337. [Google Scholar] [CrossRef]
  131. Huang, Y.; Dong, Y.; Zhao, J.; Zhang, L.; Kong, L.; Lu, J.J. Comparison of the effects of photon, proton and carbon-ion radiation on the ecto-calreticulin exposure in various tumor cell lines. Ann. Transl. Med. 2019, 7, 542. [Google Scholar] [CrossRef]
  132. Takahashi, J.; Misawa, M.; Murakami, M.; Mori, T.; Nomura, K.; Iwahashi, H. 5-Aminolevulinic acid enhances cancer radiotherapy in a mouse tumor model. Springerplus 2013, 2, 602. [Google Scholar] [CrossRef] [Green Version]
  133. Yamamoto, J.; Ogura, S.; Shimajiri, S.; Nakano, Y.; Akiba, D.; Kitagawa, T.; Ueta, K.; Tanaka, T.; Nishizawa, S. 5-aminolevulinic acid-induced protoporphyrin IX with multi-dose ionizing irradiation enhances host antitumor response and strongly inhibits tumor growth in experimental glioma in vivo. Mol. Med. Rep. 2015, 11, 1813–1819. [Google Scholar] [CrossRef]
  134. Takahashi, J.; Murakami, M.; Mori, T.; Iwahashi, H. Verification of radiodynamic therapy by medical linear accelerator using a mouse melanoma tumor model. Sci. Rep. 2018, 8, 2728. [Google Scholar] [CrossRef]
  135. Marconi, R.; Strolin, S.; Bossi, G.; Strigari, L. A meta-analysis of the abscopal effect in preclinical models: Is the biologically effective dose a relevant physical trigger? PLoS ONE 2017, 12, e0171559. [Google Scholar] [CrossRef] [Green Version]
Table 1. Details of published case reports on abscopal effects.
Table 1. Details of published case reports on abscopal effects.
HistopathologyAgeGenderRTTime for AbscopalMarkersReference
Adenocarcinoma of unknown origin35F30 Gy, 20 fr2 weeks Ehlers et al.,1973 [63]
Lymphoma44M40 Gy, 20 frNRnumbers and percentages of total leukocyte, band neutrophils, segmented neutrophils, lymphocytes, monocytes, eosinophils,.basophils,Antoniades et al., 1977 [64]
Lymphocytic lymphoma40M40 Gy, 20 frNR
Mixed-cellularity Hodgkin lymphomaNRNR35 Gy, 28 daysNR Rees et al., 1981 [65]
Hepatocellular carcinoma76M36 Gy, NR10 Monthsserum level of IL-1β, IL-2, IL-4, IL-6, HGF and TNF-αOhba et al., 1998 [66]
Renal cell carcinoma 83F32 Gy, 4 fr2 years Wersäll et al., 2006 [67]
Renal cell carcinoma 64FNRNR
Renal cell carcinoma 69MNRNR
Renal cell carcinoma 55F32 Gy, 4 fr5 months
Uterine cervix69F1.8 Gy, 16 fr
2.0 Gy 21 fr
6 Gy,4 fr (total 74.8 Gy)
NRserum levels of squamous cell carcinoma (SCC) antigenTakaya et al., 2007 [68]
Chronic lymphocytic leukemia65F24 Gy, 12 frduring treatment Lakshmanagowda et al., 2009 [69]
Hepatocellular carcinoma63M60.25 Gy, 27 frNR Okuma et al., 2011 [70]
Merkel cell carcinoma70M12 Gy, 2 fr1 month Cotter et al., 2011 [71]
Medullary thyriod carcinoma72M30 Gy, 3 fr1 month Tubin et al., 2012 [72]
Renal cell carcinoma 61M40 Gy, 5 fr1 month Ishiyama et al., 2012 [73]
Melanoma33F28.5 Gy, 3 fr4 monthslevels of CD4+ ICOShigh cells, HLA-DR expression on monocytes, MDSCs (CD14+ HLA-DRlow) of peripheral-blood mononuclear cellsPostow et al., 2012 [74]
Adenocarcinoma of lung78F26 Gy, 1 fr12 months Siva et al., 2013 [75]
Adenocarcinoma of lung64M30 Gy, 5 fr2.5 monthsthe absolute lymphocyte count (ALC), the absolute eosinophil count (AEC), white blood cells (WBCs), carcinoembryonic antigen (CEA) of peripheral-bloodGolden et al., 2013 [76]
Melanoma67M24 Gy, 3 fr8 monthsmelanoma antigen A3 (MEGA3), PAS domain containing 1 (PASD1) level of serumStamell et al., 2013 [77]
Melanoma44M30 Gy, 10 fr2 months Thallinger et al., 2014 [78]
Squamous carcinoma of the anal canal57F54 Gy, 30 fr1 monthPD-1, PD-L1, CD163, CD3, CD8 expression of tumor infiltrating lymphocytes (TILs)Joe et al., 2017 [79]
Melanoma36F20 or 24 Gy, 1 fr9 months Sperduto et al., 2017 [80]
Renal cell carcinoma66F36 Gy, 12 fr1 month van Gysen et al., 2018 [81]
Esophageal adenocarcinoma74M30 Gy, 10 fr2 months Bruton et al., 2018 [82]
Malignant melanoma of unknown primary51F20 Gy, NR Chantharasamee et al., 2018 [83]
Merkel cell carcinoma69M8 Gy, 1 fr12 months Xu et al., 2018 [84]
Merkel cell carcinoma72F8 Gy, 1 fr2 months
Mucosal melanom65F24 Gy, 3 fr1 month Tsui et al., 2018 [85]
Gastric adenocarcinoma78F30 Gy, 10 fr3 months Bonilla et al., 2019 [86]
Retroperitoneal sarcomas67F50 Cobalt Gray Equivalents, 25 fr5 monthsPD-L1, CD4, CD8 expression of tumor TILsBrenneman et al., 2019 [87]
Head and neck squamous cell carcinoma75M3.7 Gy twice a day, 2 fr
(total 14.8 Gy)
2 weeks Shinde et al., 2019 [88]
Urinary bladder cancer65M30 Gy, 12 fr4 months Abbas et al., 2019 [89]
Malignant pleural mesothelioma67M30 Gy, 10 fr Barsky et al., 2019 [90]
Cholangiocarcinoma 70M48 Gy, 4 fr3 months Kim et al., 2019 [91]
Pulmonary pleomorphic carcinoma63M30 Gy, NR PD-L1 expression of tumorYaguchi et al., 2019 [92]
Melanoma42F30 Gy, 15 fr3 weeksERCC1, MLH1, MSH2, MSH6, PMS2, TUBB3, PDL-1, TrK A/B/C, MGMT expression of tumorD’Andrea et al., 2019 [93]
Renal cell carcinoma62M36 Gy, 12 fr1.5 months Matushita et al., 2019 [94]
Renal cell carcinoma71M66 Gy, 33 fr1.5 months
Melanoma71M50 Gy, 5 fr1 month Moran et al., 2019 [95]
Mucosal melanoma66M25 Gy, 5 fr4 months Sohal et al., 2020 [96]
Salivary gland carcinoma84F50 Gy, 20 fr2 weeks Ellerin et al., 2020 [97]
Renal cell carcinoma 40F30 or 40 Gy, 10 fr6 monthsHLA class1, CD8, PD-L1 expression of tumorHori et al., 2020 [98]
F, female; M, male; fr, fraction; NR, not reported; MDSCs, myeloid-derived suppressor cells.
Table 2. Animal models used in past studies for evaluating abscopal effects.
Table 2. Animal models used in past studies for evaluating abscopal effects.
Mouse StrainAgeCancer Cell LineCell TypeCondition of InoculationRT Treatment (Total Dose, Fraction)EndpointNoteReference
C57BL/68 weeksMC38mouse colon adenocarcinomaright flank (MC38-CEA+), left frank (MC38-CEA-)8 Gy, 1 frtumor growth Hodge et al., 2012 [104]
C57BL/6 transgenic for human CEA LL/2mouse lung adenocarcinoma,right frank (LL2-CEA+), intravenously (LL2-CEA+)125I -brachytherapy 72 h exposurepulmonary metastasislung metastasis model
C57BL/6, p53 null B6.129S2- Trp53 tm1Tyj4–6 weeksLLC-LM, T241Lewis lung carcinoma, fibrosarcomamidline dorsum24 Gy, 12 frtumor growthirradiate non tumor site, legCamphausen et al., 2003 [105]
NCr nu/nu BxPC-3 pancreatic carcinoma cellsright and left flank10 Gy, 5 frtumor growthNude mouseBlanquicett et al., 2005 [106]
BALB/c, C57BL/66–8 weeksTSA, MCA38mouse breast carcinoma, mouse colon carcinomaright and left flank20 Gy, 1 fr
24 Gy, 3 fr
30 Gy, 5 fr
tumor growth Dewan et al., 2009 [107]
BALB/c8 weekscolon26mouse colon adenocarcinomaleft frank, after 3 weeks intra-splenic injection20 Gy, 10 frtumor growth (liver weight)liver metastasis modelYasuda et al., 2011 [108]
CD1 nu/nu HCT116, A549human colorectal cancer, human lung adenocarcinomaright and left flank10 Gy, 2 fr
20 Gy, 3 fr
tumor growthnude mouseStrigari et al., 2014 [109]
C57BL/64 weeksnone 2 Gy, 1 fr
10 Gy, 1 fr
10 Gy, 5 fr
norml tissue responseabscopal model without cancerAravindan et al., 2014 [110]
BALB/c 4T1, TSAmouse mammary carcinoma, mammaryadenocarcinomaright and lefl flank30 Gy, 1frtumor growth Vanpouille-Box et al., 2015 [55]
BALB/c6 weeksK7M2mouse osteosarcomasubcutaneous, right frontal lobes40 Gy (2Gy × 4, five consecutive days)immune markers from peripheral broodbrain metastasis, irradiate for subcutaneous tumorXia et al., 2018 [111]
BALB/c,
C57BL/6, athymic nude mice
12 weeks67NRbreast cancer, Lewis lung carcinomaright and left mammary fat pad10 Gy, 1 fr
15 Gy, 1 fr
tumor growth, survivalcompare the response of immunocompetent mouse with nude mouseMarkovsky et al., 2019 [112]
C57BL/6 B16-F10, D4Mmouse melanomasubcutaneous, right frontal lobes8 Gy, 4 frtumor growthbrain metastasis, irradiate for brain tumorPfannenstiel et al., 2018 [113]
FVB (JAX) Myc-CaP mouse prostate cancerfrank and leg20 Gy, 2 frtumor growth, survival Dudzinski et al., 2019 [114]
C57BL/6 B16-F10mouse melanomaright and left flank5 Gy, 1 fr
10 Gy, 1 fr
20 Gy, 1 fr
tumor growth, survival Vijayakumar et al., 2019 [115]
MXH10/Mo/Lpr FM3A-Lucmouse mamary carcinoma cellslymph node8 Gy, 1 frtumor growthlymph node metastasis modelKikuchi et al., 2019 [116]
C57BL/6 MC38, B16F10mouse colon adenocarcinoma cell, mouse melanoma upper and lower dorsum6 Gy, 3 fr
24 Gy, 3 fr
tumor growth Baba et al., 2020 [117]
BALB/c6 weeksCT26.WTmouses colon carcinomaright and left flank12 Gy, 3 frtumor growth Zhang et al., 2020 [118]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Takahashi, J.; Nagasawa, S. Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review. Int. J. Mol. Sci. 2020, 21, 9324. https://doi.org/10.3390/ijms21239324

AMA Style

Takahashi J, Nagasawa S. Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review. International Journal of Molecular Sciences. 2020; 21(23):9324. https://doi.org/10.3390/ijms21239324

Chicago/Turabian Style

Takahashi, Junko, and Shinsuke Nagasawa. 2020. "Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review" International Journal of Molecular Sciences 21, no. 23: 9324. https://doi.org/10.3390/ijms21239324

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop