Next Article in Journal
COVID-19 in Children with Down Syndrome: Data from the Trisomy 21 Research Society Survey
Next Article in Special Issue
Combined First Month Body Weight Loss and Development of Tolerance as Predictors of 6-Month Efficacy of Mazindol in Mild and Moderate Obese Subjects
Previous Article in Journal
Projections of Primary and Revision Shoulder Arthroplasty until 2040: Facing a Massive Rise in Fracture-Related Procedures
Previous Article in Special Issue
Differential Expression of the Sphingolipid Pathway Is Associated with Sensitivity to the PP2A Activator FTY720 in Colorectal Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Abscopal Effects in Metastatic Cancer: Is a Predictive Approach Possible to Improve Individual Outcomes?

by
Barbara Link
1,
Adriana Torres Crigna
1,
Michael Hölzel
2,
Frank A. Giordano
1 and
Olga Golubnitschaja
3,*
1
Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
2
Institute of Experimental Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
3
Predictive, Preventive, Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(21), 5124; https://doi.org/10.3390/jcm10215124
Submission received: 28 September 2021 / Revised: 20 October 2021 / Accepted: 29 October 2021 / Published: 31 October 2021
(This article belongs to the Special Issue Predictive Diagnostics and Personalized Treatment)

Abstract

:
Patients with metastatic cancers often require radiotherapy (RT) as a palliative therapy for cancer pain. RT can, however, also induce systemic antitumor effects outside of the irradiated field (abscopal effects) in various cancer entities. The occurrence of the abscopal effect is associated with a specific immunological activation in response to RT-induced cell death, which is mainly seen under concomitant immune checkpoint blockade. Even if the number of reported apscopal effects has increased since the introduction of immune checkpoint inhibition, its occurrence is still considered rare and unpredictable. The cases reported so far may nevertheless allow for identifying first biomarkers and clinical patterns. We here review biomarkers that may be helpful to predict the occurrence of abscopal effects and hence to optimize therapy for patients with metastatic cancers.

1. Introduction

Radiotherapy (RT) has been widely used as an extremely effective anticancer treatment resulting in local tumor management. The targeted ionizing irradiation therapeutic effect is principally described to exert direct localized cell death through deoxyribonucleic acid (DNA) damage to promptly proliferating cells. However, preclinical studies have recognized irradiation to induce tumor recession at non-irradiated distant tumor milieus by means of tumor-specific immunity induction or immunogenic cell death (ICD) [1]. This systemic response is called the “abscopal effect” (AbE), derived from the Latin words ab (position away from) and scopus (mark or target), first introduced by Mole in 1953 and later enhanced by Andrews describing of distant normal tissue effects [2,3]. In modern oncology, AbE is becoming increasingly important as the association between and understanding of localized irradiation and immune-mediated systemic antitumor effects are enhanced. Nevertheless, the occurrence of AbE is mainly described in isolated case reports.
Here, we overview the present state of clinical data concerning AbE. The aim of this review is to provide an overview of the AbE in different cancer entities, along with its employment in combination with chemo- and immunotherapy and identify potential predictors/biomarkers associated with RT immunogenic features for patient stratification and treatment optimization.

2. Historical Background

The abscopal effect was first described and named by Mole in 1953 in a preclinical model of metastatic cancer [3]. Later his definition was broadened by Andrews to include radiation-related effects of both distant tumor and normal tissue [2,3]. Furthermore, Formenti et al. determined the abscopal response as a size shrinkage of 30% of a non-irradiated metastasis irrespective of other lesions [1]. AbE must be distinguished from bystander effects, the former being an effect that occurs at distance from the original radiation field, whereas the latter occurs as a local reaction due to radiation-induced signals from nearby irradiated cells [4].
Since its first description, AbE has been rarely observed in the clinical setting despite the millions of patients treated worldwide with local irradiation of tumor tissue in the absence of any systemic therapy. From 1969 to 2018, only 47 cases for multiple metastatic cancers including melanoma, renal cell carcinoma, lymphoma, breast cancer, and hepatocellular carcinoma following RT have been reported [5,6]. In Table 1, we provide case reports of AbE after RT in which no immunotherapy was utilized. We performed a literature search in PubMed up to March 2021, using as search criteria “abscopal effect” in the title and/or abstract and filtering for case reports. Results revealed nine additional AbE reports correlated with RT only.
The reported AbEs occurred independently from the patient age, treatment dose, fractionation, modality and characteristic of the target lesion. The cases varied widely in ethnicity and age (19–89 years), as well as in the applied radiation total doses (0.45–70 Gy) and doses per fraction (fr) (0.15–26 Gy). Most patients were treated with RT delivered in conventional fractionation (≤3 Gy), whilst only 14 cases received higher fraction doses. Approximately half of the reported cases were treated with RT for their primary site. AbE was observed ranging from two weeks up to several months (1–24 months) after receiving RT [5].
Postow et al. reported for the first time a case of AbE in a female patient that received systemic immunotherapy five years post primary tumor and lung metastasis treatment. Ipilimumab as immunotherapy against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) was administered to treat metastatic thoracic lesions, along with additional palliative RT. Thereby, five months following irradiation, non-targeted hilar and splenic metastatic lesions had regressed leading to minimal stable disease, and suggesting a correlation between AbE and the immune checkpoint blockade (ICB) [49]. With the introduction of the immune checkpoint inhibitor ipilimumab in clinical practice, the number of AbE related publications considerably increased. Adjepong et al. along with Dagoglu et al. thoroughly reviewed a total of 47 cases of AbE in patients with melanoma, lymphoma, renal cell carcinoma, pancreatic cancer, breast cancer, and prostate cancer, among others, treated with radiation in addition to immunotherapy in just six years between 2012 and 2018 [6,50].
In Table 2 we provide case reports of AbE after RT in which immunotherapy was utilized. Our PubMed search identified 60 additional cases of AbE since 2018. Most of these cases (14 cases) were observed in renal cell carcinoma (RCC). Ten cases occur in lung cancer, eight of them in non-small cell lung cancer (NSCLC).
All reported cases had received prior treatments such as chemotherapy or resection surgery of the primary site or metastasis. Only a few patients had received RT previously. Immunotherapy was administered simultaneously with or immediately after RT. In these cases of AbE, the patients received markedly different RT dose schemes (8–70 Gy), fractionation sessions (1–35), and dose per fraction (1.8–28 Gy) with or immediately after additional immunotherapy. The vast majority of patients was treated with moderate hypofractionated (3–6 Gy/fr) or hypofractionated doses (>7 Gy/fr) at metastasis sites.
In fact, most AbEs have been reported in melanoma patients undergoing RT treatment along with immunotherapy (27 cases). Most patients received a total dose of 30 Gy (predominantly 3 Gy/fr), however, the RT schemes differed widely in doses (13.2–66 Gy) and number of fractionation sessions (1–52). Based on the number of AbEs reported after administration of checkpoint inhibitors, the blocking of the immune checkpoints seems to support AbE events.

3. Abscopal Effect Immune-Related Mechanisms

The precise mechanism of AbE remains unidentified, however, there are several independent and dependent mechanisms involved that are induced by certain radiation dose and fractionation schemes.
RT is a highly effective anti-cancer therapy achieved by tumor cell lethality, prompted by irradiation-induced lethal DNA double-strand breaks (DSBs). Furthermore, the generation of free radicals also plays a role, as radiolysis of water in cells leads to production of reactive oxygen species (ROS) producing cascade of DNA damage in tumor microenvironment (TME) cells [101,102].
Recent advances in RT techniques allow for a more accurate delivery of higher doses per fraction [103]. Localized RT presents not only immunosuppressive but also immune-stimulatory effects, inducing the release of immune-stimulating tumor antigens via ICD [104]. Thus, it modulates the TME as a result of the induction of endothelial cell death and vascular damage as well as increased T cell priming in lymphoid tissues [105].
Demaria et al. revealed AbE to be dependent on the immune-mediated action of functional T cells. Hence, immune promotion is mainly supported by T cell priming against novel tumor-specific antigens [106]. Thereby, irradiated tumors can act as an in situ immunization, being functional T lymphocytes essential effector cells in such response [107]. Irradiation-modulated TME releases diverse cytokines and chemokines that recruit and mature antigen-presenting cells (APCs), namely dendritic cells (DCs) and macrophages to the tumor site. Decreasing tumor cells release elevated amounts of antigens, which are recognized and processed by APCs [108,109]. Tumor-related APCs prime antigen-specific effector T cells by presenting tumor peptide antigens to naïve T cells in tumor-draining lymph nodes (LNs) [110]. In fact, cytotoxic T lymphocytes (CTLs) recognize radiation-induced antigens and activated T cells that travel through the bloodstream to combat the residual tumor cells present in the non-irradiated distant metastatic sites which seems to lead to an effective anti-tumor immune response and cancer control [111].
Due to the stress response after the irradiation of the tumor cells, the damaged and dying cells release damage-associated molecular patterns (DAMPs) including heat shock protein 70 (Hsp70), calreticulin, adenosine-5′-triphosphate (ATP), and high mobility group protein B1 (HMGB1) [112]. Hsp70 has been seen to induce resistance against apoptosis when present inside the cells, however, extracellularly, it promotes apoptosis and contributes to an innate and adaptive immune response [113,114]. Calreticulin acts as an ‘eat me’ signal for DCs when exposed to the membrane resulting in enhanced phagocytosis of the cancer cells by DCs and, thus, results in an increased release of tumor-specific antigens (TSAs) [104,109,115]. Moreover, ATP binds to P2X purinoreceptor 7 (P2X7R) on inflammatory cells, activating the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, a multiprotein complex that produces active caspase-1 and stimulates the production of interleukin (IL) 1β and IL-18 [116,117]. Il-1β mediates the antitumor-specific priming of the CD8+ T cells, IFN-γ production as well as the activation and maturation of DCs to APCs [118]. HMGB1 activates DCs, promoting their maturation and ATP release, leading to the enhancement of CD8+ effector T cells antigen presentation and their anti-tumor-specific activation [112,119].
Local RT induces the release of both pro- and antitumor cytokines due to irradiation-induced DNA damage. In the cytoplasm of irradiated cells, cyclic GMP-AMP synthase (cGAS) binds to translocated double-stranded DNA fragments (dsDNA) induced by RT and activates a signaling cascade via the second messenger 2′3′-cGAMP (cGAMP) and stimulator of interferon genes (STING) to produce type I interferons (IFNs) in the TME [120,121,122,123,124]. Radiation-induced interferons play a major role in the therapeutic effect of RT. In fact, irradiated tumor cells produce IFN-β and are, in turn, responsible for the release of IFN-γ by T cells [125,126]. IFN-γ consecutively promote cytotoxic T cell activation along with upregulating programmed cell death protein 1 (PD-1) receptor-ligand 1 (PD-L1) [127]. Following the irradiation-induced release of IFN-γ, there is an upregulation of pro-inflammatory chemokines CXCL10 and CXCL16 which are in turn responsible for the recruitment and activation of T cells into the TME [128,129,130]. ROS production leads to IL-1α, IL-1β and IL-6 production as well as tumor necrosis factor alpha (TNF-α) and transforming growth factor beta (TGF-β) [131].
Moreover, there are various mechanisms that can concomitantly suppress AbE such as those dependent on factors with immune system inhibitory effects. In particular, the environment of the irradiated volume plays an important role in this process. Thus, prevalence of immunosuppressive cytokines and cells present at the post-irradiated site could limit AbE regardless of immunotherapy treatment [132].
The hypoxic milieu within the irradiated tumor promotes recruitment of immunosuppressive regulatory T cells (Tregs) which induce immunosuppression in the TME [133,134]. Moreover, hypoxia impacts the antigen-presenting ability of APCs by reducing major histocompatibility complex (MHC) class I molecules. This reduction is correlated with a lower recognition of cytotoxic T cells and hence a reduced killing of tumor cells by these T cells [135].
In fact, a study correlated the frequency of AbE with the extent of adrenergic stress in a murine model. Enhanced norepinephrine production prevented T cell activation and consequently the abscopal response [136].
In most cases, RT alone is insufficient to overcome the established immune suppressive mechanisms of the TME. The immune effect of irradiation is affected by the applied dose, fractionation, and application volume making prediction of immunological reactions intricate [137]. RT in combination with immunotherapy may be advantageous in overcoming this immune tolerance and enhance AbE patient response. Preclinical studies have already illustrated how important the optimal RT dose, fractionation scheme, administration timing and dosing of the immunotherapeutics are to provide systemic antitumoral effects [106,138,139,140,141,142,143]. Identification of AbE prediction biomarkers may be crucial to develop an individualized medical approach.
Recently, in the course of the ongoing Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic, the effects of infectious diseases on the immune system have come into focus. The virus triggers inflammatory responses in the respiratory tract and induces a broad production of cytokines, which are also associated with the mechanism of AbE [123].
Herrscher et al. reported the first case of AbE in a SARS-CoV-2 infected patient with melanoma. The 84-year-old woman was diagnosed in early 2020 with peritoneal and a nodal recurrence of melanoma which was handled with palliative care. In late 2020, palliative RT was performed for a metastatic cervical node (20 Gy, 5 fractions). The patient was diagnosed with coronavirus disease 2019 (COVID-19) in early 2021. At this time point, metastases presented a shrinkage of 20–25% which may suggest an abscopal response linked to the activation of the innate immune system due to the SARS-CoV-2 infection and their influence on the production of pro-inflammatory cytokines [144].
Spontaneous tumor regression has been described over the last centuries being often correlated with acute bacterial, fungal, viral, or protozoal infections as well as vaccine therapy. In 25–80% of documented cases of spontaneous regression of cancer, the acute infections cause a strong immunological reaction presenting with fever, which in turn supports the regression of the tumor [145,146]. Thus, the increase in the anti-tumor response may also be responsible for the noted AbE after COVID-19 infection.

4. Abscopal Effect in Different Tumor Entities

AbEs have been reported in a variety of cancer types. In studies where additional immune therapy was absent, AbE occurred remarkably in cases of adenocarcinoma of different origins. Nevertheless, AbE were observed more frequently in melanoma patients also treated with immunotherapy. In breast and prostate cancer patients, the preclinical evidence for AbE cases is more robust compared to the frequency of reported clinical cases. Table 3 provides an overview of the frequencies of case reports of ABE in various tumor entities juxtaposed with their worldwide incidence and mortality.

4.1. Adenocarcinoma

Adenocarcinomas are malignancies arising from the epithelial cells of the glandular tissue in multiple sites of the body [148]. They usually originate in organs such as the gastrointestinal tract, esophagus, lung, breast, prostate or pancreas. However, in the case of metastasis, the determination of the primary site of origin is complex. In fact, adenocarcinomas account for 70% of cancers with unknown origin [149]. Twelve cases of AbE were observed under conventional treatment of adenocarcinomas, and 13 additional cases occurred after additional immunotherapy.
Ehlers et al. reported the first clinical case of AbE in papillary adenocarcinoma of unknown origin. The 37-year-old female patient showed metastases in the bilateral neck, right axilla, and mediastinum LN. The bilateral neck and supraclavicular regions were treated with a total dose of 40 Gy in 20 fractions. After two weeks, the non-irradiated right axilla and mediastinum mass regressed, presenting the treated neck node only a partial response [8].
Adenocarcinoma AbE case reports of a digestive origin are underrepresented (three reports in total) considering that adenocarcinoma accounts for 95% of colon and rectal cancers [150]. In fact, Chuang et al. described a case of AbE after whole-brain irradiation in a female patient with metastatic adenocarcinoma of colorectal origin. Along with multiple brain metastases, the patient presented a huge metastatic lung mass. Treatment consisted of 30 Gy in 10 fractions without any adjuvant systemic therapy. The mass in the left lung markedly regressed two months later. These results suggest that AbE inducing factors are able to trespass the blood-brain barrier (BBB) [37]. Bonilla et al. reported a case of a patient with unresectable advanced gastric adenocarcinoma with extensive retroperitoneal LN involvement. The gastric mass and margins were treated with RT (30 Gy total), observing three months later a complete response of the non-irradiated retroperitoneal paraaortic adenopathy and gastro-hepatic ligament along with the tumor markers angiotensin-converting enzyme (ACE), carbohydrate antigen 19-9 (CA19-9) and cancer antigen 125 (CA-125) [41]. Furthermore, another AbE case was described in an adenocarcinoma patient undergoing RT (48 Gy; 24 fractions) at the primary tumor site for recurrent disease, along with adoptive T-cell immunotherapy and DC therapy. Two months after RT the metastatic lesion size had decreased in addition to serum tumor marker CA19-9 [60].
AbE have also been investigated in colon adenocarcinoma preclinical murine models. In fact, Baba et al. revealed an increased AbE in C57BL/6 mice related to the irradiated tumor volume and applied radiation dose, excluding anti-PD-1 immunotherapy [151].
Two cases of AbE in esophageal adenocarcinoma were reported until today. Esophageal adenocarcinoma starts in the mucus glands that line the lower part of the esophagus. When it metastasizes, the tumor cells accumulate in the LNs, lungs, liver, bones, adrenal glands, or brain [152]. Rees et al. reported AbE of all lung metastasis in a patient with lower esophageal adenocarcinoma. After irradiation of the primary esophageal tumor, lung metastases regressed for 14 months before cancer further progressed [13]. More recently, a patient treated with RT and esophagectomy for the primary tumor and associated LNs, presented signs of metastases regression twelve months following treatment [34].

4.2. Lung Cancer

Up to 85% of lung cancers are NSCLCs, with adenocarcinoma being the main subtype [153,154]. The pulmonary cancer cells spread typically to the contralateral lung, adrenal glands, bones, brain, or liver. Even if resection is the standard of care for early-stage NSCLC, RT is utilized as a first-line treatment for elderly, comorbid patients or as a palliative treatment in later stages [155,156,157]. In advanced or metastatic NSCLC, the combination of immunotherapy and RT significantly improves 1- and 3-year overall survival; as well as progression-free survival, though but is also beneficial in adenocarcinomas and PD-L1-negative patients [158].
Rees et al. reported the first AbE in a patient with metastatic lung adenocarcinoma treated with palliative radiation to the left lung and the mediastinum. After two weeks, the patient showed a regression in subcutaneous metastases in the forehead and the left shoulder [13]. Ever since there have been 17 more case reports of AbE in NSCLCs, respectively eight in lung adenocarcinoma. Five AbE cases resulted from irradiation of malignant tissue without support from any immunotherapy [24,36,43], wherein two of these reported cases described AbE as a result of the irradiation of brain metastases [27,38].
One case of NSCLC in the right upper lung lobe and metastases in the brain, left adrenal gland, left lower lung lobe, and the liver was treated with chemotherapy and brain RT. One month after RT, the adrenal, lung and liver lesions had diminished [27]. Furthermore, another patient with brain metastasis was treated with RT (25 Gy, five fractions) resulting in the regression of the primary tumor as well as a metastatic lesion in the left mediastinum one month following treatment [38].
A more recent study described an adenocarcinoma case with an origin in the right upper lung lobe. Three months post irradiation, the patient presented regression of the primary tumor and several non-irradiated LN. Serum carcinoembryonic antigen (CEA) levels were decreased in correlation with AbE, whereas CD8+ lymphocyte counts were low at the time the AbE was documented yet increased with metastases growth [43].
Between 2012 and 2021, twelve AbE cases were reported in pulmonary adenocarcinoma after treatment with RT and immunotherapy. Thereby, the AbE seems to occur after irradiation of the contralateral lung, the brain, liver and metastatic LNs [52,58,59,64,74,87,98]. In fact, four cases were described where patients received RT for their brain metastasis (from two weeks to four months after) with a resulting abscopal regression of the primary lung tumor after receiving anti-PD-L1 (atezolizumab) or anti-PD-1 (pembrolizumab) therapy [77,81,98].

4.3. Kidney Cancer

Renal cell carcinoma represents 3–5% of all new adult malignancies. Localized RCC is normally treated first line by resection with adjuvant immunotherapy or receptor tyrosine kinase (RTK) inhibitor therapy [159,160]. Stereotactic ablative radiotherapy (SABR) is recommended as treatment option for medically inoperable patients, though RT plays mainly a role as a palliative therapeutic option for relieving the symptoms of metastatic lesions [159].
The first AbE in renal cell carcinoma was reported in 1981 by Fairlamb. The 73-year-old female patient was diagnosed with lung, hila, and pubic bone metastasis next to her primary left kidney tumor. After nephrectomy, she was treated with palliative radiation to the groin (40 Gy, 15 fractions) which resulted in regression of the lung lesions two months after RT and remained disease-free for 4.5 years [10]. Since then five AbEs have been observed in RCC without adjuvant immunotherapy, while 18 AbE cases have been reported with predominantly nivolumab (anti-PD-1) as immunotherapy. In 1994, MacManus et al. identified a RCC (right kidney) patient presenting lung and mediastinal metastases. The patient received palliative radiation (20 Gy, 10 fractions) for pain relief, and after six months the paratracheal lymphadenopathy was reduced and lung metastases were non-detectable, suggesting an AbE. Cytokine assays (TNF-α, TNF-β, interferon gamma (IFN-γ), IL-2 receptor, IL-6) performed on a single serum sample during the regression of the metastases did not reveal any significant differences compared to the reference values [14].
Moreover, AbE was detected following irradiation of brain metastasis in a patient that underwent nephrectomy. Computed tomography (CT) examination leads to the identification of multiple metastases in bone, brain, lung, and mediastinum. The patient was treated with stereotactic radiosurgery to a dose of 18 Gy. Additionally, the patients vertebral bone lesion was irradiated with 40 Gy in five fractions. Treatment resulted in the regression of lung and mediastinal disease. Follow-up CT, one month after RT, revealed slight regression of the untreated multiple lung metastases and lymphadenopathy. However, three months following RT the patient developed new brain lesions, suggesting AbE to be organ-dependent and incapable of crossing the BBB [22]. Another report described the case of an RCC patient with multiple mediastinal, retroperitoneal, and cervical LN metastasis. The subject was treated after nephrectomy with the tyrosine kinase inhibitors (sunitinib) followed by a combination of anti-PD-1 antibody (AB) pembrolizumab and stereotactic body radiation therapy (SBRT) (32 Gy, four fractions). The patient achieved a systemic complete response within two months [63].
Most recently, Wong et al. identified nine cases of AbEs in the patient cohort of a descriptive and retrospective study of RCC. Approximately 20% of all observed patients revealed a regression in a non-irradiated tumor. The patients received besides RT either a single-agent anti-PD-1 AB (nivolumab, pembrolizumab) or a combination of anti-CTLA-4 and anti-PD-1 treatment (ipilimumab-nivolumab). RT was delivered in different radiation schemes depending on treatment indication and the location of the irradiated volume. AbE occurred in 26.7% of patients treated with combined immunotherapy and 16.1% of patients treated with a single agent. In addition, the abscopal response patterns were very divergent between the patients. Most patients experienced isolated abscopal responses in one to two lesions, while two patients presented a more extensive response in lung and liver [100].

4.4. Melanoma

The most effective treatment modality for localized melanoma is surgery, but RT plays a prominent role in the management of the disease. ICB is established as a standard of care for advanced melanoma [161]. With the employment of immunotherapy (pembrolizumab and nivolumab) for melanoma treatment in 30–40% of the patients a complete response was observed [162,163]. Adjuvant RT following lymphadenectomy and systemic therapy prevents local and regional recurrence. Thus, RT is highly effective and widely used as a palliative treatment being mostly utilized in brain, lung, liver or bone metastases.
Preclinical studies have investigated the influence of PD-1 expression on the systemic antitumor response induced by RT. A combination of RT in addition to PD 1 blockade results in the complete regression of the irradiated primary tumor as well as a reduction in size of non-irradiated secondary tumors outside of the radiation field. The tumors present infiltration of activated, cytotoxic CD8+ T cells, even in the non-irradiated tumors [164,165].
Since Postow et al. first discovered a correlation between AbE and immune checkpoint inhibitors, 26 other cases of AbEs associated with immunotherapy in melanoma patients have been observed, being the cancer type with the most reported abscopal immune responses [49].
One melanoma patient with melanoma of an unknown primary showed an AbE after conventional chemotherapy and palliative radiotherapy (total dose 20 Gy) for bilateral inguinal LNs. The other metastases were found to have gradually decreased in size, as seen on CT scan [35].
Different studies aiming to determine the occurrence rate for AbE in melanoma cases handled with RT combined with ICB. In a prospective clinical trial in which the combination of RT and systemic immunotherapy (ipilimumab) for melanoma was tested, the occurrence of an AbE was detected in 3 out of 16 patients (18.75%) (NCT01449279) [166]. Similarly, in another study 20–21% of melanoma patients who received RT in addition to ipilimumab exhibited an AbE following treatment [167]. Moreover, eleven cases of AbE were detected in a retrospective analysis of 21 patients with advanced melanoma which progressed after receiving ipilimumab and were subsequently treated with local RT. The median overall survival was extended to 22.4 months as a result of the AbE [54]. Another study reported two oligometastatic melanoma patients treated with anti-PD-1 and SBRT. One patient was diagnosed with BRAF-wild-type melanoma on the upper left leg with three liver metastases, a muscle lesion and a lesion in the left groin. The treatment consisted of RT to the three liver lesions along with adjuvant ICB (nivolumab). The LN metastases presented with a strong exhaustion signature defined by the markers PD-1, PD-L1, T cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte-activation gene 3 (LAG-3), and the transcription factor thymocyte selection-associated high mobility group box protein (TOX). Moreover, a high level of the T cell-attracting chemokines CXCL9, CXCL10, CCL5, and CXCL13 were discovered. The patient rapidly developed a complete tumor regression, ongoing for more than 4.5 years. The other patient, however, was described with an unfavorable pre-treatment immune signature of the tumor mass without evidence of infiltration by exhausted T cells, the targets of anti-PD-1 treatment. The patient presented a primary melanoma in the right chest region followed by a single liver metastasis which were both surgically resected. Thereafter, more recent detected liver metastases were treated with RT (45 Gy, three fractions). A second SBRT was delivered displaying a month later, a complete regression of all metastases, lasting for over 2.5 years [97].

4.5. Breast Cancer

Breast cancer is the most commonly diagnosed cancer with an estimated 2.3 million new cases in 2020 [147]. RT is an effective tool for local tumor control, and decreases the risk of tumor cells dispersing within breast, chest area or axillary LNs. RT is also utilized as palliative treatment of symptomatic lesions in patients with metastatic breast cancer. RT is used to treat limited metastatic sites in only patients for whom surgical intervention is not an option (location of the metastases, general health state).
In fact, a study described an AbE in a patient with metastatic breast cancer, after receiving RT alone. The patient presented with a 10 cm tumor in the right breast and multiple associated metastases in lung, bones and LNs, and was treated with RT alone as a palliative treatment, due to her performance status score of 3–4 in the Eastern Cooperative Oncology Group (ECOG). The applied radiation dose was distributed into 60 Gy to the right breast, 28 Gy to the left femur and 39 Gy to the lumbar vertebrae and sacrum. Following 10 months of RT, the lesions in the lung and LNs showed a prominent regression with normalized serum levels of CEA and cancer antigen 15-3 (CA15-3) [33]. Another report described an AbE in stage IV breast carcinoma after multiple fractions of high dose radiation [39].
The combination of RT and immunotherapy was also tested in metastatic breast cancer in clinical trials. Formenti et al. reported the results of a clinical trial investigating the different doses of a TGF-β inhibitor with RT (NCT01401062). Twenty-three patients with at least three distinct metastatic sites were randomized in two treatment arms. As a result, one patient with triple-negative breast cancer (TNBC), invasive breast cancer with limited treatment options and worse prognosis, showed a 28% reduction in the non-irradiated lesions. Unfortunately, no more details of this case were reported [168]. A separate study regarding patients with TNBC, performed post-mastectomy RT and reported a beneficial impact on 3-year breast cancer-specific survival which was dependent on the pathological stage of the patients [169].
Furthermore, preclinical models of metastatic breast cancer revealed a significant survival advantage when RT is combined with immune checkpoint inhibitors [138,139,170]. In fact, a study with a metastatic breast cancer murine model (TS/A) conducted irradiation of one of the tumors with different RT regimes in combination with an anti-CTLA-4 AB. Fractionated RT in combination with the AB leads to significant regression of the secondary tumor, nonetheless, RT alone did not induce AbE in this model [139]. In another murine breast tumor model (4T1) study with bilateral tumors, the non-irradiated tumor was growth restricted by RT and combined with anti-PD-1 AB resulting in prolonged survival, in addition to a reduction in spontaneous lung metastases. These results suggest that an effective modification of the immune microenvironment of non-irradiated tumor is generated by means of combination therapy [170].
Krombach et al. employed TNBC cells to examine the initial steps of RT-induced anti-tumor priming which is crucial for AbE advent, observing the strongest effects with high single doses [171]. In addition, in another murine TNBC model, RT effect was enhanced due to AbEs by immune checkpoint inhibitors and hyperthermia (HT). After implanting 4T1-Luc cells into the mammary glands of BALB/c mice, the primary tumors of the animals received RT (24 Gy, three fractions), HT, anti-PD-1 and anti-CTLA-4 immunotherapy. Results revealed a moderate reduction in both the primary tumors and the lung metastasis but improvement with respect to survival was observed [172].

4.6. Prostate Cancer

Prostate cancer is the second most frequently diagnosed cancer with 7.3% of all estimated cases in males worldwide [147,173]. The most aggressive form of this cancer type is defined by disease progression despite androgen deprivation therapy, carrying a poor prognosis with a median survival of less than three years [174]. The so-called castration-resistant prostate cancer (CRPC) is associated with a variety of symptoms such as bone metastases, spinal cord compression and advanced pelvic disease. The bone metastases are often radiosensitive and treated with palliative RT for pain relief [175,176].
The therapeutic response improvement in prostate cancer of RT in combination with immunotherapy has also been tested in clinical trials. In a phase I/II study, patients with metastatic CRPC received ipilimumab (anti-CTLA-4) as a monotherapy or in combination with RT. Patients treated with 10 mg/kg ipilimumab and RT showed the strongest response, however, no AbE was observed [177]. Furthermore, similar results were observed in a multicenter phase III trial with 799 patients, where a combination of ipilimumab and RT was used to treat bone metastatic castration-resistant prostate cancer (8 Gy), unable to induce AbE [178]. In a DC vaccine clinical trial an AbE was detected in a patient with advanced CRPC. In addition to the prostate mass, the patient showed metastases in the lungs, bone marrow and mediastinal and inguinal LNs. Six months following several cycles of DC vaccine with an immunostimulant and irradiation of the prostatic tumor as well as the inguinal LNs (24 Gy, three fractions), the size of mediastinal and retroperitoneal lesions was significantly reduced. Prostate-specific antigen (PSA) serum concentration post RT was decreased whereas CD3+ and CD8+ T cells as well as IL-6 serum concentration were slightly enhanced. All other assessed parameters were within range [67].
In prostate cancer, the incidence of an AbE is reduced by a characteristic suppression of the anti-tumor immune response as a reaction to RT. The immune response can be hindered by the recruitment of myeloid cells with immunosuppressive function to irradiated TME. A study determined that after primary tumor site irradiation, expression of macrophage colony-stimulating factor 1 (CSF1) increased significantly both in patients and preclinical prostate models. CSF1 enhanced recruitment of immunosuppressive myeloid-derived suppressor cells (MDSCs), thereby limiting the therapeutic response to RT and suppressing the immune-related mechanism of AbE [179].
AbE prospect in prostate cancer has been demonstrated in the preclinical stage. In fact, a CRPC preclinical model observed an increased median overall survival and AbE with combination of RT and immune checkpoint inhibition. Mice’s leg tumors were irradiated with 20 Gy, and additionally treated with anti-PD-1 or anti-PD-L1 ABs. Thus, RT leads to a local response in the irradiated tumor followed by a similar regression of the non-irradiated tumor graft in this model. This response was mediated by CD8+ T cells [180].

5. Description of Hypothetical AbE Patient

Based on the reported cases, the profile of a patient who is most likely to develop an AbE was created as a summary of the current data situation.
The patient who is most likely to develop an AbE is a man aged between 42.4 and 72.6 years suffering from metastatic melanoma cancer. His primary tumor and also most likely this sentinel lymph nodes will be resected before the metastatic status is treated with systemic treatments (most probably ICB). Then, some of his metastases will be treated additionally with RT applied as a total dose of 30 Gy in 10 fractions. It will most likely be a brain metastasis that will be irradiated and the patient will receive at least four cycles of anti-PD-1 immunotherapy. This will most probably be 3 mg per kg body weight nivolumab every three weeks, and also the same dose of pembrolizumab. Around three months after this treatment, the irradiation of the brain metastases is most likely to result in the regression of non-irradiated lesions of the lungs followed by malignant LNs and liver lesions. However, a complete response could also be possible as a treatment result.
Previously reported cases of AbE were predominantly observed in male patients under immunotherapy whereas the numbers of case reports of female patients were nearly the same with or without ICB. The mean age mentioned in the case reports was 57.5 years with a standard deviation of 15.1 years. The number of reports indicates that the highest probability for an AbE is the strongest after combined RT and immunotherapy in melanoma (27 cases), followed by RCC (18 cases) and then adenocarcinomas of lung origin (12 cases). In the most reported AbE cases, the primary tumor was resected (31 cases), systemic chemotherapy (19 cases) is indicated as the second primary treatment followed by RT (seven cases) as the third treatment procedure. In most AbE cases, the RT is applied as palliative treatment and most patients received a total dose of 30 Gy applied in 10 fractions. In melanoma cases, more than half of the AbEs occurred after irradiation of brain metastasis. As the most commonly used ICB agent, nivolumab was applied in a total of 39 cases of AbE followed by 17 cases previously treated with pembrolizumab. With a spread of 3.1 months, an average of 3.2 months elapsed between the applied RT and the abscopal response. The combination of RT and ICB were correlated in 37 cases with the abscopal regression of lung metastases, in at least 17 cases with the regression of malignant LNs and in ten cases where the non-irradiated liver lesions responded. A total response was described in 11 patients who showed an abscopal response (Table 2).
With the available data, no specific size or localization of the primary tumor, metastases nor radiation field could be defined that would enable an AbE to be predicted. Most reported clinical cases do not provide detailed information regarding blood parameters or other molecular tumor markers.
Due to the relation between the appearance of an AbE and the anti-tumor immune reaction of the patient, the specific immunological pattern of patients would contribute significantly to the prediction of such a response. In particular, the change in the status of CD4+ and CD8+ T cells and the ratio of these cell groups to immunosuppressive Tregs or MDSCs during the entire treatment would be of particular interest. Unfortunately, only ten case reports in total provide details on this matter. When administering an immunotherapeutic agent, the determination of the PD-L1 status should be determined routinely. In 16 cases of AbE, this biomarker not only provided information for the benefit of an ICB but it also correlated with an abscopal response. In addition, the AbEs we have seen so far identify further markers that may potentially provide a pattern for predicting such an immunologically-driven effect.

6. Potential Predictors for Abscopal Effect

It remains unclear whether an abscopal response can be predicted. AbEs occur unexpectedly after the irradiated tumors begin to grow following RT, as some cases may have gone clinically undetected or discovered by chance during routine examinations [50]. Therefore, predictive biomarkers would promote a better monitoring and investigation of this phenomenon.
The T cell trafficking model was discussed as a predictive tool. In fact, Poleszczuk et al. developed a mathematical model to estimate the distribution of activated T cells between metastatic lesions. Thereby, they proposed that the migration of activated T cells from the activation site (irradiation field) to a specific metastasis depended on the physiologic blood flow to the organ involved. They aimed to identify treatment targets with the highest probability for AbE occurrence. They revealed that not all metastatic sites respond to the systemic immune response in an equal manner and that the dissemination of activated T cells among multiple metastatic sites is intricate. Consequently, the abscopal responses of the virtual patients presented the same metastatic sites, being highly patient-specific and could not be predicted from the data of previous cases [181]. Nevertheless, their model has some clinical limitations such as the gradual process of T cell priming and the influence of immune enhancement or suppression [182].
The current data situation only allows the application of a mathematical model to a small part of the mechanism behind an AbE, thus potentially misleading the clinical trial design. Therefore, timely identification of biomarkers associated with an abscopal response is necessary for predictive purposes. Even though some AbE biomarkers have already been discussed in literature no precise biomarkers have yet been validated to accurately predict AbE, especially in the context of combined treatment regimens. Thus, it is also worth considering immunotherapy predictive biomarkers for cancer immunotherapy and immune checkpoint inhibitors. Potential predictors for systemic responses to RT and immunotherapy include cytokine profiles as well as changes in AB titer and peripheral blood and/or tumor-infiltrating immune cells as listed in Table 4.

6.1. Tumor Protein p53

The tumor suppressor gene p53 regulates the expression of crucial proteins responsible for tumor cell proliferation, apoptosis, and DNA repair. The gene possesses a checkpoint function critical for preventing the replication of damaged DNA, thereby hindering thereby the growth of tumors, a function that is inhibited in mutated p53 [190]. Altered gene p53 functions are predominant driver mutations in numerous carcinomas.
Various preclinical studies have demonstrated p53 being involved in the mechanism behind AbE suggesting that p53 status could help predict the possibility of its occurrence.
Camphausen et al. examined the dependence of AbE on the function of p53 after RT. They irradiated the normal tissue of non-tumor-bearing legs of wild-type (wt) p53 mice and p53-null mice to induce abscopal response in distant lung carcinomas and fibrosarcomas. Only the tumors in mice with a non-inhibited p53 protein complex showed markedly reduced growth rate after irradiation. These results may suggest that intact p53 could be necessary to trigger the AbE tumor which is independent yet radiation-dose dependent. Its transcription function could be essential for the expression of cytokines or to affect a systemic antiangiogenic effect resulting in an abscopal signal [183]. Furthermore, another study proved that non-irradiated tumors respond systematically to irradiation only if they express functional p53. Implanted human colon cancer cells in wt-p53 and p53-null mice were irradiated (dose of 10 or 20 Gy) delivered to only one flank tumor leaving the contralateral side non-irradiated. The irradiated tumors reacted dose-dependently but presented a tumor retardation independent of p53 status. A significant growth inhibition of non-irradiated tumors was observed exclusively in wt-p53 animals, displaying a higher effect at higher doses [184]. This preclinical data may suggest that p53-dependent signals might be responsible for AbE and that the evaluation of p53-status could help predict the possibility of an AbE, even a verification in a clinical environment is still pending.

6.2. Calreticulin

Calreticulin, as a calcium-binding endoplasmic reticulum protein, plays an essential function in antigen presentation ensuring the appropriate loading of cellular antigens [191]. Thus, calreticulin expression may also give some predictive information about a potential abscopal response to immunotherapy and radiotherapy [192]. Calreticulin is essential for the recognition of tumor cells by DCs and subsequently by T cells, in fact, depletion of calreticulin in the tumor cells reduced the T cell killing [193]. Sublethal irradiated tumor cells increase their calreticulin expression while radiation induces the translocation of calreticulin from the endoplasmic reticulum to the cell surface. The exposure on the cell surface, in turn, affects the induction of immunogenic cell death and enhances antigen processing which promotes the uptake of irradiated tumor cells by APCs [115,194,195]. Gameiro et al. examined the calreticulin expression in vitro on human breast, lung and prostate carcinoma cells in comparison to untreated control cells. Cells exposed to sublethal doses increased their susceptibility to CD8+ CTL-mediated lysis. Moreover, exogenous calreticulin also increased the CTL lyses of non-irradiated tumor cells regardless of their p53 or triple-negative phenotype [115].

6.3. Hsp70

The highly stress-inducible heat shock protein 70 (Hsp70) is overexpressed inside tumor cells in many different cancer types, being an indicator for enhanced cell growth, protection against lethal damage, resistance to radiation-induced cell death or shorter overall survival [196,197]. Hsp70 is known as a useful biomarker for the prediction of hepatocellular carcinoma, prostate cancer, serum squamous cell carcinoma (SCC) of the esophagus, endometrial carcinoma or node-negative breast carcinoma [198,199,200,201]. Moreover, Hsp70 is actively released by viable tumor cells within exosomes. The plasma levels of the Hsp70 have been linked to tumor burden in a mice model correlating levels with tumor progression as well as radiation-induced regression of the tumors [185]. Following RT, Hsp70 is emitted into the extracellular matrix by dying tumor cells in a smaller amounts than by viable tumor cells [202]. Herein, Hsp70 acts as a danger signal stimulating the innate and adaptive immune system [203]. Natural killer cells (NKs) are activated after binding to free Hsp70, which then migrate to Hsp70 positive tumor cells to eradicate them [204]. Furthermore, the free Hsp70 can bind tumor-derived antigenic peptides that stimulate T cells via antigen cross-presentation on MHC molecules of APCs [205]. Thus, free Hsp70 seems to be a potential biomarker for a radiation-induced anti-tumor immune response and hence, for AbE.

6.4. γ-H2AX

Phosphorylated histone H2AX (γ-H2AX) molecule amount measured after irradiation is an essential biomarker utilized to determine the DNA damage induced by radiation but may also give some information about AbE. In response to irradiation-induced DNA DSBs, cells phosphorylate thousands of H2AX molecules specifically at the damaged sites [206]. The amount of these y-H2AX foci is proportional to the radiation dose and can also be visualized in the chromatin of human peripheral blood lymphocytes (PBL) and hair follicles for use in biodosimetry [186,206]. Siva et al. observed the γ-H2AX foci in non-irradiated PBL and hair follicles of irradiated NSCLC patients to determine the abscopal response of the irradiation in normal tissue. They also correlated this to changes in plasma levels of CCL22 and CCL3 cytokines [186].

6.5. PD-L1

PD-L1 expression is the sole biomarker typically used in clinical practice for the prediction of immunotherapy responses. PD-1 is an immune checkpoint receptor expressed on activated T cells that regulate the T cell response antagonistically. The interaction of PD-1 and its ligand PD-L1, which is expressed on tumor cells and other cells in the TME affects T cell activation, resulting in an immune escape for cancer cells. The expression of PD-L1 on tumor-infiltrating lymphocytes must also be considered as a factor influencing its predictive value. Herbst et al. related the response to checkpoint inhibitors with increased levels of PD-L1 on the surface of the tumor-infiltrating lymphocytes but not on T cells [207].
Numerous studies have shown a correlation between a high PD-L1 expression and the response to immunotherapy [208,209,210,211]. PD-L1 expression alone has limitations in successfully predicting responses to immunotherapy, as it may also indicate T cell exhaustion and thus reduced systemic efficacy. Thus, it may imply either a limitation or complete absence of AbE [132,212].
Nevertheless, PD-L1 expression may deliver important information for an AbE event but not as a comprehensive and independent clinical biomarker. Several AbE reports correlated the expression of PD-L1 with patients’ systemic response [88,97]. Yaguchi et al. reported a PD-L1 positive immunohistochemistry (IHC) analysis in an AbE of a patient with recurrent pulmonary pleomorphic carcinoma and rapidly progressive metastases in the brain, bone, and pulmonary pleurae. The resected lung tumor strongly expressed PD-L1 and the non-irradiated metastases responsed as nearly complete after three cycles of anti-PD-1 immunotherapy [88]. Watanabe et al. analyzed PD-L1 status in two melanoma patients. One patient presented PD-L1 positive tumor lesions with a 40% chance of complete response due to the anti-PD-1 treatment alone. Hence, the authors questioned the role of RT in the outcome. The AbE in the other patient was correlated with the RT due to lack of the PD-L1 expression [97].
PD-L1 expression, may also serve to define the role of the RT in an AbE, however, the temporal and spatial heterogeneity of PD-L1 expression must also be taken into account [213]. Nonetheless, PD-L1 negative patients may also benefit from checkpoint therapy because PD-L1 expression in metastases do not correspond with the expression in the primary tumor [73,82,90]. A study of metastatic NSCLC demonstrated a substantial variation in the PD-L1 expression across different sites and clinical states. PD-L1 expression levels in the lung or distant metastatic biopsies correlated with a higher response rate, whilst no association could be observed in LN metastases [214].

6.6. Tumor Markers

Tumor markers could be used to monitor the response of the tumor masses; therefore, longitudinal marker measurement also allows detection of an ongoing AbE.
Serum squamous cell carcinoma antigen (SCC Ag), a specific marker in SCC, could be associated with the outcome of RT. Elevated SCC Ag represents RT-resistant tumors whereas a reduction in its levels predicts a positive tumor response [215]. In the case of AbE, the SCC Ag levels also decrease due to irradiation and the following systemic response [31].
Similarly, the melanoma marker S100, commonly used as a marker for immunohistochemical identification of malignant melanoma, could help determine an abscopal response if measured longitudinally in short intervals. S100 levels were seen to correlate both with the tumor burden in distant metastases and the degree of infiltration of the liver and skeleton [216,217]. Immunotherapy combined with RT can lead to a decrease in S100 which could be associated with an AbE [97].
CEA is expressed as a non-specific tumor marker on the surface of many cancer cells, whereas CEA plasma levels are very faint (0 to 5 ng/mL). Increased CEA levels act as a determiner for the tumor stage and as a predictor of the response to therapy for gastric cancer [218,219,220]. In NSCLC, a CEA plasma level could be used as a prognostic marker for overall survival, recurrence or progression-free survival [221]. Additionally, a group analyzed CEA levels in a patient with metastatic NSCLC and reported an AbE following treatment, consistent with a drastic drop to normal levels in CEA levels during the systemic response [52]. Kuroda et al. also observed a significant drop in the plasma levels of a lung adenocarcinoma patient after RT and during the time of the occurrence of the AbE. CEA levels increased in accordance with the progression of a non-responder metastasis [43].
In breast cancer, high preoperative CA15-3 and CEA serum levels were described as directly related to tumor burden and outcome predictors [222,223]. In fact, Leung et al. reported a substantial decline CA15-3 levels in a breast cancer patient, along with a continuous decrease in CEA during the occurrence of the abscopal response [39].
Carbohydrate antigen 19-9 (CA19-9) is most commonly used as a tumor marker for pancreatic ductal adenocarcinoma [224], during an abscopal response this marker has seen to be reduced. Bonilla et al. reported a corresponding plasma concentration curve for a patient with gastric adenocarcinoma presenting an AbE following palliative RT [41]. Most recently, a case report of a patient with endometrioid adenocarcinoma reported decreasing plasma levels of CA125 during the time of the AbE event [48].
Unfortunately, most patients with AbE do not reflect the disease progress through specific tumor marker signatures [60,98].

6.7. ctDNA Amount and Tumor Mutational Burden

Circulating tumor DNA (ctDNA) are found in cell-free blood fraction and are described as a potent biomarker that enables the detection of tumor-specific sequence alterations [225,226,227]. Tissue tumor mutational burden (TMB) is a viable biomarker for the response to immunotherapy, helping to quantify tumor immunogenicity [228]. Elevated TMB supports the enhanced expression of tumor-associated neo-antigens following RT and subsequent anti-tumor immune response [229,230].
ctDNA abundance, the peripheral blood ratio between ctDNA and total circulating DNA, might also give information about tumor burden or disease progression. An independent predictor of the response to immunotherapy has been described as having a TMB higher than 16 when measured in ctDNA abundance [231]. ctDNA levels seem to be correlated to pseudo-progression in KRAS-mutated adenocarcinoma, a mutation associated with an improved outcome under immunotherapy. In pseudo-progressive patients, ctDNA levels decrease drastically whereas in progressive patients they appears strongly elevated [232].
Xie et al. measured the ratio between ctDNA and the total free DNA in the peripheral blood of an RCC patient which presented an AbE both before and during RT and pembrolizumab treatment. ctDNA abundance decreased before it unexpectedly rose two months after combination treatment and then drastically decreased again, suggesting its correlation with the extinction of tumor cells as corroborated by CT images [63]. Another study reported an AbE in a neuroendocrine cervical carcinoma patient with metastatic lesions in the liver, pelvic and retroperitoneal LNs. After performing a ctDNA test (70 genes), they found alterations in 19 genes indicating an elevated TMB. Tissue next-generation sequencing (NGS) also revealed a high TMB (53 mutations per megabase) [61].
Zhao et al. presented a case of an abscopal response in a patient with esophageal carcinoma and multiple LN metastases. Esophagectomy was performed followed by several cycles of chemotherapy, pembrolizumab and RT (42 Gy, six fractions) targeting the left retroperitoneal LN. After two months all LN metastases displayed complete regression. The ctDNA was analyzed to monitor the TP53 mutation. Results suggested that these mutations decreased significantly due to the applied combination therapy [72].
The changes in ctDNA release during or immediately after RT are a result of irradiation-induced immunogenic cell death. ctDNA presents a half-life of 0.5–2 h, therefore blood samples should be taken periodically and at shorter intervals to obtain more accurate information on the ctDNA alterations during patient treatment [233].

6.8. Tumor Antigens and Antibodies

RT generates tumor tissue damage leading to enhanced tumor-specific antigen release, which could function as an in situ immunization and induce AbE [107]. Consequently, serum analysis of tumor antigens alterations may be used as an abscopal response marker for RT alone and/or in combination with immunotherapy.
The antigen load or more precisely the load of neoantigens, which are the number of mutations actually targeted by T cells, is associated with the response to immunotherapy [234,235]. Indeed, a study correlated a significantly increased neoantigen load in tumors of NSCLC patients after PD-1 blockade with a higher clinical response and improved progression-free survival. Thereby, the neoantigen-induced T cell response was detected after therapy initiation with its highest level three weeks following treatment starting with a progressive reduction [236]. AbE have been described to occur at different time points, suggesting that abscopal immune reactions might stimulate the priming of new T cells in addition to the initially primed one, originating a broader and more specific T cell response [182].
Measuring the concentration of specific antibodies against tumor-associated antigens may be illustrative. In effect, the serum of a melanoma patient was tested for auto-AB levels against melanoma antigen A3 (MAGEA3), revealing a systemic antitumor immune response. Following RT and immunotherapy the patient responded to cancer antigen PAS domain-containing 1 (PASD1) achieving complete remission [53]. Another research group reported an AbE of a melanoma patient treated with RT and ipilimumab. The cancer cells of the patient expressed the cancer antigen NY-ESO-1, an antigen found in 30–40% of advanced melanoma patients. The titers of AB against epitopes within the central portion of NY-ESO-1 in the serum samples collected during the treatment correlated positively with the disease progression. In fact, after RT and immunotherapy, the titers were increased by a factor of more than 30 corresponding to disease resolution [49].
Antigen immunogenicity, the ability of released antigens to elicit an anti-tumor immune response, is fundamental to the occurrence of abscopal responses and thus may be the limiting factor for an AbE [237]. Therefore, a further study promoted the determination of the tumor immunogenicity score (TIGS), represented as tumor antigenicity multiplied by antigen processing and presenting status, as an effective biomarker for immunotherapy response prediction [238]. TIGS may be best determined during the treatment period, on account of the relation between AbE and the immune response to tumor antigens.

6.9. Absolute Lymphocyte Count and Amount of Blood Immune Cells

The absolute lymphocyte count (ALC) and/or the count of specific immune cells could also act as conceivable predictive biomarkers for AbE. However, to date, only a few cases have given further details on the immune cell status of the respective patients during treatment.
In 1977, Antoniades et al. counted the absolute leukocytes as well as the subpopulations of neutrophils, monocytes, eosinophils, and basophils in their patients’ blood before and after irradiation compared with values of a healthy donor. The two patients with metastatic lymphoma exhibited a marked reduction in the size of non-treated abdominal LNs. Both patients showed an irradiation-induced decrease in the ALC corresponding with an increased ratio of neutrophils [9]. A more recent study analyzed leukocytes, neutrophils, lymphocytes, and monocytes in the blood of a patient with a diffuse-type giant cell tumor showing an AbE at one month after RT treatment of the right hilar in the left lung [28].
Diverse studies have shown that if more of these standard values were reported, changes in the immune cell amounts may be correlated with the occurrence of an AbE [43,49,52,54,187,188]. More precise information of the immune condition also enables an assessment of the immunosuppressive status that counteracts a possible AbE.
Golden et al. reported a case of AbE in a treatment-resistant lung cancer patient treated with RT and ipilimumab. Two to three months following RT to hepatic metastases, the non-irradiated lesions showed a decrease in size. Disease progression was tracked by ALCs, white blood cells (WBCs) and absolute eosinophil counts (AECs). Other studies associated an ALC increase of ≥ 1000/μL in whole blood with improved survival rates [239,240]. Indeed, the patient showed a correspondingly elevated ALC due to the RT and ipilimumab treatment combined with an AbE. The AEC was also enhanced by more than 100/μL whole blood, an increase which was previously also correlated with prolonged survival. Tumor-infiltrating lymphocytes (cytotoxic T cell, cytotoxic granules and regulatory T cells) also increased due to the treatment [52]. In a retrospective analysis of 21 advanced melanoma patients, an AbE was reported in 11 patients treated with a combination of ipilimumab and RT. In patients presenting AbE, ALC was increased prior to RT [54]. Another retrospective study analyzed 153 cases of patients with various cancer types treated with a combination of RT and immunotherapy, where at least one non-contiguous lesion was not irradiated. The results suggested a correlation between an enhanced post-RT ALC and abscopal response. Moreover, a lower post-RT APC results in a poorer progression-free and overall survival [188]. In brief, these data suggest ALC as a potential predictive AbE marker, however, this hypothesis requires additional data from clinical trials.
Due to their prominent role in AbE, different case reports also reported CD8+ lymphocytes values in blood at different treatment time points. There are reports of low CD8+ lymphocyte counts at the time of AbE, which increased again with disease progression [43]. A study measured the amounts of patients CD14+ MDSCs, CD8+ and CD4+ T cells as well as their activation status during the treatment period via flow cytometry. CD4+ ICOShigh T cells decreased during RT response while MDSCs first decreased severely before increasing again within the abscopal response period. The frequencies of IFN-γ-producing CD8+ and CD4+ T cells were decreased due to cell irradiation, yet increased slightly during AbE [49].
Additionally, the neutrophil-to-lymphocyte ratio may be an effective AbE predictive factor. In fact, eleven patients with different cancer entities presented an AbE with a combined RT and granulocyte-macrophage colony-stimulating factor (GM-CSF) treatment. A comparison was made between the baseline features of abscopal responders and non-responders. Accordingly, no significant differences were observed in phenotype parameters such as age, sex, previous numbers of RT or chemotherapy regimens nor in laboratory values (hemoglobin, albumin, WBC count) between responders and non-responders. However, a significantly lower baseline was noted in non-responders for the neutrophil-to-lymphocyte ratio [189].

6.10. Tumor-Infiltrating Lymphocytes

The evaluation of the tumor-infiltrating lymphocytes (TILs) may also give a potential indication of abscopal response. In preclinical studies, enhanced amounts of CD8+ cells in non-irradiated tumors were associated with the enhanced probabilities of AbE [151]. Clinically, the effectiveness of immunotherapy could be predicted as a result of TIL count determination [241].
Joe et al. analyzed the tissue of squamous carcinoma of the anal canal tumor with a complete response four months after palliative RT to the pelvic metastasis. Immunohistochemical staining for CD163, CD3, CD4, and CD8 showed heterogeneity of cell distribution. Some tumor regions were densely infiltrated with lymphocytes, including CD8+ and CD4+ T cells, suggesting a profuse immune response [31].
Moreover, a recent case report of a metastatic RCC patient reported a high heterogeneous infiltration of the non-irradiated primary lesion by CD8+ T cells. The patient showed regression of a lung metastasis six months after RT and immunotherapy treatment. Nevertheless, only a part of the tumor was considered immunogenic before anti-PD-1 treatment [90]. Teulings et al. performed a detailed immunological analysis of biopsies from a melanoma patient presenting AbE twelve months after irradiation of the axillary region and brain. Axillary LN sections exhibited a lymphocyte infiltration, whereas brain metastasis was mainly infiltrated by CD8+ T cells. Melanocyte-specific CD8+ T cells were found to represent an effector memory subgroup in both skin biopsies and blood. These data suggested a systemic and local anti-melanoma response against melanocyte differentiation antigens from the primary tumor and metastases [25]. Furthermore, the immunohistochemical analysis of the non-irradiated retropharyngeal metastasis of a patient with malignant pleural mesothelioma revealed a moderate infiltration of CD3+ cells after the occurrence of AbE. The intra-tumoral T cell population was composed of around 25% CD8+ T cells [99].

6.11. Cytokine Profiles

Radiation, as part of the cellular repair response, induces the release of cytokines and chemokines which in turn play various roles in systemic immune response modulation. Longitudinal observation of cytokine profiles could provide an overview about the systemic immune response. This information could provide some valuable therapy success predictive data in addition to assessing AbE incidence.
Animal models have already shown that increased levels of specific cytokines have a significant role in cytotoxic T cell responses and AbE [139]. In prostate cancer, several blood-based biomarker studies correlated elevated levels of different cytokines with disease progression and poor prognosis. After exposure to RT, the plasma levels of diverse cytokines, namely IL-1α, IL-4, IL-6, TNF-α, TGF-β, IFN-ɣ and macrophage colony stimulating factor (M-CSF) were increased and correlated with RT-induced toxicity [242,243,244,245]. In breast cancer studies, IL-6 and IL-8 were considered as potential markers for metastasis and disease progression [246,247,248]. Moreover, in patients with NSCLC undergoing palliative thoracic RT, significant changes in CCL15, CCL23, CCL24, CXCL2, CXCL6, CX3CL1, IL-6, and IL-8 serum levels were observed before, during, and after RT, correlating with metabolic tumor burden [249]. In hepatocellular carcinoma patients treated with RT, levels of IL-6 and IL-10 were seen to be valuable predictors of infield- and outfield-intrahepatic treatment failure [250]. Ohba et al. measured the serum concentrations of IL-1β, IL-2, IL-4, IL-6, TNF-α, and hepatocyte growth factor (HGF) in series before and after RT in a patient with hepatocellular carcinoma who presented AbE ten months after the irradiation of a thoracic vertebral metastasis. Most cytokine levels did not significantly change, except for TNF-α serum levels which had increased. The authors associated this increase with an enhanced NK cell activity along with increased AbE occurrence [15].
Immunosuppressive cytokines reduce the probability of AbE occurrence. Consequently, the balance between immune promoting and suppressing cytokines systemically and within the tumor environment before and after irradiation is crucial for treatment response. Levels of the cytokines TGF-β, IL-10 and adenosine should be particularly monitored. Radiation-induced cell death leads to the activation of M2 macrophages and TGF-β and IL-10 production within the irradiation field [251]. Moreover, Tregs are enriched in the irradiated tumor, releasing TGF-β and IL-10 [252]. In fact, increased Treg amounts together with radiation-induced recruitment of MDSCs exerts an impact on the production of immunosuppressive adenosine by irradiation stressed or injured cells [253]. On the other hand, IL-8, which primarily comes from circulating and intra-tumoral myeloid cells, inhibits the adaptive immune response by blocking the antitumor activity of effector T cells as well as antigen presentation [254,255]. Studies examining the clinical benefits of immune-checkpoint inhibitors linked high baseline levels of IL-8 in patients plasma to the poor prognosis of checkpoint inhibitor therapy. Consequently, a reduced immune response as a result of the IL-8 expression additionally suppresses the occurrence of an AbE.

6.12. Exosomes

Extracellular vesicles, including microvesicles and exosome, are small vesicles that contain bioactive cargo such as proteins, DNA or RNA, mediating intercellular communication in both physiological and pathological settings [256]. Exosomes secreted from tumor cells can carry both immune-stimulatory and immune-suppressive factors that partially mimic the profile of the releasing tumoral cells. Most previous studies have highlighted the immune-suppressive functions of these tumor-derived exosomes (TEXs). However, TEXs also transport immune-stimulatory TSAs as well as (co-)stimulatory immune molecules [257]. They play a crucial role in RT-associated immunity due to the antigen presentation and immune regulation dependent on the immune status of the TME [256]. TEXs have been studied to serve as liquid biopsy markers with high clinical diagnostic and prognostic value in lung, pancreatic, gastrointestinal cancer and hepatocellular carcinoma [258]. TEXs not only affect the local microenvironment around their releasing site but also circulate in the blood to distant metastatic sites promoting/suppressing tumor growth [257]. The immune-promoting effect is mainly based on the delivery of exosome-derived TSA to dendritic cells and on the activation of cytotoxic T cells to generate a specific anti-tumor response [259].
Following the irradiation production of TEXs containing DAMPs, chemokines and TSA are enhanced, however they vary according to the originally irradiated tissue [257,260,261]. The TEXs are processed by antigen-presenting cells, and trigger the activation of cytotoxic T cells which then migrate to distant non-targeted tumor cells and can induce AbE [257]. In case of AbE, Hsp70 surface-positive tumor-derived exosomes may be enriched in the irradiated field, facilitating their uptake by APC and thus triggering an enhanced anti-tumor response by NK cells [262,263].
In metastatic melanoma, TEXs could be used to predict the tumor response to anti-PD-L1 checkpoint inhibition. In this case, the level of circulating TEXs with surface PD-L1 could correlate with IFN-γ levels, which could distinguish responders from non-responders [264]. Additionally, exosomes may potentially be used to identify NSCLS patients who respond positively to immune therapy [265].

7. Clinical Studies on Abscopal Effect

Even after almost a decade of radioimmunotherapy and numerous studies on different immunotherapeutic agents, the occurrence of abscopal immune reactions has not been regularly achieved in clinical trials. The most frequently reported cases of AbE were correlated with the co-administration of RT and ICB, in particular with the administration of the anti-PD-1 therapy nivolumab. Nevertheless, a similar effectiveness has not yet been observed in clinical trials. Accordingly, trial design and evaluation must be performed taking into consideration many different aspects, namely, patient and endpoint selection, RT parameters, the particular definition of AbE, therapy sequence, among others, making the discovery of the optimal conditions extremely challenging [266].
A clinical trial tested the combination of RT with GM-CSF in patients with metastatic solid tumors of different origin (NCT02474186). The primary endpoint of the study was the proportion of patients with an abscopal response. Eleven of the fourty-one patients enrolled (26.8%) presented abscopal responses (four patients with metastatic NSCLC, 5, breast cancer patients and two, thymic cancer patients) [189]. A phase I study treated 20 stage IV melanoma patients with a combination of RT, nivolumab (anti-PD-1), and ipilimumab (anti-CTLA4), receiving immunotherapy shortly before and after palliative RT. Patients who received a conventional irradiation dose (3 Gy, 10 fractions) experienced in a 50% ratio an abscopal reaction extrinsic of the irradiation field whereas 11.1% of patients treated with hypofractionated irradiated scheme (9 Gy, three fractions) responded in a similar manner [267].
Contrary results were reported by a phase II trial examining the combination of RT and immune therapy in 20 patients with inoperable or metastatic melanoma (NCT02821182). The overall response rate (ORR) of the non-irradiated lesions increased by up to 45% with three complete and six partial responses similar to nivolumab monotherapy, however, no AbE was detected [268]. Subsequently, another study failed to observe an AbE in patients with metastatic head and neck squamous cell carcinoma (HNSCC) in a randomized, phase II trial of nivolumab combined with RT (NCT02684253). RT was delivered to one safely irradiated metastatic lesion between the first and second dose of nivolumab in three fractions adding up to a dose of 27 Gy. The patients in this treatment arm showed no significant prolonged survival compared to the immune monotherapy arm [269].
Current ongoing trials with AbE as their primary or secondary outcome mainly investigate the combination of RT and the inhibition of the PD-1/PD-L1 pathway in various cancers such as NSCLC, metastatic gastro-intestinal cancer, metastatic melanoma and metastatic breast cancer (Table 5). Several of them depict promising outcomes which may provide further insights into potential AbE predictive biomarkers.
In the ongoing clinical trial NCT03385226, among other objectives, the combination of RT (12 Gy, three fractions) and pembrolizumab is being observed over a period of 2 years for AbE rate. Immune cells will be functionally analyzed along with the diversity of T cell clones and tumor-infiltrating lymphocyte-specific neo-antigens and the expression of immunological checkpoints [270]. Moreover, another clinical trial aims to evaluate the treatment response at non-irradiated and extra-cranial sites of melanoma or NSCLC patients with brain metastasis, after receiving pembrolizumab along with stereotactic radiosurgery (6 Gy, 9 Gy, 18–21 Gy) (NCT02858869). As a secondary objective, potential immune biomarkers will be compared, analyzing blood samples for absolute cell counts, major lymphocyte populations as well as other serum immune biomarkers pre-, during and post-treatment for up to three years [271]. Another phase II study investigates the effect of pembrolizumab and palliative radiation therapy in patients with a metastatic esophagus, stomach or with gastroesophageal junction cancer (NCT02830594). Its primary endpoint aims to determine changes in tumor-infiltrating and circulating cytotoxic T cells, immunosuppressive Tregs and MDSCs at the metastatic examined sites. Cell populations will be correlated with the response rates of irradiated and non-irradiated tumors [272]. The benefit of radio-immunotherapy for advanced metastatic patients is being tested in a trial with a stratified patient cohort (NCT03453892). The systemic immune-modulating effect of the treatment will be assessed by deep immunophenotyping, evaluating up to 30 immune cell subtypes with their activation markers at various time points of the treatment [273]. Similar targets are being investigated in another trial (NCT03042156), which researches the effect of immune therapy and palliative RT combined treatment, along with inflammatory and irradiation-induced signatures as indicators for an AbE, particularly those extrinsic to irradiated sections [274].

8. Conclusions and Recommendations in the Frame-Work of 3P Medicine

AbE is a rare clinical phenomenon associated with RT, which, however, is becoming more frequent now towards improved radiation delivery and the development of immunotherapeutic drugs. The addition of ICB to conventional RT has led to an increase in the incidence of abscopal responses due to the involvement of various immune pathways in their occurrence. Nevertheless, the exact mechanism responsible for an AbE is still in question. It is observed after different treatment regimens and it takes weeks to months before an AbE becomes noticeable which makes it difficult to predict under typical clinical conditions.
The wide variation of the applied treatments often does not allow for a precise identification of the AbE’s origin, its triggers and contributors. Systemic therapies namely chemotherapy as well as immunotherapy may have delayed effects that later cannot be assigned to a specific treatment. To this end, the immune system of the patients can be affected by typical cancer pain management or immunosuppressive drugs such as corticosteroids. Furthermore, normal tissue in the radiation field also receives a small but still significant radiation dose during RT. Therefore, the observed AbE could also be related to low-dose radiation effects which are a result of the applied irradiation.
The costs of a combined therapy of ICB and RT has been estimated by Giuliani et al. to be 5866 EUR more expensive than immunotherapy alone, translates to 707 € to 1086 € per month of overall survival of NSCLC patients [275]. Targeted induction or an accurate prediction of an AbE by biomarkers could lead to a personalized, more precise and more cost-effective treatment strategy.
The reported clinical cases allow for the identification of several promising biomarker candidates which need to be further analyzed in the context of their predictive power. Validated biomarkers might be of great clinical utility
-
to stratify patients
-
to prognose treatment response individually by applying associated diagnostics
-
to optimize treatment strategies tailored to every individual
-
to create advanced multi-level diagnostic approaches based on the application of artificial intelligence (AI) for big-data analysis in the context of 3P medicine improving cost-efficacy of treatments and individual outcomes [276,277,278,279,280].

Author Contributions

Conceptualization, B.L. and O.G.; validation, B.L. and A.T.C.; investigation; B.L.; writing—original draft preparation, B.L., A.T.C., M.H., F.A.G. and O.G., writing—review and editing, B.L. and O.G.; supervision, O.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data have been included within the article. Therefore, no additional data file is required.

Conflicts of Interest

There is no conflict of interest with respect to this manuscript.

References

  1. Formenti, S.C.; Demaria, S. Systemic effects of local radiotherapy. Lancet Oncol. 2009, 10, 718–726. [Google Scholar] [CrossRef] [Green Version]
  2. Andrews, J.R. The Radiobiology of Human Cancer Radiotherapy, 2nd ed.; University Park Press: Baltimore, MD, USA, 1978; ISBN 0-8391-1161-4. [Google Scholar]
  3. Mole, R.H. Whole body irradiation; radiobiology or medicine? Br. J. Radiol. 1953, 26, 234–241. [Google Scholar] [CrossRef]
  4. Marín, A.; Martín, M.; Liñán, O.; Alvarenga, F.; López, M.; Fernández, L.; Büchser, D.; Cerezo, L. Bystander effects and radiotherapy. Rep. Pract. Oncol. Radiother. 2014, 20, 12–21. [Google Scholar] [CrossRef] [Green Version]
  5. Abuodeh, Y.; Venkat, P.; Kim, S. Systematic review of case reports on the abscopal effect. Curr. Probl. Cancer 2016, 40, 25–37. [Google Scholar] [CrossRef]
  6. Dagoglu, N.; Karaman, S.; Caglar, H.B.; Oral, E.N. Abscopal Effect of Radiotherapy in the Immunotherapy Era: Systematic Review of Reported Cases. Cureus 2019, 11, e4103. [Google Scholar] [CrossRef] [Green Version]
  7. Lome, L.G.; Navani, S.; Aral, I.M. Spontaneous regression of pulmonary metastases from transitional cell carcinoma of the bladder. Cancer 1970, 26, 415–418. [Google Scholar] [CrossRef]
  8. Ehlers, G.; Fridman, M. Abscopal effect of radiation in papillary adenocarcinoma. Br. J. Radiol. 1973, 46, 220–222. [Google Scholar] [CrossRef] [PubMed]
  9. 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]
  10. Fairlamb, D.J. Spontaneous regression of metastases of renal cancer: A report of two cases including the first recorded regression following irradiation of a dominant metastasis and review of the world literature. Cancer 1981, 47, 2102–2106. [Google Scholar] [CrossRef]
  11. Rees, G.J.G. Abscopal regression in lymphoma: A mechanism in common with total body irradiation? Clin. Radiol. 1981, 32, 475–480. [Google Scholar] [CrossRef]
  12. Robin, H.I.; AuBuchon, J.; Varanasi, V.R.; Weinstein, A.B. The abscopal effect: Demonstration in lymphomatous involvement of kidneys. Med. Pediatr. Oncol. 1981, 9, 473–476. [Google Scholar] [CrossRef] [PubMed]
  13. Rees, G.J.; Ross, C.M. Abscopal regression following radiotherapy for adenocarcinoma. Br. J. Radiol. 1983, 56, 63–66. [Google Scholar] [CrossRef] [PubMed]
  14. MacManus, M.P.; Harte, R.J.; Stranex, S. Spontaneous regression of metastatic renal cell carcinoma following palliative irradiation of the primary tumour. Ir. J. Med. Sci. 1994, 163, 461–463. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Nam, S.W.; Han, J.-Y.; Kim, J.I.; Park, S.H.; Cho, S.H.; Han, N.I.; Yang, J.M.; Kim, J.K.; Choi, S.W.; Lee, Y.S.; et al. Spontaneous regression of a large hepatocellular carcinoma with skull metastasis. J. Gastroenterol. Hepatol. 2005, 20, 488–492. [Google Scholar] [CrossRef] [PubMed]
  17. 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]
  18. 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]
  19. Isobe, Y.; Aritaka, N.; Sasaki, M.; Oshimi, K.; Sugimoto, K. Spontaneous regression of natural killer cell lymphoma. J. Clin. Pathol. 2009, 62, 647–650. [Google Scholar] [CrossRef]
  20. 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]
  21. 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]
  22. 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]
  23. Tubin, S. 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]
  24. 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]
  25. Teulings, H.E.; Tjin, E.P.M.; Willemsen, K.J.; Krebbers, G.; van Noesel, C.J.; Kemp, E.H.; Nieuweboer-Krobotova, L.; van der Veen, J.P.W.; Luiten, R.M. Radiation-induced melanoma-associated leucoderma, systemic antimelanoma immunity and disease-free survival in a patient with advanced-stage melanoma: A case report and immunological analysis. Br. J. Dermatol. 2013, 168, 733–738. [Google Scholar] [CrossRef]
  26. Lock, M.; Muinuddin, A.; Kocha, W.I.; Dinniwell, R.; Rodrigues, G.; D’souza, D. Abscopal Effects: Case Report and Emerging Opportunities. Cureus 2015, 7, e344. [Google Scholar] [CrossRef] [Green Version]
  27. Yarchoan, M.; Lim, M.; Brahmer, J.R.; Ettinger, D. Oligometastatic Adenocarcinoma of the Lung: A Therapeutic Opportunity for Long-Term Survival. Cureus 2015, 7, e409. [Google Scholar] [CrossRef] [Green Version]
  28. Desar, I.M.E.; Braam, P.M.; Kaal, S.E.J.; Gerritsen, W.R.; Oyen, W.J.G.; van der Graaf, W.T.A. Abscopal effect of radiotherapy in a patient with metastatic diffuse-type giant cell tumor. Acta Oncol. 2016, 55, 1510–1512. [Google Scholar] [CrossRef]
  29. Orton, A.; Wright, J.; Buchmann, L.; Randall, L.; Hitchcock, Y.J. A Case of Complete Abscopal Response in High-Grade Pleiomorphic Sarcoma Treated with Radiotherapy Alone. Cureus 2016, 8, e821. [Google Scholar] [CrossRef] [Green Version]
  30. Saba, R.; Saleem, N.; Peace, D. Long-term survival consequent on the abscopal effect in a patient with multiple myeloma. BMJ Case Rep. 2016, 2016, bcr2016215237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. 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]
  32. Lesueur, P.; Chevalier, F.; Stefan, D.; Habrand, J.-L.; Lerouge, D.; Gervais, R. Review of the mechanisms involved in the abscopal effect and future directions with a focus on thymic carcinoma. Tumori 2017, 103, 217–222. [Google Scholar] [CrossRef]
  33. Azami, A.; Suzuki, N.; Azami, Y.; Seto, I.; Sato, A.; Takano, Y.; Abe, T.; Teranishi, Y.; Tachibana, K.; Ohtake, T. Abscopal effect following radiation monotherapy in breast cancer: A case report. Mol. Clin. Oncol. 2018, 9, 283–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. 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]
  35. 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]
  36. Chino, F.; Pollis, K.E.; Choi, S.; Salama, J.K.; Palta, M. Stereotactic Body Radiation Therapy-Induced Abscopal Effect on Hepatocellular Carcinoma After Treatment for Lung Cancer: A Case Report. Hepatology 2018, 68, 1653–1655. [Google Scholar] [CrossRef]
  37. Chuang, C.-H.; Hsu, J.-F.; Shen, Y.-T.; Yang, C.-J. Regression of a metastatic lung mass after receiving whole brain irradiation: Can the abscopal effect cross the blood-brain barrier? Asia Pac. J. Clin. Oncol. 2018, 14, e548–e550. [Google Scholar] [CrossRef]
  38. Hamilton, A.J.; Seid, J.; Verdecchia, K.; Chuba, P. Abscopal Effect after Radiosurgery for Solitary Brain Metastasis from Non-small Cell Lung Cancer. Cureus 2018, 10, e3777. [Google Scholar] [CrossRef] [Green Version]
  39. Leung, H.W.; Wang, S.-Y.; Jin-Jhih, H.; Chan, A.L. Abscopal effect of radiation on bone metastases of breast cancer: A case report. Cancer Biol. Ther. 2018, 19, 20–24. [Google Scholar] [CrossRef]
  40. Agyeman, M.B.; Vanderpuye, V.D.; Yarney, J. Abscopal Effect of Radiotherapy in Imatinib-resistant Dermatofibrosarcoma Protuberans. Cureus 2019, 11, e3857. [Google Scholar] [CrossRef] [Green Version]
  41. 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] [PubMed] [Green Version]
  42. 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]
  43. Kuroda, A.; Tabuchi, T.; Iwami, E.; Sasahara, K.; Matsuzaki, T.; Nakajima, T.; Tsutsumi, Y.; Eguchi, K.; Terashima, T. Abscopal effect of radiation on multiple lung metastases of lung adenocarcinoma: A case report. BMC Cancer 2019, 19, 336. [Google Scholar] [CrossRef]
  44. Ellerin, B.E.; Demandante, C.G.N.; 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] [PubMed]
  45. Guan, S.; Wang, H.; Qi, X.-H.; Guo, Q.; Zhang, H.-Y.; Liu, H.; Zhu, B.-J. Abscopal effect of local irradiation treatment for thymoma: A case report. Am. J. Transl. Res. 2020, 12, 2234–2240. [Google Scholar]
  46. Ohmatsu, K.; Hashimoto, Y.; Kawanishi, M.; Ishii, Y.; Kono, S.; Kuribayashi, S.; Ariizumi, S.; Karasawa, K. Abscopal complete regression of hepatocellular carcinoma with multiple pleural metastases. Int. Cancer Conf. J. 2021, 10, 54–58. [Google Scholar] [CrossRef]
  47. Mazzaschi, G.; Tommasi, C.; Pietri, E.; Corcione, L.; de Giorgi, A.; Bini, P.; Bui, S. Abscopal effect as part of treatment of oligometastatic head and neck cancer: A case report. Clin. Case Rep. 2021, 9, 1334–1338. [Google Scholar] [CrossRef]
  48. Tomita, N.; Ogawa, S.; Aikawa, G. Abscopal Effect of Pelvic Intensity Modulated Radiation Therapy on Lung Metastases in a Patient with Recurrent Endometrial Cancer. Adv. Radiat. Oncol. 2021, 6, 100563. [Google Scholar] [CrossRef]
  49. Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Adjepong, D.; Malik, B.H. Radiation Therapy as a Modality to Create Abscopal Effects: Current and Future Practices. Cureus 2020, 12, e7054. [Google Scholar] [CrossRef] [Green Version]
  51. Hiniker, S.M.; Chen, D.S.; Reddy, S.; Chang, D.T.; Jones, J.C.; Mollick, J.A.; Swetter, S.M.; Knox, S.J. A systemic complete response of metastatic melanoma to local radiation and immunotherapy. Transl. Oncol. 2012, 5, 404–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. 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]
  53. 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]
  54. Grimaldi, A.M.; Simeone, E.; Giannarelli, D.; Muto, P.; Falivene, S.; Borzillo, V.; Giugliano, F.M.; Sandomenico, F.; Petrillo, A.; Curvietto, M.; et al. Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 2014, 3, e28780. [Google Scholar] [CrossRef]
  55. Thallinger, C.; Prager, G.; Ringl, H.; Zielinski, C. Abscopal-Effekt in der Therapie des malignen Melanoms. Hautarzt 2015, 66, 545–548. [Google Scholar] [CrossRef]
  56. Okwan-Duodu, D.; Pollack, B.P.; Lawson, D.; Khan, M.K. Role of radiation therapy as immune activator in the era of modern immunotherapy for metastatic malignant melanoma. Am. J. Clin. Oncol. 2015, 38, 119–125. [Google Scholar] [CrossRef]
  57. Michot, J.-M.; Mazeron, R.; Dercle, L.; Ammari, S.; Canova, C.; Marabelle, A.; Rose, S.; Rubin, E.; Deutsch, E.; Soria, J.-C.; et al. Abscopal effect in a Hodgkin lymphoma patient treated by an anti-programmed death 1 antibody. Eur. J. Cancer 2016, 66, 91–94. [Google Scholar] [CrossRef]
  58. Cong, Y.; Shen, G.; Wu, S.; Hao, R. Abscopal regression following SABR for non-small-cell-lung cancer: A case report. Cancer Biol. Ther. 2017, 18, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Komatsu, T.; Nakamura, K.; Kawase, A. Abscopal Effect of Nivolumab in a Patient with Primary Lung Cancer. J. Thorac. Oncol. 2017, 12, e143–e144. [Google Scholar] [CrossRef] [Green Version]
  60. Sato, H.; Suzuki, Y.; Yoshimoto, Y.; Noda, S.-E.; Murata, K.; Takakusagi, Y.; Okazaki, A.; Sekihara, T.; Nakano, T. An abscopal effect in a case of concomitant treatment of locally and peritoneally recurrent gastric cancer using adoptive T-cell immunotherapy and radiotherapy. Clin. Case Rep. 2017, 5, 380–384. [Google Scholar] [CrossRef]
  61. Sharabi, A.; Kim, S.S.; Kato, S.; Sanders, P.D.; Patel, S.P.; Sanghvi, P.; Weihe, E.; Kurzrock, R. Exceptional Response to Nivolumab and Stereotactic Body Radiation Therapy (SBRT) in Neuroendocrine Cervical Carcinoma with High Tumor Mutational Burden: Management Considerations from the Center For Personalized Cancer Therapy at UC San Diego Moores Cancer Center. Oncologist 2017, 22, 631–637. [Google Scholar] [CrossRef] [Green Version]
  62. Shi, F.; Wang, X.; Teng, F.; Kong, L.; Yu, J. Abscopal effect of metastatic pancreatic cancer after local radiotherapy and granulocyte-macrophage colony-stimulating factor therapy. Cancer Biol. Ther. 2017, 18, 137–141. [Google Scholar] [CrossRef] [Green Version]
  63. Xie, G.; Di, G.; Zhang, L.; Chen, S.; Wu, D. A rapid and systemic complete response to stereotactic body radiation therapy and pembrolizumab in a patient with metastatic renal cell carcinoma. Cancer Biol. Ther. 2017, 18, 547–551. [Google Scholar] [CrossRef] [PubMed]
  64. Britschgi, C.; Riesterer, O.; Burger, I.A.; Guckenberger, M.; Curioni-Fontecedro, A. Report of an abscopal effect induced by stereotactic body radiotherapy and nivolumab in a patient with metastatic non-small cell lung cancer. Radiat. Oncol. 2018, 13, 102. [Google Scholar] [CrossRef]
  65. Gutkin, P.M.; Hiniker, S.M.; Swetter, S.M.; Reddy, S.A.; Knox, S.J. Complete Response of Metastatic Melanoma to Local Radiation and Immunotherapy: 6.5 Year Follow-Up. Cureus 2018, 10, e3723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. 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. 2019, 8, 29–32. [Google Scholar] [CrossRef] [PubMed]
  67. Rodríguez-Ruiz, M.E.; Perez-Gracia, J.L.; Rodríguez, I.; Alfaro, C.; Oñate, C.; Pérez, G.; Gil-Bazo, I.; Benito, A.; Inogés, S.; López-Diaz de Cerio, A.; et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann. Oncol. 2018, 29, 1312–1319. [Google Scholar] [CrossRef] [PubMed]
  68. 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] [PubMed] [Green Version]
  69. 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]
  70. Wight, J.C.; Hawkes, E.A.; Berlangieri, S.U.; Khor, R.; Grigg, A.P. An abscopal effect may augment PD-1 inhibition in refractory classical Hodgkin lymphoma. Leuk. Lymphoma 2018, 59, 2749–2751. [Google Scholar] [CrossRef]
  71. 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]
  72. Zhao, X.; Kang, J.; Zhao, R. Abscopal effect of radiation on lymph node metastasis in esophageal carcinoma: A case report and literature review. Oncol. Lett. 2018, 16, 3555–3560. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Bitran, J. The Abscopal Effect Exists in Non-small Cell Lung Cancer: A Case Report and Review of the Literature. Cureus 2019, 11, e4118. [Google Scholar] [CrossRef] [Green Version]
  75. Choi, J.S.; Sansoni, E.R.; Lovin, B.D.; Lindquist, N.R.; Phan, J.; Mayo, L.L.; Ferrarotto, R.; Su, S.Y. Abscopal Effect Following Immunotherapy and Combined Stereotactic Body Radiation Therapy in Recurrent Metastatic Head and Neck Squamous Cell Carcinoma: A Report of Two Cases and Literature Review. Ann. Otol. Rhinol. Laryngol. 2020, 129, 517–522. [Google Scholar] [CrossRef]
  76. 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]
  77. Garelli, E.; Rittmeyer, A.; Putora, P.M.; Glatzer, M.; Dressel, R.; Andreas, S. Abscopal effect in lung cancer: Three case reports and a concise review. Immunotherapy 2019, 11, 1445–1461. [Google Scholar] [CrossRef]
  78. Gounder, M.M.; Zhu, G.; Roshal, L.; Lis, E.; Daigle, S.R.; Blakemore, S.J.; Michaud, N.R.; Hameed, M.; Hollmann, T.J. Immunologic Correlates of the Abscopal Effect in a SMARCB1/INI1-negative Poorly Differentiated Chordoma after EZH2 Inhibition and Radiotherapy. Clin. Cancer Res. 2019, 25, 2064–2071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Grimaux, X.; Delva, R.; Jadaud, E.; Croue, A. Nivolumab-induced bullous pemphigoid after radiotherapy and abscopal effect. Australas. J. Dermatol. 2019, 60, e235–e236. [Google Scholar] [CrossRef]
  80. Ishiyama, Y.; Takagi, T.; Yoshida, K.; Iizuka, J.; Kakuta, Y.; Okumi, M.; Ishida, H.; Tanabe, K. Possible abscopal effect in urothelial carcinoma of the upper urinary tract after treatment with immune checkpoint inhibitors. IJU Case Rep. 2020, 3, 25–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Lin, X.; Lu, T.; Xie, Z.; Qin, Y.; Liu, M.; Xie, X.; Li, S.; Zhou, C. Extracranial abscopal effect induced by combining immunotherapy with brain radiotherapy in a patient with lung adenocarcinoma: A case report and literature review. Thorac. Cancer 2019, 10, 1272–1275. [Google Scholar] [CrossRef]
  82. Liu, X.; Yao, J.; Song, L.; Zhang, S.; Huang, T.; Li, Y. Local and abscopal responses in advanced intrahepatic cholangiocarcinoma with low TMB, MSS, pMMR and negative PD-L1 expression following combined therapy of SBRT with PD-1 blockade. J. Immunother. Cancer 2019, 7, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. 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]
  84. Qin, Q.; Nan, X.; Miller, T.; Fisher, R.; Teh, B.; Pandita, S.; Farach, A.M.; Pingali, S.R.; Pandita, R.K.; Butler, E.B.; et al. Complete Local and Abscopal Responses from a Combination of Radiation and Nivolumab in Refractory Hodgkin’s Lymphoma. Radiat. Res. 2018, 190, 322–329. [Google Scholar] [CrossRef]
  85. 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]
  86. Suzuki, G.; Masui, K.; Yamazaki, H.; Takenaka, T.; Asai, S.; Taniguchi, H.; Nakamura, T.; Ukimura, O.; Yamada, K. Abscopal effect of high-dose-rate brachytherapy on pelvic bone metastases from renal cell carcinoma: A case report. J. Contemp. Brachytherapy 2019, 11, 458–461. [Google Scholar] [CrossRef]
  87. Trommer, M.; Yeo, S.Y.; Persigehl, T.; Bunck, A.; Grüll, H.; Schlaak, M.; Theurich, S.; von Bergwelt-Baildon, M.; Morgenthaler, J.; Herter, J.M.; et al. Abscopal Effects in Radio-Immunotherapy-Response Analysis of Metastatic Cancer Patients with Progressive Disease under Anti-PD-1 Immune Checkpoint Inhibition. Front. Pharmacol. 2019, 10, 511. [Google Scholar] [CrossRef] [Green Version]
  88. 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]
  89. Forner, D.; Horwich, P.; Trites, J.R.; Hollenhorst, H.; Bullock, M.; Lamond, N.W.D. The abscopal effect in head-and-neck squamous cell carcinoma treated with radiotherapy and nivolumab: A case report and literature review. Curr. Oncol. 2020, 27, 330–335. [Google Scholar] [CrossRef] [PubMed]
  90. 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] [Green Version]
  91. Igarashi, H.; Fukuda, M.; Konno, Y.; Takano, H. Abscopal effect of radiation therapy after nivolumab monotherapy in a patient with oral mucosal melanoma: A case report. Oral Oncol. 2020, 108, 104919. [Google Scholar] [CrossRef]
  92. Kuhara, Y.; Ninomiya, M.; Hirahara, S.; Doi, H.; Kenji, S.; Toyota, K.; Yano, R.; Kobayashi, H.; Hashimoto, Y.; Yokoyama, Y.; et al. A long-term survival case of unresectable gastric cancer with multidisciplinary therapy including immunotherapy and abscopal effect. Int. Cancer Conf. J. 2020, 9, 193–198. [Google Scholar] [CrossRef]
  93. Levitin, M.; Ofori, J.; Shin, W.J.; Huang, J.; Daly, M.; Cao, D.; Pachynski, R. Radiation and Checkpoint Inhibitor Immunotherapy Lead to Long Term Disease Control in a Metastatic RCC patient with Brain Metastases. Front. Oncol. 2020, 10, 566070. [Google Scholar] [CrossRef]
  94. Nakajima, N.; Kano, T.; Oda, K.; Uchida, T.; Otaki, T.; Nagao, K.; Shimizu, Y.; Kawakami, M.; Kim, H.; Nitta, M.; et al. Possible abscopal effect after discontinuation of nivolumab in metastatic renal cell carcinoma. IJU Case Rep. 2020, 3, 215–218. [Google Scholar] [CrossRef] [PubMed]
  95. 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] [PubMed]
  96. Wang, W.; Huang, C.; Wu, S.; Liu, Z.; Liu, L.; Li, L.; Li, S. Abscopal effect induced by modulated radiation therapy and pembrolizumab in a patient with pancreatic metastatic lung squamous cell carcinoma. Thorac. Cancer 2020, 11, 2014–2017. [Google Scholar] [CrossRef] [PubMed]
  97. Watanabe, T.; Firat, E.; Scholber, J.; Gaedicke, S.; Heinrich, C.; Luo, R.; Ehrat, N.; Multhoff, G.; Schmitt-Graeff, A.; Grosu, A.-L.; et al. Deep abscopal response to radiotherapy and anti-PD-1 in an oligometastatic melanoma patient with unfavorable pretreatment immune signature. Cancer Immunol. Immunother. 2020, 69, 1823–1832. [Google Scholar] [CrossRef]
  98. Hotta, T.; Okuno, T.; Nakao, M.; Amano, Y.; Isobe, T.; Tsubata, Y. Reproducible abscopal effect in a patient with lung cancer who underwent whole-brain irradiation and atezolizumab administration. Thorac. Cancer 2021, 12, 985–988. [Google Scholar] [CrossRef]
  99. Mampuya, W.A.; Bouchaab, H.; Schaefer, N.; Kinj, R.; La Rosa, S.; Letovanec, I.; Ozsahin, M.; Bourhis, J.; Coukos, G.; Peters, S.; et al. Abscopal effect in a patient with malignant pleural mesothelioma treated with palliative radiotherapy and pembrolizumab. Clin. Transl. Radiat. Oncol. 2021, 27, 85–88. [Google Scholar] [CrossRef] [PubMed]
  100. Wong, A.; Vellayappan, B.; Cheng, L.; Zhao, J.J.; Muthu, V.; Asokumaran, Y.; Low, J.-L.; Lee, M.; Huang, Y.-Q.; Kumarakulasinghe, N.B.; et al. Atypical Response Patterns in Renal Cell Carcinoma Treated with Immune Checkpoint Inhibitors-Navigating the Radiologic Potpourri. Cancers 2021, 13, 1689. [Google Scholar] [CrossRef]
  101. Azzam, E.I.; Jay-Gerin, J.-P.; Pain, D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012, 327, 48–60. [Google Scholar] [CrossRef] [Green Version]
  102. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef]
  103. Garibaldi, C.; Jereczek-Fossa, B.A.; Marvaso, G.; Dicuonzo, S.; Rojas, D.P.; Cattani, F.; Starzyńska, A.; Ciardo, D.; Surgo, A.; Leonardi, M.C.; et al. Recent advances in radiation oncology. Ecancermedicalscience 2017, 11, 785. [Google Scholar] [CrossRef] [Green Version]
  104. Golden, E.B.; Apetoh, L. Radiotherapy and immunogenic cell death. Semin. Radiat. Oncol. 2015, 25, 11–17. [Google Scholar] [CrossRef] [Green Version]
  105. 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, 11, e4100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Demaria, S.; Ng, B.; Devitt, M.L.; Babb, J.S.; Kawashima, N.; Liebes, L.; Formenti, S.C. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 862–870. [Google Scholar] [CrossRef]
  107. Formenti, S.C.; Demaria, S. Radiation therapy to convert the tumor into an in situ vaccine. Int. J. Radiat. Oncol. Biol. Phys. 2012, 84, 879–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. 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]
  109. Grass, G.D.; Krishna, N.; Kim, S. The immune mechanisms of abscopal effect in radiation therapy. Curr. Probl. Cancer 2016, 40, 10–24. [Google Scholar] [CrossRef]
  110. Gupta, A.; Probst, H.C.; van Vuong Landshammer, A.; Muth, S.; Yagita, H.; Schwendener, R.; Pruschy, M.; Knuth, A.; van den Broek, M. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J. Immunol. 2012, 189, 558–566. [Google Scholar] [CrossRef] [Green Version]
  111. Kaur, P.; Asea, A. Radiation-induced effects and the immune system in cancer. Front. Oncol. 2012, 2, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Rodriguez-Ruiz, M.E.; Rodriguez, I.; Leaman, O.; López-Campos, F.; Montero, A.; Conde, A.J.; Aristu, J.J.; Lara, P.; Calvo, F.M.; Melero, I. Immune mechanisms mediating abscopal effects in radioimmunotherapy. Pharmacol. Ther. 2019, 196, 195–203. [Google Scholar] [CrossRef]
  113. Lanneau, D.; de Thonel, A.; Maurel, S.; Didelot, C.; Garrido, C. Apoptosis versus cell differentiation: Role of heat shock proteins HSP90, HSP70 and HSP27. Prion 2007, 1, 53–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Chen, T.; Cao, X. Stress for maintaining memory: HSP70 as a mobile messenger for innate and adaptive immunity. Eur. J. Immunol. 2010, 40, 1541–1544. [Google Scholar] [CrossRef] [PubMed]
  115. 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]
  116. Di Virgilio, F. Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol. Sci. 2007, 28, 465–472. [Google Scholar] [CrossRef] [PubMed]
  117. Martinon, F.; Mayor, A.; Tschopp, J. The inflammasomes: Guardians of the body. Annu. Rev. Immunol. 2009, 27, 229–265. [Google Scholar] [CrossRef] [Green Version]
  118. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
  119. Sato, H.; Okonogi, N.; Nakano, T. Rationale of combination of anti-PD-1/PD-L1 antibody therapy and radiotherapy for cancer treatment. Int. J. Clin. Oncol. 2020, 25, 801–809. [Google Scholar] [CrossRef] [Green Version]
  120. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef] [Green Version]
  121. Ablasser, A.; Goldeck, M.; Cavlar, T.; Deimling, T.; Witte, G.; Röhl, I.; Hopfner, K.-P.; Ludwig, J.; Hornung, V. cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 2013, 498, 380–384. [Google Scholar] [CrossRef] [Green Version]
  122. Ishikawa, H.; Ma, Z.; Barber, G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef] [Green Version]
  123. Guo, Y.-R.; Cao, Q.-D.; Hong, Z.-S.; Tan, Y.-Y.; Chen, S.-D.; Jin, H.-J.; Tan, K.-S.; Wang, D.-Y.; Yan, Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak—An update on the status. Mil. Med. Res. 2020, 7, 11. [Google Scholar] [CrossRef] [Green Version]
  124. 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] [PubMed] [Green Version]
  125. Burnette, B.C.; Liang, H.; Lee, Y.; Chlewicki, L.; Khodarev, N.N.; Weichselbaum, R.R.; Fu, Y.-X.; Auh, S.L. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011, 71, 2488–2496. [Google Scholar] [CrossRef] [Green Version]
  126. 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]
  127. 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] [PubMed]
  128. 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]
  129. Matsumura, S.; Demaria, S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat. Res. 2010, 173, 418–425. [Google Scholar] [CrossRef] [Green Version]
  130. Lim, J.Y.H.; 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]
  131. Hallahan, D.E.; Spriggs, D.R.; Beckett, M.A.; Kufe, D.W.; Weichselbaum, R.R. Increased tumor necrosis factor alpha mRNA after cellular exposure to ionizing radiation. Proc. Natl. Acad. Sci. USA 1989, 86, 10104–10107. [Google Scholar] [CrossRef] [Green Version]
  132. 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]
  133. Ben-Shoshan, J.; Maysel-Auslender, S.; Mor, A.; Keren, G.; George, J. Hypoxia controls CD4+CD25+ regulatory T-cell homeostasis via hypoxia-inducible factor-1alpha. Eur. J. Immunol. 2008, 38, 2412–2418. [Google Scholar] [CrossRef] [PubMed]
  134. Yan, M.; Jene, N.; Byrne, D.; Millar, E.K.A.; O’Toole, S.A.; McNeil, C.M.; Bates, G.J.; Harris, A.L.; Banham, A.H.; Sutherland, R.L.; et al. Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers. Breast Cancer Res. 2011, 13, R47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Sethumadhavan, S.; Silva, M.; Philbrook, P.; Nguyen, T.; Hatfield, S.M.; Ohta, A.; Sitkovsky, M.V. Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells. PLoS ONE 2017, 12, e0187314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Chen, M.; Qiao, G.; Hylander, B.L.; Mohammadpour, H.; Wang, X.-Y.; Subjeck, J.R.; Singh, A.K.; Repasky, E.A. Adrenergic stress constrains the development of anti-tumor immunity and abscopal responses following local radiation. Nat. Commun. 2020, 11, 1821. [Google Scholar] [CrossRef]
  137. Donlon, N.E.; Power, R.; Hayes, C.; Reynolds, J.V.; Lysaght, J. Radiotherapy, immunotherapy, and the tumour microenvironment: Turning an immunosuppressive milieu into a therapeutic opportunity. Cancer Lett. 2021, 502, 84–96. [Google Scholar] [CrossRef]
  138. Demaria, S.; Kawashima, N.; Yang, A.M.; Devitt, M.L.; Babb, J.S.; Allison, J.P.; Formenti, S.C. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 2005, 11, 728–734. [Google Scholar]
  139. Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef] [Green Version]
  140. Rodriguez-Ruiz, M.E.; Rodriguez, I.; Barbes, B.; Mayorga, L.; Sanchez-Paulete, A.R.; Ponz-Sarvise, M.; Pérez-Gracia, J.L.; Melero, I. Brachytherapy attains abscopal effects when combined with immunostimulatory monoclonal antibodies. Brachytherapy 2017, 16, 1246–1251. [Google Scholar] [CrossRef]
  141. Rekers, N.H.; Olivo Pimentel, V.; Yaromina, A.; Lieuwes, N.G.; Biemans, R.; Zegers, C.M.L.; Germeraad, W.T.V.; van Limbergen, E.J.; Neri, D.; Dubois, L.J.; et al. The immunocytokine L19-IL2: An interplay between radiotherapy and long-lasting systemic anti-tumour immune responses. Oncoimmunology 2018, 7, e1414119. [Google Scholar] [CrossRef] [Green Version]
  142. Yasmin-Karim, S.; Bruck, P.T.; Moreau, M.; Kunjachan, S.; Chen, G.Z.; Kumar, R.; Grabow, S.; Dougan, S.K.; Ngwa, W. Radiation and Local Anti-CD40 Generate an Effective in situ Vaccine in Preclinical Models of Pancreatic Cancer. Front. Immunol. 2018, 9, 2030. [Google Scholar] [CrossRef] [PubMed]
  143. Kwon, Y.-S.; Lee, M.-G.; Baek, J.; Nam, K.-S.; Im Lee, J.; Kim, S.; Jang, H. Evaluation of Radiotherapy-Induced Systemic Antitumor Effects in Mice Bearing 4T1 Mouse Breast Cancer Cells. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef] [PubMed]
  144. Herrscher, H.; Sauer, B.; Truntzer, P.; Robert, C. Abscopal antitumor effect in a patient with melanoma and coronavirus disease 2019. Eur. J. Cancer 2021, 149, 91–93. [Google Scholar] [CrossRef] [PubMed]
  145. Kucerova, P.; Cervinkova, M. Spontaneous regression of tumour and the role of microbial infection—Possibilities for cancer treatment. Anticancer Drugs 2016, 27, 269–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Thomas, J.A.; Badini, M. The role of innate immunity in spontaneous regression of cancer. Indian J. Cancer 2011, 48, 246–251. [Google Scholar] [CrossRef]
  147. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  148. Mullangi, S.; Lekkala, M.R. Adenocarcinoma; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  149. Varadhachary, G.R. Carcinoma of unknown primary origin. Gastrointest. Cancer Res. 2007, 1, 229–235. [Google Scholar]
  150. Dicken, B.J.; Bigam, D.L.; Cass, C.; Mackey, J.R.; Joy, A.A.; Hamilton, S.M. Gastric adenocarcinoma: Review and considerations for future directions. Ann. Surg. 2005, 241, 27–39. [Google Scholar] [CrossRef]
  151. 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]
  152. Shaheen, O.; Ghibour, A.; Alsaid, B. Esophageal Cancer Metastases to Unexpected Sites: A Systematic Review. Gastroenterol. Res. Pract. 2017, 2017, 1657310. [Google Scholar] [CrossRef] [Green Version]
  153. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. Non-small cell lung cancer: Epidemiology, risk factors, treatment, and survivorship. Mayo Clin. Proc. 2008, 83, 584–594. [Google Scholar] [CrossRef]
  154. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Putora, P.M.; Fruh, M.; Kern, L. The place of radiotherapy in the palliative management of NSCLC. Breathe 2011, 8, 134–143. [Google Scholar] [CrossRef]
  156. Lindberg, K.; Nyman, J.; Riesenfeld Källskog, V.; Hoyer, M.; Lund, J.Å.; Lax, I.; Wersäll, P.; Karlsson, K.; Friesland, S.; Lewensohn, R. Long-term results of a prospective phase II trial of medically inoperable stage I NSCLC treated with SBRT—The Nordic experience. Acta Oncol. 2015, 54, 1096–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Postmus, P.E.; Kerr, K.M.; Oudkerk, M.; Senan, S.; Waller, D.A.; Vansteenkiste, J.; Escriu, C.; Peters, S. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv1–iv21. [Google Scholar] [CrossRef]
  158. Fiorica, F.; Tebano, U.; Gabbani, M.; Perrone, M.; Missiroli, S.; Berretta, M.; Giuliani, J.; Bonetti, A.; Remo, A.; Pigozzi, E.; et al. Beyond Abscopal Effect: A Meta-Analysis of Immune Checkpoint Inhibitors and Radiotherapy in Advanced Non-Small Cell Lung Cancer. Cancers 2021, 13, 2352. [Google Scholar] [CrossRef] [PubMed]
  159. Escudier, B.; Porta, C.; Schmidinger, M.; Rioux-Leclercq, N.; Bex, A.; Khoo, V.; Grunwald, V.; Gillessen, S.; Horwich, A. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 706–720. [Google Scholar] [CrossRef] [Green Version]
  160. Ljungberg, B.; Albiges, L.; Bedke, J.; Bex, A.; Capitanio, U.; Giles, R.H.; Hora, M.; Klatte, T.; Lam, T.; Marconi, L.; et al. European Association of Urology Guidelines on Renal Cell Carcinoma Presented at the EAU Annual Congress Milan 2021; EAU Guidelines Office: Arnhem, The Netherlands, 2021; ISBN 978-94-92671-13-4. [Google Scholar]
  161. Sullivan, R.J.; Atkins, M.B.; Kirkwood, J.M.; Agarwala, S.S.; Clark, J.I.; Ernstoff, M.S.; Fecher, L.; Gajewski, T.F.; Gastman, B.; Lawson, D.H.; et al. An update on the Society for Immunotherapy of Cancer consensus statement on tumor immunotherapy for the treatment of cutaneous melanoma: Version 2.0. J. Immunother. Cancer 2018, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  162. Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
  163. Robert, C.; Ribas, A.; Schachter, J.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.M.; Lotem, M.; et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): Post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019, 20, 1239–1251. [Google Scholar] [CrossRef]
  164. Park, S.S.; Dong, H.; Liu, X.; Harrington, S.M.; Krco, C.J.; Grams, M.P.; Mansfield, A.S.; Furutani, K.M.; Olivier, K.R.; Kwon, E.D. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol. Res. 2015, 3, 610–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. 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 2019, 8, e1507669. [Google Scholar] [CrossRef] [Green Version]
  166. Hiniker, S.M.; Reddy, S.A.; Maecker, H.T.; Subrahmanyam, P.B.; Rosenberg-Hasson, Y.; Swetter, S.M.; Saha, S.; Shura, L.; Knox, S.J. A Prospective Clinical Trial Combining Radiation Therapy with Systemic Immunotherapy in Metastatic Melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 578–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Theurich, S.; Rothschild, S.I.; Hoffmann, M.; Fabri, M.; Sommer, A.; Garcia-Marquez, M.; Thelen, M.; Schill, C.; Merki, R.; Schmid, T.; et al. Local Tumor Treatment in Combination with Systemic Ipilimumab Immunotherapy Prolongs Overall Survival in Patients with Advanced Malignant Melanoma. Cancer Immunol. Res. 2016, 4, 744–754. [Google Scholar] [CrossRef] [Green Version]
  168. Formenti, S.C.; Lee, P.; Adams, S.; Goldberg, J.D.; Li, X.; Xie, M.W.; Ratikan, J.A.; Felix, C.; Hwang, L.; Faull, K.F.; et al. Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2493–2504. [Google Scholar] [CrossRef] [Green Version]
  169. Zhang, L.; Tang, R.; Deng, J.-P.; Zhang, W.-W.; Lin, H.-X.; Wu, S.-G.; He, Z.-Y. The effect of postmastectomy radiotherapy in node-positive triple-negative breast cancer. BMC Cancer 2020, 20, 1146. [Google Scholar] [CrossRef]
  170. Rodriguez-Ruiz, M.E.; Rodriguez, I.; Garasa, S.; Barbes, B.; Solorzano, J.L.; Perez-Gracia, J.L.; Labiano, S.; Sanmamed, M.F.; Azpilikueta, A.; Bolaños, E.; et al. Abscopal Effects of Radiotherapy Are Enhanced by Combined Immunostimulatory mAbs and Are Dependent on CD8 T Cells and Crosspriming. Cancer Res. 2016, 76, 5994–6005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Krombach, J.; Hennel, R.; Brix, N.; Orth, M.; Schoetz, U.; Ernst, A.; Schuster, J.; Zuchtriegel, G.; Reichel, C.A.; Bierschenk, S.; et al. Priming anti-tumor immunity by radiotherapy: Dying tumor cell-derived DAMPs trigger endothelial cell activation and recruitment of myeloid cells. Oncoimmunology 2019, 8, e1523097. [Google Scholar] [CrossRef] [Green Version]
  172. Oei, A.L.; Korangath, P.; Mulka, K.; Helenius, M.; Coulter, J.B.; Stewart, J.; Velarde, E.; Crezee, J.; Simons, B.; Stalpers, L.J.A.; et al. Enhancing the abscopal effect of radiation and immune checkpoint inhibitor therapies with magnetic nanoparticle hyperthermia in a model of metastatic breast cancer. Int. J. Hyperth. 2019, 36, 47–63. [Google Scholar] [CrossRef]
  173. Ferlay, J.; Ervik MLam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today. Available online: https://gco.iarc.fr/today (accessed on 4 February 2021).
  174. Kirby, M.; Hirst, C.; Crawford, E.D. Characterising the castration-resistant prostate cancer population: A systematic review. Int. J. Clin. Pract. 2011, 65, 1180–1192. [Google Scholar] [CrossRef]
  175. Hotte, S.J.; Saad, F. Current management of castrate-resistant prostate cancer. Curr. Oncol. 2010, 17 (Suppl. 2), S72–S79. [Google Scholar] [CrossRef] [PubMed]
  176. Rose, J.N.; Crook, J.M. The role of radiation therapy in the treatment of metastatic castrate-resistant prostate cancer. Ther. Adv. Urol. 2015, 7, 135–145. [Google Scholar] [CrossRef] [Green Version]
  177. Slovin, S.F.; Higano, C.S.; Hamid, O.; Tejwani, S.; Harzstark, A.; Alumkal, J.J.; Scher, H.I.; Chin, K.; Gagnier, P.; McHenry, M.B.; et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: Results from an open-label, multicenter phase I/II study. Ann. Oncol. 2013, 24, 1813–1821. [Google Scholar] [CrossRef] [PubMed]
  178. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; van den Eertwegh, A.J.M.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef] [Green Version]
  179. 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]
  180. 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] [Green Version]
  181. Poleszczuk, J.T.; Luddy, K.A.; Prokopiou, S.; Robertson-Tessi, M.; Moros, E.G.; Fishman, M.; Djeu, J.Y.; Finkelstein, S.E.; Enderling, H. Abscopal Benefits of Localized Radiotherapy Depend on Activated T-cell Trafficking and Distribution between Metastatic Lesions. Cancer Res. 2016, 76, 1009–1018. [Google Scholar] [CrossRef] [Green Version]
  182. Demaria, S.; Formenti, S.C. Can abscopal effects of local radiotherapy be predicted by modeling T cell trafficking? J. Immunother. Cancer 2016, 4, 29. [Google Scholar] [CrossRef] [Green Version]
  183. 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]
  184. 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]
  185. Bayer, C.; Liebhardt, M.E.; Schmid, T.E.; Trajkovic-Arsic, M.; Hube, K.; Specht, H.M.; Schilling, D.; Gehrmann, M.; Stangl, S.; Siveke, J.T.; et al. Validation of heat shock protein 70 as a tumor-specific biomarker for monitoring the outcome of radiation therapy in tumor mouse models. Int. J. Radiat. Oncol. Biol. Phys. 2014, 88, 694–700. [Google Scholar] [CrossRef]
  186. Siva, S.; Lobachevsky, P.; MacManus, M.P.; Kron, T.; Möller, A.; Lobb, R.J.; Ventura, J.; Best, N.; Smith, J.; Ball, D.; et al. Radiotherapy for Non-Small Cell Lung Cancer Induces DNA Damage Response in Both Irradiated and Out-of-field Normal Tissues. Clin. Cancer Res. 2016, 22, 4817–4826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Blattner, C.; Fleming, V.; Weber, R.; Himmelhan, B.; Altevogt, P.; Gebhardt, C.; Schulze, T.J.; Razon, H.; Hawila, E.; Wildbaum, G.; et al. CCR5+ Myeloid-Derived Suppressor Cells Are Enriched and Activated in Melanoma Lesions. Cancer Res. 2018, 78, 157–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Chen, D.; Verma, V.; Patel, R.R.; Barsoumian, H.B.; Cortez, M.A.; Welsh, J.W. Absolute Lymphocyte Count Predicts Abscopal Responses and Outcomes in Patients Receiving Combined Immunotherapy and Radiation Therapy: Analysis of 3 Phase 1/2 Trials. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 196–203. [Google Scholar] [CrossRef]
  189. Golden, E.B.; Chhabra, A.; Chachoua, A.; Adams, S.; Donach, M.; Fenton-Kerimian, M.; Friedman, K.; Ponzo, F.; Babb, J.S.; Goldberg, J.; et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: A proof-of-principle trial. Lancet Oncol. 2015, 16, 795–803. [Google Scholar] [CrossRef]
  190. Vousden, K.H.; Lane, D.P. p53 in health and disease. Nat. Rev. Mol. Cell Biol. 2007, 8, 275–283. [Google Scholar] [CrossRef]
  191. Fucikova, J.; Spisek, R.; Kroemer, G.; Galluzzi, L. Calreticulin and cancer. Cell Res. 2021, 31, 5–16. [Google Scholar] [CrossRef]
  192. Ngwa, W.; Irabor, O.C.; Schoenfeld, J.D.; Hesser, J.; Demaria, S.; Formenti, S.C. Using immunotherapy to boost the abscopal effect. Nat. Rev. Cancer 2018, 18, 313–322. [Google Scholar] [CrossRef] [PubMed]
  193. Kroemer, G.; Zitvogel, L. Subversion of calreticulin exposure as a strategy of immune escape. Cancer Cell 2021, 39, 449–451. [Google Scholar] [CrossRef]
  194. 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]
  195. Obeid, M.; Tesniere, A.; Panaretakis, T.; Tufi, R.; Joza, N.; van Endert, P.; Ghiringhelli, F.; Apetoh, L.; Chaput, N.; Flament, C.; et al. Ecto-calreticulin in immunogenic chemotherapy. Immunol. Rev. 2007, 220, 22–34. [Google Scholar] [CrossRef] [PubMed]
  196. Gehrmann, M.; Specht, H.M.; Bayer, C.; Brandstetter, M.; Chizzali, B.; Duma, M.; Breuninger, S.; Hube, K.; Lehnerer, S.; van Phi, V.; et al. Hsp70—A biomarker for tumor detection and monitoring of outcome of radiation therapy in patients with squamous cell carcinoma of the head and neck. Radiat. Oncol. 2014, 9, 131. [Google Scholar] [CrossRef] [Green Version]
  197. Juhasz, K.; Lipp, A.-M.; Nimmervoll, B.; Sonnleitner, A.; Hesse, J.; Haselgruebler, T.; Balogi, Z. The complex function of hsp70 in metastatic cancer. Cancers 2013, 6, 42–66. [Google Scholar] [CrossRef] [Green Version]
  198. Abe, M.; Manola, J.B.; Oh, W.K.; Parslow, D.L.; George, D.J.; Austin, C.L.; Kantoff, P.W. Plasma levels of heat shock protein 70 in patients with prostate cancer: A potential biomarker for prostate cancer. Clin. Prostate Cancer 2004, 3, 49–53. [Google Scholar] [CrossRef] [PubMed]
  199. Fujita, Y.; Nakanishi, T.; Miyamoto, Y.; Hiramatsu, M.; Mabuchi, H.; Miyamoto, A.; Shimizu, A.; Takubo, T.; Tanigawa, N. Proteomics-based identification of autoantibody against heat shock protein 70 as a diagnostic marker in esophageal squamous cell carcinoma. Cancer Lett. 2008, 263, 280–290. [Google Scholar] [CrossRef] [PubMed]
  200. Chuma, M.; Sakamoto, M.; Yamazaki, K.; Ohta, T.; Ohki, M.; Asaka, M.; Hirohashi, S. Expression profiling in multistage hepatocarcinogenesis: Identification of HSP70 as a molecular marker of early hepatocellular carcinoma. Hepatology 2003, 37, 198–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Davidson, B.; Valborg Reinertsen, K.; Trinh, D.; Reed, W.; Bøhler, P.J. BAG-1/SODD, HSP70, and HSP90 are potential prognostic markers of poor survival in node-negative breast carcinoma. Hum. Pathol. 2016, 54, 64–73. [Google Scholar] [CrossRef] [PubMed]
  202. Multhoff, G.; Pockley, A.G.; Schmid, T.E.; Schilling, D. The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett. 2015, 368, 179–184. [Google Scholar] [CrossRef] [Green Version]
  203. Vega, V.L.; Rodríguez-Silva, M.; Frey, T.; Gehrmann, M.; Diaz, J.C.; Steinem, C.; Multhoff, G.; Arispe, N.; de Maio, A. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J. Immunol. 2008, 180, 4299–4307. [Google Scholar] [CrossRef] [Green Version]
  204. Multhoff, G. Activation of natural killer cells by heat shock protein 70. Int. J. Hyperth. 2002, 18, 576–585. [Google Scholar] [CrossRef]
  205. Srivastava, P.K.; Menoret, A.; Basu, S.; Binder, R.J.; McQuade, K.L. Heat shock proteins come of age: Primitive functions acquire new roles in an adaptive world. Immunity 1998, 8, 657–665. [Google Scholar] [CrossRef] [Green Version]
  206. Redon, C.E.; Dickey, J.S.; Bonner, W.M.; Sedelnikova, O.A. γ-H2AX as a biomarker of DNA damage induced by ionizing radiation in human peripheral blood lymphocytes and artificial skin. Adv. Space Res. 2009, 43, 1171–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Herbst, R.S.; Soria, J.-C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [Green Version]
  208. Carbognin, L.; Pilotto, S.; Milella, M.; Vaccaro, V.; Brunelli, M.; Caliò, A.; Cuppone, F.; Sperduti, I.; Giannarelli, D.; Chilosi, M.; et al. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS ONE 2015, 10, e0130142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Shimizu, A.; Kaira, K.; Okubo, Y.; Utsumi, D.; Yasuda, M.; Asao, T.; Nishiyama, M.; Takahashi, K.; Ishikawa, O. Positive PD-L1 Expression Predicts Worse Outcome in Cutaneous Angiosarcoma. J. Glob. Oncol. 2017, 3, 360–369. [Google Scholar] [CrossRef]
  210. Lyu, X.; Zhang, M.; Li, G.; Jiang, Y.; Qiao, Q. PD-1 and PD-L1 Expression Predicts Radiosensitivity and Clinical Outcomes in Head and Neck Cancer and is Associated with HPV Infection. J. Cancer 2019, 10, 937–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Valentinuzzi, D.; Simončič, U.; Uršič, K.; Vrankar, M.; Turk, M.; Jeraj, R. Predicting tumour response to anti-PD-1 immunotherapy with computational modelling. Phys. Med. Biol. 2019, 64, 25017. [Google Scholar] [CrossRef] [Green Version]
  212. Davis, A.A.; Patel, V.G. The role of PD-L1 expression as a predictive biomarker: An analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef]
  213. Nishino, M.; Ramaiya, N.H.; Hatabu, H.; Hodi, F.S. Monitoring immune-checkpoint blockade: Response evaluation and biomarker development. Nat. Rev. Clin. Oncol. 2017, 14, 655–668. [Google Scholar] [CrossRef]
  214. Hong, L.; Negrao, M.V.; Dibaj, S.S.; Chen, R.; Reuben, A.; Bohac, J.M.; Liu, X.; Skoulidis, F.; Gay, C.M.; Cascone, T.; et al. Programmed Death-Ligand 1 Heterogeneity and Its Impact on Benefit from Immune Checkpoint Inhibitors in NSCLC. J. Thorac. Oncol. 2020, 15, 1449–1459. [Google Scholar] [CrossRef]
  215. Fu, J.; Wang, W.; Wang, Y.; Liu, C.; Wang, P. The role of squamous cell carcinoma antigen (SCC Ag) in outcome prediction after concurrent chemoradiotherapy and treatment decisions for patients with cervical cancer. Radiat. Oncol. 2019, 14, 146. [Google Scholar] [CrossRef] [Green Version]
  216. Henze, G.; Dummer, R.; Joller-Jemelka, H.I.; Böni, R.; Burg, G. Serum S100—A marker for disease monitoring in metastatic melanoma. Dermatology 1997, 194, 208–212. [Google Scholar] [CrossRef] [PubMed]
  217. Hauschild, A.; Engel, G.; Brenner, W.; Gläser, R.; Mönig, H.; Henze, E.; Christophers, E. Predictive value of serum S100B for monitoring patients with metastatic melanoma during chemotherapy and/or immunotherapy. Br. J. Dermatol. 1999, 140, 1065–1071. [Google Scholar] [CrossRef] [PubMed]
  218. Park, Y.-A.; Sohn, S.-K.; Seong, J.; Baik, S.-H.; Lee, K.-Y.; Kim, N.-K.; Cho, C.-W. Serum CEA as a predictor for the response to preoperative chemoradiation in rectal cancer. J. Surg. Oncol. 2006, 93, 145–150. [Google Scholar] [CrossRef] [PubMed]
  219. Saito, G.; Sadahiro, S.; Ogimi, T.; Miyakita, H.; Okada, K.; Tanaka, A.; Suzuki, T. Relations of Changes in Serum Carcinoembryonic Antigen Levels before and after Neoadjuvant Chemoradiotherapy and after Surgery to Histologic Response and Outcomes in Patients with Locally Advanced Rectal Cancer. Oncology 2018, 94, 167–175. [Google Scholar] [CrossRef] [PubMed]
  220. Luo, H.; Shen, K.; Li, B.; Li, R.; Wang, Z.; Xie, Z. Clinical significance and diagnostic value of serum NSE, CEA, CA19-9, CA125 and CA242 levels in colorectal cancer. Oncol. Lett. 2020, 20, 742–750. [Google Scholar] [CrossRef]
  221. Grunnet, M.; Sorensen, J.B. Carcinoembryonic antigen (CEA) as tumor marker in lung cancer. Lung Cancer 2012, 76, 138–143. [Google Scholar] [CrossRef] [PubMed]
  222. Park, B.-W.; Oh, J.-W.; Kim, J.-H.; Park, S.H.; Kim, K.-S.; Kim, J.H.; Lee, K.S. Preoperative CA 15-3 and CEA serum levels as predictor for breast cancer outcomes. Ann. Oncol. 2008, 19, 675–681. [Google Scholar] [CrossRef]
  223. Lee, J.S.; Park, S.; Park, J.M.; Cho, J.H.; Kim, S.I.; Park, B.-W. Elevated levels of preoperative CA 15-3 and CEA serum levels have independently poor prognostic significance in breast cancer. Ann. Oncol. 2013, 24, 1225–1231. [Google Scholar] [CrossRef] [PubMed]
  224. Lee, T.; Teng, T.Z.J.; Shelat, V.G. Carbohydrate antigen 19–9—Tumor marker: Past, present, and future. World J. Gastrointest. Surg. 2020, 12, 468–490. [Google Scholar] [CrossRef]
  225. Dawson, S.-J.; Tsui, D.W.Y.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.-F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Murtaza, M.; Dawson, S.-J.; Tsui, D.W.Y.; Gale, D.; Forshew, T.; Piskorz, A.M.; Parkinson, C.; Chin, S.-F.; Kingsbury, Z.; Wong, A.S.C.; et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013, 497, 108–112. [Google Scholar] [CrossRef] [PubMed]
  227. Yong, E. Cancer biomarkers: Written in blood. Nature 2014, 511, 524–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Lee, M.; Samstein, R.M.; Valero, C.; Chan, T.A.; Morris, L.G.T. Tumor mutational burden as a predictive biomarker for checkpoint inhibitor immunotherapy. Hum. Vaccin. Immunother. 2020, 16, 112–115. [Google Scholar] [CrossRef]
  229. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: A pan-cancer analysis. Lancet Oncol. 2017, 18, 1009–1021. [Google Scholar] [CrossRef] [Green Version]
  230. Chan, T.A.; Yarchoan, M.; Jaffee, E.; Swanton, C.; Quezada, S.A.; Stenzinger, A.; Peters, S. Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann. Oncol. 2019, 30, 44–56. [Google Scholar] [CrossRef]
  231. Gandara, D.R.; Paul, S.M.; Kowanetz, M.; Schleifman, E.; Zou, W.; Li, Y.; Rittmeyer, A.; Fehrenbacher, L.; Otto, G.; Malboeuf, C.; et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018, 24, 1441–1448. [Google Scholar] [CrossRef] [PubMed]
  232. Guibert, N.; Mazieres, J.; Delaunay, M.; Casanova, A.; Farella, M.; Keller, L.; Favre, G.; Pradines, A. Monitoring of KRAS-mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget 2017, 8, 38056–38060. [Google Scholar] [CrossRef] [Green Version]
  233. Chaudhuri, A.A.; Binkley, M.S.; Osmundson, E.C.; Alizadeh, A.A.; Diehn, M. Predicting Radiotherapy Responses and Treatment Outcomes through Analysis of Circulating Tumor DNA. Semin. Radiat. Oncol. 2015, 25, 305–312. [Google Scholar] [CrossRef] [Green Version]
  234. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
  235. Yi, M.; Qin, S.; Zhao, W.; Yu, S.; Chu, Q.; Wu, K. The role of neoantigen in immune checkpoint blockade therapy. Exp. Hematol. Oncol. 2018, 7, 28. [Google Scholar] [CrossRef] [PubMed]
  236. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Mahanty, S.; Prigent, A.; Garraud, O. Immunogenicity of infectious pathogens and vaccine antigens. BMC Immunol. 2015, 16, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Wang, S.; He, Z.; Wang, X.; Li, H.; Liu, X.-S. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. Elife 2019, 8, e49020. [Google Scholar] [CrossRef] [PubMed]
  239. Ku, G.Y.; Yuan, J.; Page, D.B.; Schroeder, S.E.A.; 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] [PubMed]
  240. Delyon, J.; Mateus, C.; Lefeuvre, D.; Lanoy, E.; Zitvogel, L.; Chaput, N.; Roy, S.; Eggermont, A.M.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]
  241. Bai, R.; Lv, Z.; Xu, D.; Cui, J. Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark. Res. 2020, 8, 34. [Google Scholar] [CrossRef]
  242. Kovacs, C. Cytokine profiles in patients receiving wide-field + prostate boost radiotherapy (xRT) for adenocarcinoma of the prostate. Cytokine 2003, 23, 151–163. [Google Scholar] [CrossRef]
  243. Johnke, R.M.; Edwards, J.M.; Evans, M.J.; Nangami, G.N.; Bakken, N.T.G.; Kilburn, J.M.; Lee, T.-K.; Allison, R.R.; Karlsson, U.L.; Arastu, H.H. Circulating cytokine levels in prostate cancer patients undergoing radiation therapy: Influence of neoadjuvant total androgen suppression. In Vivo 2009, 23, 827–833. [Google Scholar]
  244. Dirksen, S.R.; Kirschner, K.F.; Belyea, M.J. Association of symptoms and cytokines in prostate cancer patients receiving radiation treatment. Biol. Res. Nurs. 2014, 16, 250–257. [Google Scholar] [CrossRef]
  245. Bedini, N.; Cicchetti, A.; Palorini, F.; Magnani, T.; Zuco, V.; Pennati, M.; Campi, E.; Allavena, P.; Pesce, S.; Villa, S.; et al. Evaluation of Mediators Associated with the Inflammatory Response in Prostate Cancer Patients Undergoing Radiotherapy. Dis. Markers 2018, 2018, 9128128. [Google Scholar] [CrossRef]
  246. Salgado, R.; Junius, S.; Benoy, I.; van Dam, P.; Vermeulen, P.; van Marck, E.; Huget, P.; Dirix, L.Y. Circulating interleukin-6 predicts survival in patients with metastatic breast cancer. Int. J. Cancer 2003, 103, 642–646. [Google Scholar] [CrossRef]
  247. Benoy, I.H.; Salgado, R.; van Dam, P.; Geboers, K.; van Marck, E.; Scharpé, S.; Vermeulen, P.B.; Dirix, L.Y. Increased serum interleukin-8 in patients with early and metastatic breast cancer correlates with early dissemination and survival. Clin. Cancer Res. 2004, 10, 7157–7162. [Google Scholar] [CrossRef] [Green Version]
  248. Kawaguchi, K.; Sakurai, M.; Yamamoto, Y.; Suzuki, E.; Tsuda, M.; Kataoka, T.R.; Hirata, M.; Nishie, M.; Nojiri, T.; Kumazoe, M.; et al. Alteration of specific cytokine expression patterns in patients with breast cancer. Sci. Rep. 2019, 9, 2924. [Google Scholar] [CrossRef]
  249. Eide, H.A.; Knudtsen, I.S.; Sandhu, V.; Løndalen, A.M.; Halvorsen, A.R.; Abravan, A.; Kure, E.H.; Bogsrud, T.V.; Brustugun, O.T.; Kyte, J.A.; et al. Serum cytokine profiles and metabolic tumor burden in patients with non-small cell lung cancer undergoing palliative thoracic radiation therapy. Adv. Radiat. Oncol. 2018, 3, 130–138. [Google Scholar] [CrossRef] [Green Version]
  250. Cha, H.; Lee, E.J.; Seong, J. Multi-analyte analysis of cytokines that predict outcomes in patients with hepatocellular carcinoma treated with radiotherapy. World J. Gastroenterol. 2017, 23, 2077–2085. [Google Scholar] [CrossRef]
  251. Shevtsov, M.; Sato, H.; Multhoff, G.; Shibata, A. Novel Approaches to Improve the Efficacy of Immuno-Radiotherapy. Front. Oncol. 2019, 9, 156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Kachikwu, E.L.; Iwamoto, K.S.; Liao, Y.-P.; DeMarco, J.J.; Agazaryan, N.; Economou, J.S.; McBride, W.H.; Schaue, D. Radiation enhances regulatory T cell representation. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 1128–1135. [Google Scholar] [CrossRef] [Green Version]
  253. De Leve, S.; Wirsdörfer, F.; Jendrossek, V. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy. Front. Immunol. 2019, 10, 698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Schalper, K.A.; Carleton, M.; Zhou, M.; Chen, T.; Feng, Y.; Huang, S.-P.; Walsh, A.M.; Baxi, V.; Pandya, D.; Baradet, T.; et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 2020, 26, 688–692. [Google Scholar] [CrossRef] [PubMed]
  255. Yuen, K.C.; Liu, L.-F.; Gupta, V.; Madireddi, S.; Keerthivasan, S.; Li, C.; Rishipathak, D.; Williams, P.; Kadel, E.E.; Koeppen, H.; et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat. Med. 2020, 26, 693–698. [Google Scholar] [CrossRef]
  256. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, 6478. [Google Scholar] [CrossRef]
  257. Shan, Z.; Wang, H.; Zhang, Y.; Min, W. The Role of Tumor-Derived Exosomes in the Abscopal Effect and Immunotherapy. Life 2021, 11, 381. [Google Scholar] [CrossRef]
  258. Zhou, B.; Xu, K.; Zheng, X.; Chen, T.; Wang, J.; Song, Y.; Shao, Y.; Zheng, S. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct. Target. Ther. 2020, 5, 144. [Google Scholar] [CrossRef] [PubMed]
  259. Greening, D.W.; Gopal, S.K.; Xu, R.; Simpson, R.J.; Chen, W. Exosomes and their roles in immune regulation and cancer. Semin. Cell Dev. Biol. 2015, 40, 72–81. [Google Scholar] [CrossRef]
  260. Tuncay Cagatay, S.; Mayah, A.; Mancuso, M.; Giardullo, P.; Pazzaglia, S.; Saran, A.; Daniel, A.; Traynor, D.; Meade, A.D.; Lyng, F.; et al. Phenotypic and Functional Characteristics of Exosomes Derived from Irradiated Mouse Organs and Their Role in the Mechanisms Driving Non-Targeted Effects. Int. J. Mol. Sci. 2020, 21, 8389. [Google Scholar] [CrossRef] [PubMed]
  261. Yin, T.; Xin, H.; Yu, J.; Teng, F. The role of exosomes in tumour immunity under radiotherapy: Eliciting abscopal effects? Biomark. Res. 2021, 9, 22. [Google Scholar] [CrossRef]
  262. Todryk, S.; Melcher, A.A.; Hardwick, N.; Linardakis, E.; Bateman, A.; Colombo, M.P.; Stoppacciaro, A.; Vile, R.G. Heat shock protein 70 induced during tumor cell killing induces Th1 cytokines and targets immature dendritic cell precursors to enhance antigen uptake. J. Immunol. 1999, 163, 1398–1408. [Google Scholar]
  263. Gastpar, R.; Gehrmann, M.; Bausero, M.A.; Asea, A.; Gross, C.; Schroeder, J.A.; Multhoff, G. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 2005, 65, 5238–5247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  265. Del Re, M.; Marconcini, R.; Pasquini, G.; Rofi, E.; Vivaldi, C.; Bloise, F.; Restante, G.; Arrigoni, E.; Caparello, C.; Bianco, M.G.; et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Br. J. Cancer 2018, 118, 820–824. [Google Scholar] [CrossRef] [Green Version]
  266. Kang, J.; Demaria, S.; Formenti, S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J. Immunother. Cancer 2016, 4, 51. [Google Scholar] [CrossRef] [Green Version]
  267. Postow, M.A.; Knox, S.J.; Goldman, D.A.; Elhanati, Y.; Mavinkurve, V.; Wong, P.; Halpenny, D.; Reddy, S.K.; Vado, K.; McCabe, D.; et al. A Prospective, Phase 1 Trial of Nivolumab, Ipilimumab, and Radiotherapy in Patients with Advanced Melanoma. Clin. Cancer Res. 2020, 26, 3193–3201. [Google Scholar] [CrossRef] [Green Version]
  268. Sundahl, N.; Seremet, T.; van Dorpe, J.; Neyns, B.; Ferdinande, L.; Meireson, A.; Brochez, L.; Kruse, V.; Ost, P. Phase 2 Trial of Nivolumab Combined with Stereotactic Body Radiation Therapy in Patients with Metastatic or Locally Advanced Inoperable Melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 828–835. [Google Scholar] [CrossRef]
  269. McBride, S.; Sherman, E.; Tsai, C.J.; Baxi, S.; Aghalar, J.; Eng, J.; Zhi, W.I.; McFarland, D.; Michel, L.S.; Young, R.; et al. Randomized Phase II Trial of Nivolumab with Stereotactic Body Radiotherapy Versus Nivolumab Alone in Metastatic Head and Neck Squamous Cell Carcinoma. J. Clin. Oncol. 2021, 39, 30–37. [Google Scholar] [CrossRef]
  270. University College, London, Merck Sharp & Dohme Corp. A Trial Assessing the Effect of Pembrolizumab Combined with Radiotherapy in Patients with Relapsed, Refractory, Specified Stages of Cutaneous T-Cell Lymphoma (CTCL) Mycosis Fungoides (MF)/Sezary Syndrome (SS): NCT03385226, UCL/17/0053. Available online: https://clinicaltrials.gov/show/NCT03385226 (accessed on 13 May 2021).
  271. Emory University|Merck Sharp & Dohme Corp. Pembrolizumab and Stereotactic Radiosurgery for Melanoma or Non-Small Cell Lung Cancer Brain Metastases: NCT02858869, IRB00086461|NCI-2016-00718|RAD3179-16. Available online: https://clinicaltrials.gov/show/NCT02858869 (accessed on 13 May 2021).
  272. City of Hope Medical Center|National Cancer Institute (NCI). Pembrolizumab and Palliative Radiation Therapy in Treating Patients with Metastatic Esophagus, Stomach, or Gastroesophageal Junction Cancer: NCT02830594, 16099|NCI-2016-00686. Available online: https://clinicaltrials.gov/show/NCT02830594 (accessed on 13 May 2021).
  273. University of Erlangen-Nürnberg Medical School. Investigation of the Timely-Coordinated Therapy of Patients with Metastatic Cancer by Radiotherapy Together with Immune Checkpoint Inhibition: NCT03453892, ST-ICI. Available online: https://clinicaltrials.gov/show/NCT03453892 (accessed on 13 May 2021).
  274. University Health Network, Toronto. Immunotherapy and Palliative Radiotherapy Combined in Patients with Advanced Malignancy: NCT03042156, 16-6189. Available online: https://clinicaltrials.gov/show/NCT03042156 (accessed on 13 May 2021).
  275. Giuliani, J.; Fiorica, F. Cost-effectiveness of immune checkpoint inhibitors and radiotherapy in advanced non-small cell lung cancer. J. Oncol. Pharm. Pract. 2021, 10781552211038925. [Google Scholar] [CrossRef]
  276. Golubnitschaja, O.; Topolcan, O.; Kucera, R.; Costigliola, V. 10th Anniversary of the European Association for Predictive, Preventive and Personalised (3P) Medicine—EPMA World Congress Supplement 2020. EPMA J. 2020, 5, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Goldstein, E.; Yeghiazaryan, K.; Ahmad, A.; Giordano, F.A.; Fröhlich, H.; Golubnitschaja, O. Optimal multiparametric set-up modelled for best survival outcomes in palliative treatment of liver malignancies: Unsupervised machine learning and 3 PM recommendations. EPMA J. 2020, 11, 505–515. [Google Scholar] [CrossRef] [PubMed]
  278. Kucera, R.; Pecen, L.; Topolcan, O.; Dahal, A.R.; Costigliola, V.; Giordano, F.A.; Golubnitschaja, O. Prostate cancer management: Long-term beliefs, epidemic developments in the early twenty-first century and 3PM dimensional solutions. EPMA J. 2020, 11, 399–418. [Google Scholar] [CrossRef]
  279. Liskova, A.; Samec, M.; Koklesova, L.; Giordano, F.A.; Kubatka, P.; Golubnitschaja, O. Liquid Biopsy is Instrumental for 3PM Dimensional Solutions in Cancer Management. J. Clin. Med. 2020, 9, 2749. [Google Scholar] [CrossRef] [PubMed]
  280. Gerner, C.; Costigliola, V.; Golubnitschaja, O. Multiomic patterns in body fluids: Technological challenge with a great potential to implement the advanced paradigm of 3P medicine. Mass Spectrom. Rev. 2020, 39, 442–451. [Google Scholar] [CrossRef] [PubMed]
Table 1. Abscopal effect case reports (no immunotherapy).
Table 1. Abscopal effect case reports (no immunotherapy).
ReferenceAgeGenderHisto-pathologyPrimary Tumor SiteTreatment of Orimary TumorMetastasis SiteSystemic TreatmentRTIrradiation SiteNon-Irradiated Abscopal RegressionTime Frame for Abscopal ResponseReported Parameters
Lome et al. (1970)
[7]
66MTransitional cell carcinoma of bladderBladderRTLung, mediastinal + retroperitoneal LNs, adrenal glandsn/a40 Gy,
4 weeks
PelvisPulmonary lesions5 monthsBlood pressure, hematocrit, blood urea nitrogen,
Ehlers et al. (1973)
[8]
35FAdeno-carcinoma (unknown origin)Unknownn/aneck, axilla + mediastinumn/a40 Gy, 20 frNeck/supra-clavicular LNAxilla and mediastinum2 weeks
Antoniades et al. (1977)
[9]
44MLymphomaLeft axillary + bilateral supra-clavicular LNsRTRight lung, abdominal LNn/a30 Gy, 20 frLN above diaphragm (mantle field)Abdominal LNn/aBlood cunt values: hemoglobin, hematocrit, white blood cells (WBC), platelets, differential count of neutrophils, eosinophils, basophils, lymphocytes + monocytes
40MLympho-cytic lymphomaRight axillary LNRTAbdominal LNn/a30 Gy, 20 frLN above diaphragm (mantle field)Abdominal LNn/a
Fairlamb et al. (1981)
[10]
73FRenal cell carcinoma (RCC)Left kidneyNephrec-tomyLeft groin, lung, hila, pubic bonen/a40 Gy, 15 frGroinLung3 month
Rees et al. (1981)
[11]
n/an/aMixed-cellularity Hodgkin lymphoman/an/an/an/a35 Gy,
28 days
Mantle fieldPelvisn/a
n/an/aPre-dominant Hodgkinn/an/an/an/a36 Gy,
24 days
Mantle fieldPelvis, para-sortic nodesn/a
n/an/aRecurrenced Hodgkin unspecificn/an/an/an/a38 Gy,
39 days
Para-aortic and pelvic nodesLeft axilla LNn/a
n/an/aMixed-cellularity Hodgkin lymphoman/an/an/an/a40 Gy,
29 days
Mantle fieldPara-aortic regionn/a
n/an/aBrill Symmers diseasen/an/an/an/a21 Gy,
15 days
Chest wall, right axillaryLeft axilla, spleenn/a
n/an/aLympho-sacroman/an/an/an/a18 Gy,
13 days
Right groinRight neck LN2 month
n/an/aLymphoman/an/an/an/a35/20 Gy, 34 daysRight breast, pelvisMediastinal massn/a
n/an/aReticulum cell sarcoman/an/an/an/a39 Gy, 43 daysPara-aortic and pelvic nodesRight axillan/a
n/an/aFollicular lymphoman/an/an/an/a30 Gy,
22 days
Neck, supra-clavicular areaBilateral axillan/a
Robin et al. (1981)
[12]
59FHistiocytic lymphomaRight kidneyCOPP, BACOPLeft kidneyn/a20 Gy, 10 frRight kidneyLeft kidneyn/a
Rees et al. (1983)
[13]
49MAdeno-carcinoma (esophagus)Esophagusn/aLungn/a40 Gy, 20 frEsophagusLung14 month
n/an/aAdeno-carcinoma (lung)Lung (left lower lobe)n/aMediastinum, subcutaneous metastases (forehead, left shoulder)n/a35 Gy, 10 frMedi-astinum and left lower lungSubcutaneous metastases (forehead, left shoulder)2 weeks
MacManus et al. (1994)
[14]
58MRenal cell carcinoma (RCC)Right kidneyRTLeft lung, paratracheal lymphadeno-pathyMedroxy-proges-terone acetate20 Gy, 10 frRight kidneyLung, paratracheal LN4 monthTNF-α, TNF-β,
INF-γ, IL-2 receptor, IL-6
Ohba et al. (1998)
[15]
76MHepato-cellular carcinomaLiverResection/CTSecond thoracic vertebraAcetate36 Gy, n/aThoracic vertebralLiver10 monthsHemoglobin, leucocyte, platelet count, bilirubin, aspartate amino-transferase, alanine amino-transferase, albumin, alkaline phosphatase, AFP, PIVKA-II, IL-1β, IL-2, IL-4, IL-6, HGF, TNF-α
Nam et al. (2005)
[16]
65MHepato-cellular carcinomaLivern/aSkull, ribs (3–6th), sternumn/a30 Gy,SkullLiver, ribs, sternum10 monthsAST, ALT, AFP
Wersäll et al. (2006)
[17]
83FRenal cell carcinoma (RCC)Right kidneyn/aLung, LN, abdomenn/a32 Gy,
4 fr
KidneyLN, lung2 years
55FRenal cell carcinoma (RCC)Right kidneyn/aLN, aorta, livern/a32 Gy,
4 fr
KidneyLung5 months
Takaya et al. (2007)
[18]
69FUterine cervixPelvicn/aPara-aortic LNn/a28.8 Gy, 16 frPelvisPara-aortic LNn/aserum levels of squamous cell carcinoma (SCC) antigen
22 Gy, 21 fr
24 Gy,
4 fr
Isobe et al. (2009)
[19]
65MNatural killer cell lymphoman/an/aSkin, submandibular LNChemo-therapy40 GySkinSub-mandibular LN2 monthsWBC, haemoglobin, platelet count, serum lactate dehydrogenase, IL-2, TIA-1, granzyme b, CD2, CD3, CD4, CD5, CD7, CD8, CD16, CD19, CD20, CD25, CD30, CD38, CD56, TCR αβ, TCR γδ.
Lakshmanagowda et al. (2009)
[20]
65FChronic lymphocytic leukemiaLeukemian/aRight axillary LN, right neck LNn/a24 Gy, 12 frRight axillary LNRight neck LNDuring treatment
Okuma et al. (2011)
[21]
63MHepato-cellular carcinomaLivern/aLung, mediastinal LNn/a60.75 Gy,
27 fr
Mediastinal LNLung1 month
Ishiyama et al. (2012)
[22]
61MRenal cell carcinoma (RCC)Left kidneyNephrec-tomyLeft adrenal gland, lung, multiple mediastinal + hilar LN, bone, spine, brainn/a
n/a
18 GyBrainLung, LN1 month
40 Gy,
5 fr
bone, spine
Tubin et al. (2012)
[23]
72MMedullary thyroid carcinomaThyroidThyroidec-tomyLeft supraclavicular LN, left infraclavicular LN, mediastinal LN levels 4R + 6n/a30 Gy,
3 fr
4R LN level6 LN level1 month
Siva et al. (2013)
[24]
78FNon-small-cell lung carcinoma (NSCLC)Lung (left upper lobe + right lower lobe)Carboplatin, paclitaxel, RTRight adrenal, right humeral headChemo-therapy (carboplatin, paclitaxel)60 Gy, 30 frLeft upper lobeRight adrenal, right humeral head12 months
26 Gy,
1 fr
Right lower lobe
Teulings et al. (2013)
[25]
67MMelanomaLeft scapulaResectionAxilla + suprascapular region (LN), brainChemo-therapy50 Gy,
30 fr
Axilla + supra-scapular regionn/an/aS100, CD8, CD68, CD3, IL-4, IL-10, Il-17, TNF-α, IFN-γ,
20 Gy,
4 fr
Whole brainLung, mediastinum2 weeks
Lock et al. (2015)
[26]
71MHepato-cellular carcinomaLiverRTLungn/a70 Gy,
15 fr
LiverLung4 monthsAFP, liver enzymes
Yarchoan et al. (2015)
[27]
60MNon-small-cell lung carcinoma (NSCLC)Lung (right upper lobe)n/aBrain, hilar LN, left adrenal gland, left lower lung lobe, liverChemo-therapyUnknownBrainAdrenal, lung, + liver1 monthHistory of tobacco use, CK7, TTF-1, CK20, EGFR
Desar et al. (2016)
[28]
19MDiffuse-type giant cell tumorKneeImatinib, femoral amputationLung, mediastinal LN, right hilarTyrosine kinase receptor inhibitor (imatinib), steroid30 Gy,
10 fr
Right hilarLeft lung1 monthHemoglobulin, leukocytes, platelets, neutrophils, lymphocytes, monocytes, sodium, albumin, CRP
Orton et al. (2016)
[29]
84MPleomorphic soft tissue sarcomaScalpResectionPinna of left ear, parotid gland, lungn/a40 Gy,
8 fr
Post-auricular lesionLung2 months
Saba et al. (2016)
[30]
69FMultiple myeloman/aChemo-therapy (melphalan, prednisone)Left humerus, bilateral clavicles, right scapula left skull, right anterior + posterior thigh, stomachChemo-therapy (melphalan, prednisone)150.5 GyLeft humerus, bilateral clavicles, right scapula, left skull, right anterior LN + posterior thigh, stomachHead of left triceps5 monthsSerum IgG levels
Joe et al. (2017)
[31]
57FSquamous carcinoma of anal canal (SCCA)Anal canalRTMesorectum, perirectal, right internal iliac + obturator LN, liver, right liliac boneChemo-therapy54 Gy,
30 fr
Primary anal tumor, affected pelvic lymph nodes, iliac bone metastasisLiver1 monthSCC antigen, PD-1, PD-L1, CD163, CD3, CD8 expression of tumor-infiltrating lymphocytes (TILs)
Lesueur et al. (2017)
[32]
89MNeuroendocrine large-cell thymic carcinomaThymus with sternal extensionn/aLung, pancreas, right lower paratracheal LNn/a29.6 Gy,
8 fr
SternumLung4 monthsCK7, synaptophysin + chromogranin A + negative for CK5, CK6, CK20, TTF-1, S100, PSA, melan-A
Azami et al. (2018)
[33]
64FBreast carcinomaRight breastIrradiationLung, bone (femur, lumbar vertebrae + sacrum), LNs (lung, right axilla, right supraclavicular area, mediastinum)n/aa60 GyRight breastLung, LNs10 monthsHER2, KI-67, CEA, CA15-3
28 GyReft femur
39 GyLumbar vertebrae + sacrum
Bruton Joe et al. (2018)
[34]
74MAdeno-carcinoma (esophagus)EsophagusEsophagec-tomyLN, right renal vesselsn/a30 Gy,
10 fr
Primary tumor, closest LNRenal vein12 monthsHemoglobin
Chantharasamee et al. (2018)
[35]
51FMelanoma of unknown primaryLeft lower extremities left inguinal massn/aBilateral inguinal LN, multiple LN throughout abdominal + pelvic cavityChemo-therapy (carboplatin, paclitaxel)20 Gy,
NR
Bilateral inguinal LNLNs6 monthsVimentin, S100, HMB-45
Chino et al. (2018)
[36]
58MNon-small-cell lung carcinoma (NSCLC)LungSurgery, chemotherapyLiverChemo-therapy60 Gy,
8 fr
LungLiver5 monthsAFP
Chuang et al. (2018)
[37]
74FAdeno-carcinoma (colon)Colorectal cancern/aLeft lung, liver, brainn/a30 Gy,
10 fr
BrainLeft lung2 monthsCK20, CDX2, thyroid transcription factor 1 (TTF-1), CK7
Hamilton et al. (2018)
[38]
47MNon-small-cell lung carcinoma (NSCLC)lung (left upper lobe)n/aLeft mediastinum, bilateral hilar, brainn/a25 Gy,
5 fr
BrainLeft upper lobe left mediastinum1 monthPrevious illness
Leung et al. (2018)
[39]
65FDuctal carcinomaRight breastn/a8th thoracic vertebra, axillary LNn/a225 Gy,
15 fr
BreastAxillary LN12 monthsHemoglobin concentration, CEA, CA-125, CA15-3
50 Gy,
25 fr
Thoracic bone
Agyeman et al. (2019)
[40]
56MDermatofibro-sarcoma protuberans (DFSP)Left lower legLocal excisionPosterior torsoChemo-therapy (imatinib mesylate)40 Gy, 20 fr (Cobalt-60)Left lower limbPosterior torso5 monthsCD34, actin, desmin, S100
Bonilla et al. (2019)
[41]
78FAdeno-carcinoma (gastric)Gastric antrumGastrojejunostomyRetroperitoneal space 30 Gy,
10 fr
Gastric mass + marginRetro-peritoneal paraaortic adenopathies + gastrohepatic ligament3 monthscarbohydrate antigen 19-9 (CA 19-9), CA 125, ACE
Kim et al. (2019)
[42]
70MCholangiocarcinomaLung (right upper lobe)Chemo-therapyLiverChemo-therapy48 Gy,
4 fr
Right upper lung lobeLiver3 monthsBilirubin, history of tobacco use, alcohol consumption, CK7, TTF-1, CA 19-9, Napsin A, CK20, anaplastic lymphoma kinase (ALK), PD-L1,
Kuroda et al. (2019)
[43]
76FAdeno-carcinoma (lung)Lung (right upper lobeRight upper lobectomyMultiple mediastinal + right hilar LNsn/a60 Gy,
30 fr
Multiple mediastinal + right hilar LNsNew left hilar + right supra-clavicular LN, lung3 monthsEGFR mutation, PD-L1 tumor proportion score, CEA, CD8+
Ellerin et al. (2020)
[44]
84FNon-small cell carcinoma (right gland)Deep lobe of right parotid glandn/aLeft upper lobe, bilateral pulmonary masses, bilateral hilar, left paratracheal LNs in right upper lobe, right cervical LN, mediastinal LNn/a50 Gy,
20 fr
Primary siteLung, mediastinal LN2 weeksHistory of tobacco use, previous illness, Lactate dehydrogenase, CK7, GATA-3, CK5/6, p40, PD-L1, mutations in NTRK1, NTRK2, NTRK3
Guan et al. (2020)
[45]
76FB3 thymomaThymusPalliative RTMultiple lung/LN metastasesn/a66 Gy,
33 fr
Not clarifiedRegression of thymic lesion + lung metastases in non-irradiated area2 monthsPD-L1
Ohmatsu et al. (2020)
[46]
77MHepato-cellular carcinomaLiver
(right lobe)
hepatic segment-ectomyLiver, inferior vena cava, lungn/a42 Gy (boost),
14 fr
Inferior vena cavaLung1 monthMediacal history, AFP,
Mazzaschi et al. (2021)
[47]
66MHigh-grade squamous cell carcinomaLateral pharyngeal wallChemo-therapy, RTMultiple pathological left cervical LNs left humerusChemo-therapy (docetaxel, cisplatin, 5-fluorouracil)70 GyOropharynx, rhino-pharynx, hypo-pharynx, larynx, bilateral jugular-digastric LNsHumerus2 monthsp53 hyper-expressed
40 GyCervical + V level LNs
Tomita et al. (2021)
[48]
88FAdeno-carcinomaUterusRadical hysterectomy with bilateral salphingooph-erectomyLung, recurrent primaryn/a65 Gy, 26 frRecurrent primaryMultiple lung metastases5 monthsMedical history, CA-125
Abbreviations: M: male; F: female; n/a: not available; RT: radiotherapy; LN: lymph nodes; WBC: white blood cells; fr: fractions; RCC: renal cell carcinoma; TNF-α: tumor necrosis factor alpha; IFN-γ: interferon gamma; IL: interleukin; CT: computed tomography; AFP: α fetoprotein; HGF: hepatocyte growth factor; AST: aspartate aminotransferase;ALT: alanine aminotransferase; SCC: squamous cell carcinoma; TIA-1: TIA1 Cytotoxic Granule Associated RNA Binding Protein; TCR: T-cell receptor; NSCLC: non-small-cell lung carcinoma; CK: cytokeratin; TTF-1: thyroid transcription factor 1; EGFR: epidermal growth factor receptor; CRP: C-reactive protein; SCCA: squamous carcinoma of anal canal; PD-1: programmed cell death protein 1; PD-L1: PD-1 receptor-ligand 1; TIL: tumor-infiltrating lymphocyte; PSA: Prostate-specific antigen; HER2: human epidermal growth factor receptor 2; CEA: carcinoembryonic antigen; CA15-3: cancer antigen 15-3; CA-125: cancer antigen 125; CA 19-9: carbohydrate antigen 19-9; ALK: anaplastic lymphoma kinase; DFSP: Dermatofibro-sarcoma protuberans.
Table 2. Abscopal effect case reports with immunotherapy.
Table 2. Abscopal effect case reports with immunotherapy.
ReferenceAgeGenderHisto-PathologyPrimary Tumor SiteTreatment of Primary TumorMetastasis SiteSystemic TreatmentRTIrradiation SiteNon-Irradiated Abscopal RegressionTime Frame for Abscopal ResponseImmuno-TherapyReported Parameters
Postow et al. (2012)
[49]
33FMelanomaUpper backExcisionLung, right hilar LN, paraspinal region, spleenChemo-therapy, immuno-therapy28.5 Gy,
3 fr
Paraspinal regionRight hilar LN, spleen4 monthsIpilimumab
(CTLA-4)
BRAF mutation, NY-ESO-1 expression, CD4+ ICOShigh, CD14+ HLA-DRlow, CD8+, CD4+ T cells, seromic analysis
Hiniker et al. (2012)
[51]
57MMelanomaLeft posterior armExcisionLeft axillary, liver, left upper armImmuno-therapy54 Gy,
3 fr
LiverAll metastases6 monthsIpilimumab
(CTLA-4)
Golden et al. (2013)
[52]
64MAdeno-carcinoma (lung)LungChemo-therapy (pemetrexed, carboplatin, gemcitabine, vinorelbine)Left + right supra-clavicular LN, right upper lobe, left lower lobe, bilateral hilar, mediastinal adenopathy, liver, sacrumChemo-therapy (pemetrexed, carboplatin, gemcitabine, vinorelbine), immuno-therapy59.4 Gy,
33 fr
Right lung, right supra-clavicular, right hilar, mediastinal adenopathy 2 monthsIpilimumab
(CTLA-4)
Absolute lymphocyte counts (ALCs), absolute eosinophil counts (AECs), metabolic activity LN, CD8 cytotoxic T cells, TIA-1 (cytotoxic granules), FoxP3+ (Tregs), CK7, TTF-1, CK20, CDX2, CEA level
30 Gy,
5 fr
LiverLeft lung, right lung, hilar adenopathy3 months
Stamell et al. (2013)
[53]
67MMelanomaHeadRTForehead, scalp, neck, nodal, brainChemo-therapy, immuno-therapy24 Gy,
3 fr
Primary tumorSkin metastasis8 months Melanoma antigen A3 (MEGA3), PAS domain containing 1 (PASD1) level of serum
SRSBrainComplete remissionIpilimumab
(CTLA-4)
Grimaldi et al. (2014)
[54]
n/an/aMelanoman/an/an/aImmuno-therapy30 Gy,
10 fr
BrainLivern/aIpilimumab
(CTLA-4)
n/an/aMelanoman/an/an/a30 Gy,
10 fr
BrainPelvic relapsen/a
n/an/aMelanoman/an/an/a50 Gy,
25 fr
Chest wall, right axillaLiver, cutaneous metastasesn/a
n/an/aMelanoman/an/an/a20 Gy,
5 fr
Right inguinal lymph nodeGastric, cutaneous, lung, lymph nodal + retroperitoneal abdominal metastasesn/a
n/an/aMelanoman/an/an/a30 Gy,
10 fr
Brain (WBRT)Liver, bilateral axillary + right ovaric metastasesn/a
n/an/aMelanoman/an/an/a30 Gy,
10 fr
Brain (WBRT)Lung, cutaneous, lymph nodal + abdominal metastasesn/a
n/an/aMelanoman/an/an/a30 Gy,
10 fr
Right chest wallLymph nodal, cutaneous + chest wall metastasesn/a
n/an/aMelanoman/an/an/a30 Gy,
10 fr
Vertebral metastasisLung metastasesn/a
n/an/aMelanoman/an/an/a24 Gy,
1 fr
Brain (SRT)Cutaneous metastasesn/a
n/an/aMelanoman/an/an/a20 Gy,
1 fr
Brain (SRT)Liver metastasesn/a
n/an/aMelanoman/an/an/a24 Gy,
1 fr
Brain (SRT)Lung metastasesn/a
Thallinger et al. (2014)
[55]
44MMelanoman/an/aLiver, lung, right kidney, right adrenal gland, LN, bone, brainChemo-therapy, immuno-therapy30 Gy,
10 fr
BrainKidney, lunge, liver2 monthsIpilimumab
(CTLA-4)
S100, HMB45, Melan A, BRAF, NRAS + c-KIT mutations
Okwan-Duodu et al. (2015)
[56]
50FMelanomaRight lower backExcisionRight groin, right occipital area, right suboccipital node, sub-centimeter right pulmonary node, brain, multiple retroperitoneal, subcutaneous, aortocaval, left periaortic + peripancreatic LNsImmuno-therapyWhole-brain RTBrainPulmonary, retroperitoneal, mesenteric nodes.5 monthsIL-2 therapy
Michot et al. (2016)
[57]
33MClassical Hodgkin diseaseNodal supra-diaphrag-matic areaChemo-therapy (doxorubicin (Adriamycin), bleomycin, vinblastine, dacarbazine)Mediastinal right hilar + coeliac areas LN, supra-diaphragmatic + subdiaphragmatic LN, mediastinal lymphadenopathyChemo-therapy (doxorubicin (Adriamycin), bleomycin, vinblastine, dacarbazine), immuno-therapy30 Gy, 10 frRight hilar mediastinal LNn/an/aPembrolizumab
(PD-1)
Cong et al. (2017)
[58]
64FNon-small-cell lung carcinoma (NSCLC)Left lungChemo-therapy (cisplatin, pemetrexed)Para-mediastinal tumor, lingual segment tumorChemo-therpay, immuno-therapy, EGFR inhibitor37.5 Gy, 5 frPara-mediastinal tumorLung10 monthsEndritic cells + cytokine-induced killers (DC-CIK) immunotherapyEGFR mutation
Komatsu et al. (2017)
[59]
60MAdeno-carcinoma (lung)Lung (right upper lobe)LobectomyLiver metastasis, intrapulmonary metastasisChemo-radio-therapy, immuno-therapy40 Gy,
20 fr
LiverLung3 weeksNivolumab
(PD-1)
Sato et al. (2017)
[60]
54MAdeno-carcinoma (gastric cancer)ColonDistal gastrectomyPeritoneal tumorChemo-therapy (TS-1, paclitaxel), immun-otherapy48 Gy,
24 fr
ColonPeritoneal tumor2 monthsT-cell immuno-therapy, dendritic cell (DC) therapyCEA, CA19-9
Sharabi et al. (2017)
[61]
48FNeuro-endocrine cervical carcinomaUterine, cervixChemo-therapy, RTLiver, pelvic, retro-peritoneal + pelvic LNChemo-therapy (cisplatin, etoposide), immuno-therapy20 Gy,
4 fr
Abdominal massHepatic lesion, pelvic mass, pelvic + retroperitoneal LN4 monthNivolumab
(PD-1)
Pan-cytokeratin, synaptophysin, CD99, EMA, p16, circulating tumor DNA (ctDNA), NGS mutation analysis, tumor mutational burden, PD-L1, microsatellite instability (MSI-H) status, CA-125
Shi et al. (2017)
[62]
67FPancreatic cancerPancreasChemo-therapy (gemcitabine, paclitaxel albumin)Liver, right pleura metastasisChemo-therapy (gemcitabine, paclitaxel albumin), tyrosine kinase inhibitor, immuno-therapy45 Gy,
15 fr
PancreasMetasases1 monthGM-CSFCA19-9
Xie et al. (2017)
[63]
54MRenal cell carcinoma (RCC)Left kidneyNephrectomyMultiple mediastinal, retro-peritoneal, bilateral cervical, pelvis LNTyrosine kinase inhibitors (sunitinib)32 Gy,
4 fr
Left mediastinal LNAll metastasis2 monthsPembrolizumab
(PD-1)
Circulating tumor DNA (ctDNA)
Britschgi et al. (2018)
[64]
47MNon-small-cell lung carcinoma (NSCLC)LungChemo-therapy cetuximab, RT, resectionRretro-peritoneal + abdominal lymph nodeChemo-therapy (pemetrexed), immuno-therapy18 Gy,
3 fr
2 retro-peritoneal LNsUnirradiated LNs10 weeksNivolumab
(PD-1)
History of tobacco use
Gutkin et al. (2018)
[65]
57MMelanomaLeft posterior armExcision (priamry, LN)Liver. left upper armImmuno-therapy50 Gy,
20 fr
Left posterior arm IFN therapy, ipilimumabBRAF mutation, V600
54 Gy,
3 fr
2 liver metastasesComplete response1 year
Matsushita et al. (2018)
[66]
62MRenal cell carcinoma (RCC)Right kidneyNephrectomyRight adrenal gland, lumbar vertebrae (L4)Tyrosine kinase inhibitors (sunitinib, axitinib), immuno-therapy36 Gy,
12 fr
Lumbar vertebrae (L4)Right adrenal1.5 monthsNivolumab
(PD-1)
71MRenal cell carcinoma (RCC)Right kidneyNephrectomyLeft parotid gland, soft tissues, lung, pancreas, right iliopsoas muscleInterferon-α therapy, tyrosine kinase inhibitors (sunitinib, axitinib), immuno-therapy66 Gy,
33 fr
Right iliopsoas muscleLung, pancreas1.5 monthsNivolumab
(PD-1)
Tumor burden
Rodriguez-Ruiz et al. (2018)
[67]
68MCastration-resistant prostate carcinomaProstateSurgery, chemotherapyMediastinal + inguinal LN, liver, bone, lungChemo-therapy, immuno-therapy24 Gy,
3 fr
Prostate, inguinal LNsMediastinal + retroperitoneal lesions6 monthsPoly ICLC,
DC vaccine
PSA serum concentration
Tsui et al. (2018)
[68]
65FMelanomaMaxillary gingiva + hard palateResectionFloor of mouth, right neck, lung, cervical LNImmuno-therapy50 Gy,
20 fr
Primary tumor Pembrolizumab
(PD-1)
24 Gy,
3 fr
NeckLung, mouth1 month
van Gysen et al. (2018)
[69]
66FRenal cell carcinoma (RCC)Kidney (right)Nephrectomy, TKI, immuno-therapyAbdominal mass. retroperitoneal + peritoneal LNs, lungTyrosine kinase inhibitors (sunitinib, axitinib)36 Gy,
12 fr
Abdominal massLung1 monthNivolumab
(PD-1)
Wight et al. (2018)
[70]
24MHodgkin lymphomaLeft axillary LNBEAM conditioning (carmustine, etoposide, cytarabine, melphalan)lung, widespread lymph-adenopathyBEAM conditioning (carmustine, etoposide, cytarabine, melphalan), immuno-therapy20 GyRight axillary LN Nivolumab
(PD-1)
36 GyInfra-diaphrag-matic sitesLeft pulmonary hilar, left parotid, right cervical nodal areas1.5 month
Xu et al. (2018)
[71]
69MMerkel cell carcinomaRight upper backSurgical excision, RTLNs, peripancreatic abdominal mass, left adrenal noduleImmuno-therapy50 Gy,
25 fr
Right axilla, posterior chest wall Pembrolizumab
(PD-1)
CK20
8 Gy,
1 fr
Peripan-creatic abdominal mass, left adrenal nodule, omental nodule, enlarged para-aortic LNsHypermetabolic malignancy12 months
72MMerkel cell carcinomaLeft thighSurgical excisionLeft inguinal adenopathy, bilateral inguinal LNs, supra-clavicular, mediastinal, hilar, upper abdominal nodalImmuno-therapy8 Gy,
1 fr
Mediastinal + right hilar LNUpraclavicular + abdominal LN, mediastinum, bilateral hila, left inguinal region4 monthsPembrolizumab
(PD-1)
Zhao et al. (2018)
[72]
65MSquamous cell carcinoma (SCC)Esoph-agusEsophagec-tomyLNs (left retro-peritoneal, pelvic)Chemo-therapy (cisplatin, docetaxel), immuno-therapy42 Gy,
6 fr
LNs (left retro-peritoneal)Non -irradiated LN (pelvic)2 monthsPembrolizumab
(PD-1)
ctDNA, mutant allele frequency (MAF)
Abbas et al. (2019)
[73]
69MUrothelial carcinomaBladderChemo-therapy (gemcitabine, carboplatin)Left internal iliac, para-aortic LNChemo-therapy, immuno-therapy30 Gy,
12 fr
Bladder left iliac LNAll sites4 monthsNivolumab
(PD-1)
History of tobacco use, PD-L1
Bitran et al. (2019)
[74]
62FAdeno-carcinoma (lung)Lung (left)Chemo-therapy (carboplatin, pemetrexed)Left adrenalChemo-therapy (carboplatin, pemetrexed), immuno-therapy27 Gy,
9 fr
Left lungLeft adrenal7 monthsNivolumab
(PD-1)
Epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), hemoglobin
Choi et al. (2019)
[75]
67MSquamous cell carcinoma (SCC)Lower lipResectionHilar nodes, liver, peritoneum, right parotid, right neck, mediastinum, left hilum, posterior neck subcutaneous tissue, lung, left adrenalImmuno-therapy, cobimetinibPost-operativeResection margin Atezolizumab
(PDL1)
45 Gy,
5 fr
Right sub-mandibular + neck nodesAbdomen, chest13
months
D’Andrea et al. (2019)
[76]
42FMelanomaRight upper skin of backSurgical excisionUpper-right retro pectoral region of chest wall, right ovary, brain axillaImmuno-therapy30 Gy,
15 fr
Whole brainChest, axilla3 weeksMAPK kinase (MEK) inhibitorBRAF mutation, RBI mutation, ERCC1, MLH1, MSH2, MSH6, PMS2, TUBB3, PDL-1, TrK A/B/C, MGMT expression
Garelli et al. (2019)
[77]
54MPulmonary large cell neuro-endocrine carcinomaLung (right upper lobe)Four cycles of chemotherapy (pemetrexed, cisplatin, bevacizumab)Bilateral adrenal metastasesChemo-therapy, immuno-therapy30 Gy,
10 fr
Second + third thoracic vertebraePartial regression of lung tumor + adrenal metastases4 monthsNivolumab
(PD-1)
PD-L1, EGFR, or ALK mutations
64MAdeno-carcinoma (lung)Lung (left upper lobe)Nabpaclitaxel/carboplatin with atezolizumabContralateral LN, brain, ocularChemo-therapy, immuno-therapy30 Gy,
10 fr
BrainComplete remission of lung + mediastinal tumor masses4 monthsAtezolizumab
(PDL1)
70MAdeno-carcinoma (lung)Lung (middle lob)n/aLN, brainImmuno-therapy30 Gy,
10 fr
BrainPartial regression of lung tumor2 weeksPembrolizumab
(PD-1)
Gounder et al. (2019)
[78]
25FChordomaSacrumn/aLungEZH2 inhibitor (tazemetostat)70 Gy,
35 fr
SacrumLung4 monthsNivolumab
(PD-1)
Next-generation sequencing, tumor mutation burden
Grimaux et al. (2019)
[79]
78MRenal cell carcinoma (RCC)KidneyImmuno-therapyPulmonary + costal metastasis, cutaneous, oral + genital blisters,Immuno-therapy30 Gy,
10 fr
Costal metastasesLung1 monthNivolumab
(PD-1)
Ishiyamal et al. (2019)
[80]
68FRenal pelvic cancerLeft kidneyNephroureterectomy + regional LN dissection, chemotherapy (cisplatin + gemcitabine)Bladder, paraaortic LN, left subclavian LN, right renal hilum LN, left back (local recurrence in surrounding muscles)Chemo-therapy30 Gy,
10 fr
Left backReductiomn 2 paraaortic LNs, right renal hilar LN2 monthsPembrolizumab
(PD-1)
Immuno-therapy2 non-irradiated lesions stable (paraaortic LN, right renal hilum LN)21 months
Lin et al. (2019)
[81]
71MAdeno-carcinoma (lung)Lung (right lobe)Chemo-therapyBrain, right lower lung, left lower lobeChemo-therapy, immuno-therapy48 Gy,
8 fr
BrainLung, LNs (mediastinum)4 monthsAtezolizumab
(PDL1)
Liu et al. (2019)
[82]
52FIntra-hepatic cholangio-carcinomaLiverRTLN in hepatic hilar + retro-peritoneumAapatinib + lenvatinib55Gy,
5 fr
Right hepatic lobeLN in hepatic hilar + retroperitoneum1 monthNivolumab
(PD-1)
Whole-exome sequencing, tumor mutation burden, PD-L1, ERBB2, HBV infection
59MIntra-hepatic cholangio-carcinomaLiverResection of middle hepatic lobeLeft + right lobes, hepatic hilar + retroperitoneal LNLapatinib52 Gy,
4 fr
Right hepatic lobeHepatic hilar + retroperitoneal LN2 to 5 monthsPembrolizumab
(PD-1)
51MIntra-hepatic cholangio-carcinomaLiverResection in left hepatic lobeRight lobe hepatic hilar + retro-peritoneal lymph nodeChemo-therapy, endostatin52 Gy,
4 fr
Left hepatic lobe, left retroperitoneal LNIntrahepatic LN1 monthPembrolizumab
(PD-1)
Moran et al. (2019)
[83]
71MMelanomaUnknownn/aLung, pelvis, omental mass, bilateral hilar nodesImmuno-therapy50 Gy,
5 fr
left lungPelvis1 monthIpilimumab (CTLA-4), Nivolumab
(PD-1)
S100, Melan A, TTF1, P63, CK7/20, BRAF mutations
Qin et al. (2019)
[84]
21MNodular sclerosing Hodgkin’s lymphoman/aDifferent chemotherapy regimesMultiple sites of adenopathy, osseous lesionsChemo-therapy, stell cell trans-plantation, immuno-therapy20 Gy,
5 fr
L2, left iliac crestComplete response4 monthsNivolumab
(PD-1)
PD-L1 expression, gene mutations, tumor mutation burden, hemoglobin, alkaline phosphatase, telomere FISH
34MNodular sclerosing Hodgkin’s lymphoman/aChemo-therapy (ICE)Preaortic + pelvic LNsChemo-therapy (ICE), stell cell trans-plantation, immuno-therapy36 Gy,
20 fr
Pelvic LNsComplete response1 year
23MNodular sclerosing Hodgkin’s lymphoman/aABVD chemotherapyMediastinum, multiple LNsABVD chemo-therapy, immuno-therapy40 Gy,
20 fr
Cervical + mediastinal LNsComplete response5 monthsNivolumab
(PD-1)
Shinde et al. (2019)
[85]
75MHead + neck squamous cell carcinomaLeft neck, hypo-pharynx + oropharynxImmuno-therapyLeft lungImmunotherapy14.8 Gy,
4 fr
NeckLung2 weeksIpilimumab (CTLA-4), Nivolumab
(PD-1)
History of tobacco use
13.2 Gy,
4 fr
Margin
Suzuki et al. (2019)
[86]
30FRenal cell carcinoma (RCC)Left kidneyNephrectomyLung, right ovarian, pelvic + lumber vertebrae, hilar LN, mediastinal LNs, liver, left iliac boneSunitinib, axitinib60 Gy,
30 fr
Mediastinal LNs Nivolumab
(PD-1)
LDH levels
35 Gy,
5 fr
(brachy-therapy)
Left iliac lesion left internal iliac LNLumbar vertebrae (L4)3 months
Trommer et al. (2019)
[87]
n/an/aMelanomaLeft thighResectionLung, paraaortal LN, brainImmuno-therapy20 Gy,
1 fr
Brain Pembrolizumab
(PD-1)
50 Gy,
2 fr
BrainLung1 month
n/an/aMelanomaLeft kneeResectionPerirenal region, brainImmuno-therapy40 Gy,
2 fr
Whole brain Pembrolizumab
(PD-1)
54 Gy,
3 fr
Popliteal fossa + lower left leg, brainPerirenal region1.5 month
20 Gy,
1 fr
Brain
n/an/aMelanoman/an/aLung, brainImmuno-therapy20 Gy,
1 fr
Brain Pembrolizumab
(PD-1)
20 Gy,
1 fr
Brain
20 Gy,
1 fr
BrainLung5 weeks
n/an/aNon-small cell lung carcinoma (NSCLC)Lungn/aSuprarenal glands, brain, right femur, os. sacrum left os. ischiadicumImmuno-therapy27 Gy,
3 fr;
20 Gy
Brain Nivolumab
(PD-1)
30 Gy,
3 fr
Right femurSuprarenal glands1 month
30 Gy,
3 fr
Os. sacrum, left os. ischiadicum
n/an/aNon-small cell lung carcinoma (NSCLC)Lungn/aCervival + left super-vlavicular, mediastinal Ln, left hilar, axillar LN, intracarinal LNImmuno-therapy54 Gy,
2 fr
Cervival + left supervlavicularaxillar LN6 monthsNivolumab
(PD-1)
n/an/aNon-small cell lung carcinoma (NSCLC)Lungn/aSupra- + infra-clavicular lymph drainage area, 3rd right rib, right iliac sacral joint, left inguinal, lungImmuno-therapy50.4 Gy,
1.8 fr
Supra- + infra-clavicular lymph drainage area Nivolumab
(PD-1)
30 Gy,
3 fr
3rd right rib, right iliac sacral joint, left inguinal
20 Gy,
1 fr
Left occipitalLung7 weeks
n/an/aRenal cell carcinoma (RCC)Kidneyn/aLeft os. ilium, left + right hip, right os. pubis, thoracic vertebra, lumbar vertebra, mediastinal LN, hilar, pleuralImmuno-therapy36 Gy,
3 fr
Left os. iliummediastinal LN2 monthsNivolumab
(PD-1)
30 Gy,
3 fr
Left + right hip, right os. pubis
30 Gy,
3 fr
Thoracic vertebra, lumbar vertebra
Yaguchi et al. (2019)
[88]
63MPulmonary pleo-morphic carcinomaLung (right upper lobe)Chemo-therapy (carboplatin, paclitaxel)Brain, bone + pleural metastasesChemo-therapy (carboplatin, paclitaxel), immuno-therapy30 Gy, NRRight hip joint over right femurPleura, brain + bone Nivolumab
(PD-1)
History of tobacco use, PD-L1 expression of tumor, EGFR mutation, ALK
Forner et al. (2020)
[89]
57MHead + neck squamous cell carcinomaLeft frontal sinus + ethmoid sinusesResection, chemotherapyRight lower lobe, left cervical LNs, subcarinal LN, extraconal intraorbital lesionChemo-therapy (cisplatin)66 Gy,
33 fr
Nivolumab
(PD-1)
Medical history
30 Gy,
5 fr
Intraorbital massThorax metastases, lung1 month
Hori et al. (2020)
[90]
40FRenal cell carcinoma (RCC)Left kidneyNephrectomyLung, right supra-clavicular + para-aortic LNInterferon-α, axitinib, everolimus, pazopanib30 or 40 Gy, 10 frRight supra-clavicular + para-aortic LNLung6 monthsNivolumab
(PD-1)
HLA class1, CD8, PD-L1 expression of tumor
Igarashi et al. (2020)
[91]
74MMelanomaMaxillaMaxillary resectionCervical LNs, brain, spleen, liverImmuno-therapy30 Gy,
10 fr
Whole brainLiver, spleen2 monthsNivolumab
(PD-1)
BRAF mutation
Kuhara et al. (2020)
[92]
69FUn-resectable gastric cancer (UGC)Lower portion of stomachChemo-therapy (S-1 plus oxaliplatin (SOX) + Herceptin (HER))Para-aortic, mediastinal, right iliac + Virchow LNs, bilateral subclavian + mediastinal LNs, left axillary LNH, peritoneal metastasis, left iliac, right diaphragm LN, left adrenal glandChemot-herapy (S-1 plus oxaliplatin (SOX) + Herceptin (HER)), immuno-therapy55 Gy,
22 fr
Neck + mediastinumBilateral subclavian + mediastinal LNs13 monthsNivolumab
(PD-1)
EGF-2
50 Gy,
10 fr
Left adrenalPara-aortic, bilateral iliac, right diaphragm LNs
Levitin et al. (2020)
[93]
83MRenal cell carcinoma (RCC)Left kidneyPazopanib, SBRT 40Gy,
5 fr
Right lung left femur, brain, mediastinal lymphadenopathy, pelvisReceptor tyrosine kinase inhibitor (pazopanib), immuno-therapyGKS
20 Gy
BrainRegression metastases2 yearsNivolumab
(PD-1)
Nakajima et al. (2020)
[94]
56FClear cell carcinomaKidneyNephrectomyLeft lung hilum, subcutaneous + lung metastases, right renal, right iliac boneReceptor tyrosine kinase inhibitor (pazopanib, axitinib), immuno-therapy30 Gy,
10 fr
Right iliac boneLung, right kidney, subcutaneous tissue5 monthsNivolumab
(PD-1)
Primary tumor9 months
Sohal et al. (2020)
[95]
66MMelanomaRight nasal cavityRTLeft proximal humerus, upper thoracic spine, liver, lateral left fifth rib, third, eighth, eleventh thoracic vertebral bodiesImmuno-therapy25 Gy,
5 fr
NoseAll metastasis4 monthsNivolumab
(PD-1)
Cancer history, transplantation history, BRAF, NRAS mutation
Wang et al. (2020)
[96]
57MSquamous cell carcinoma (SCC)Right upper lungResection, adjuvant RT, chemotherapyPancreas, metastatic nodules in left lower lobeChemo-therapy (gemcitabine, cisplatin)55 Gy,
25 fr
Resection margin Pembrolizumab
(PD-1)
PD-L1, ALK genetic status, history of tobacco use, whole-exome sequencing
60.2 Gy, 28 frPancreasLeft lung nodules1 month
Watanabe et al. (2020)
[97]
69FMelanoma (BRAF-wild-type)Upper left legResection, also sentinel LNsLeft groin LN, liver, muscle in upper left leg, left groinImmuno-therapy, corticosteroids, adjuvant IFN-α therapy45 Gy,
3 fr
Liver lesionsLeft groin LN, liver, muscle in upper left leg, left groin8 weeksNivolumab
(PD-1)
CD8 cells, PD-1, PD-L1, TIM-3, LAG-3, TOX, IFN-γ, perforin, granzymes, IL-2, TNF-α, DCXCL9, CXCL10, CCL, CXCL13, IDO1, CD4+ Tregs, arginase, MHC I, MHC II, APCs, β2-microglobulin, Ki67, CD3+ cells, tumor burden, FoxP3, exhausted T cells, BRAF-mutation
77MMelanoma (BRAF-wild-type)Right breast regionResection, also sentinel lymphnodesLiver, lung, abdominal LNImmunotherapy45 Gy,
3 fr
Liver metastasesStable6 weeksPembrolizumab
(PD-1)
60 Gy,
8 fr
LN, liverOther liver lesions, lung1 month
Hotta et al. (2021)
[98]
42FAdeno-carcinoma (lung)Lung (left upper lobe)EGFR tyrosine kinase inhibitor, steroidBone, brain, thoracic spineEGFR tyrosine kinase inhibitor, steroid30 Gy,
10 fr
Whole brainLung4 daysAtezolizumab
(PDL1)
EGFR. PD-L1, CEA
36 Gy,
12 fr
Thoracic spineLung2 weeks
Mampuya et al. (2021)
[99]
72MMalignant pleural mesothe-liomaRight lungChemo-therapy (carboplatin, pemetrexed),VEGF-A inhibitor (bevacizumab)Para-mediastinal + retro-pharyngeal lesionsChemo-therapy (carboplatin, pemetrexed),VEGF-A inhibitor (bevacizumab), immuno-therapy30 Gy,
10 fr
Para-mediastinal lesionsRight lung3 monthsPembrolizumab
(PD-1)
Medical history, PD-L1, tumorinfiltrating CD3 T cells, CD8 T cells
Wong et al. (2021)
[100]
n/an/aRenal cell carcinoma (RCC)Kidneyn/aLungImmuno-therapy55 Gy,
20 fr
LungLungn/aIpilimumab (CTLA-4), Nivolumab
(PD-1)
n/an/aRenal cell carcinoma (RCC)Kidneyn/aFemur, rib25 Gy,
5 fr
Femur, ribNephrectomy bedn/aIpilimumab (CTLA-4), Nivolumab
(PD-1)
n/an/aRenal cell carcinoma (RCC)Kidneyn/aLung, adrenal36 Gy,
3 fr
LungLung, several lesionsn/aIpilimumab (CTLA-4), Nivolumab
(PD-1)
30 Gy,
5 fr
AdrenalLung
n/an/aRenal cell carcinoma (RCC)Kidneyn/aBrain, intramuscularGKS 25GyBrainIntramuscularn/aIpilimumab (CTLA-4), Nivolumab
(PD-1)
55FRenal cell carcinoma (RCC)Kidneyn/aFemur, pubic bone, liver8 Gy, 1 frFemurLiver, several lesionsn/aPembrolizumab
(PD-1)
27 Gy,
3 fr
Pubic bone
n/an/aRenal cell carcinoma (RCC)Kidneyn/aLung54 Gy,
3 fr
LungLung, several lesionsn/aNivolumab
(PD-1)
42 Gy,
5 fr
Lung
25 Gy,
5 fr
Lung
n/an/aRenal cell carcinoma (RCC)Kidneyn/aLung50 Gy,
20 fr
LungLungn/aNivolumab
(PD-1)
48 Gy,
3 fr
Lung
48 Gy,
3 fr
Lung
n/an/aRenal cell carcinoma (RCC)Kidneyn/aFemur, lliac bone, brain, lung20 Gy,
5 fr
FemurLung, several lesionsn/aNivolumab
(PD-1)
24 Gy,
3 fr
Iliac bone
GKS 25 GyBrain
n/an/aRenal cell carcinoma (RCC)Kidneyn/aIliac bone, spine, lung27 Gy,
3 fr
Iliac boneLung, several lesionsn/aNivolumab
(PD-1)
24 Gy,
3 fr
SpineKidney
Abbreviations: M: male; F: female; n/a: not available; RT: radiotherapy; LN: lymph nodes; fr: fractions; CTLA-4: cytotoxic T-lymphocyte-associated protein 4; ALC: Absolute lymphocyte counts; AEC: absolute eosinophil counts; TIA-1: TIA1 Cytotoxic Granule Associated RNA Binding Protein; Tregs: regulatory T cells; CK: cytokeratin; TTF-1: thyroid transcription factor 1; CDX2: Caudal Type Homeobox 2; CEA: carcinoembryonic antigen; MEGA3: Melanoma antigen A3; PASD1: PAS domain containing 1; DC-CIK: cytokine-induced killer cells; PD-1: programmed cell death protein 1; DC: dendritic cell; CA 19-9: carbohydrate antigen 19-9; ctDNA: circulating tumor DNA, NGS: next generation sequencing; PD-L1: PD-1 receptor-ligand 1; MSI-H: high levels of microsatellite instability; CA-125: cancer antigen 125; GM-CSF: granulocyte-macrophage colony-stimulating factor; MAF: mutant allele frequency; EGFR: epidermal growth factor receptor; ALK: anaplastic lymphoma kinase; FISH: fluorescent in situ hybridization; LDH: lactate dehydrogenase; EGF: epidermal growth factor; TIM-3: T cell immunoglobulin and mucin-domain containing-3; LAG-3: lymphocyte-activation gene 3; TOX: thymocyte selection-associated high mobility group box protein; IFN-γ: interferon gamma; TNF-α: tumor necrosis factor alpha; MHC: major histocompatibility complex; APC: antigen-presenting cells.
Table 3. Abscopal effect case reports in different tumor entities.
Table 3. Abscopal effect case reports in different tumor entities.
Tumor EntitiesIncidenceMortality Cases of AbE
New Cases Worldwide in 2020 (% of All Sites) [147]New Deaths Worldwide in 2020 (% of All Sites) [147]TotalWithout ICB Under ICB
Breast11.7%6.9%22-
Lung11.4%18.0%18612
Colorectum10.0%9.4%211
Prostate7.3%3.8%1-1
Stomach5.6%7.7%11-
Esophagus3.1%5.5%22-
Thyroid3.0%0.4%11-
Bladder3.0%2.1%211
Kidney2.2%1.8%23520
Melanoma1.7%0.6%29227
Table 4. Potential predictive biomarkers for abscopal effects.
Table 4. Potential predictive biomarkers for abscopal effects.
Potential PredictorPreclinical EvidenceClinical Evidence
Tumor protein p53Camphausen et al. [183]Requirement of intact p53 to induce AbE
Strigari et al. [184]Non-irradiated tumors with functional p53 respond systematically to irradiation
CalreticulinGameiro et al. [115]Calreticulin expression as evidence for enhanced CTL lysis of non-irradiated tumor cells
Hsp70Bayer et al. [185]Plasma levels of Hsp70 associated with tumor burden, level correlates with tumor progression and radiation-induced tumor regression
γ-H2AX Siva et al. [186]γ-H2AX foci in non-irradiated PBL and hair follicles indicates AbE in normal tissue
PD-L1 Yaguchi et al. [88]Correlation of PD-L1 expression to abscopal response
Watanabe et al. [97]
Tumor markers
• SCC antigen Joe et al. [31]SCC Ag levels decreases due to systemic response
• S100 (melanoma) Watanabe et al. [97]Correlation of decrease in S100 with AbE
• CEA Golden et al. [52]Drastic drop to normal levels during systemic response
Kuroda et al. [43]Drop in plasma levels during occurrence of AbE
• CA15-3 Leung et al. [39]Significant decrease in CA15-3 levels during abscopal reaction
• CA19-9 Bonilla et al. [41]Reduced plasma levels during AbE
• CA125 Tomita et al. [48]Reduced plasma levels during AbE
ctDNA abundance and tumor mutational burden (TMB) Xie et al. [63]Correlation of ctDNA abundance with abscopal extinction of tumor cells
Zhao et al. [72]ctDNA analysis for mutation monitoring associated with AbE
Sharabi et al. [61]Correlation of elevated TMB with AbE
Tumor antigens
• melanoma antigen A3 (MAGEA3) Stamell et al. [53]Association of AB to tumor antigens with AbE
• PAS domain-containing 1 (PASD1)
• NY-ESO-1 Postow et al. [49]AB titers against NY-ESO-1 correlated positively with disease progression
Absolute lymphocyte count (ALC) Antoniades et al. [9]Correlation of irradiation-induced decrease in ALC with increase in neutrophils
Desar et al. [28]Monitoring changes of lymohocytes during AbE
Kuroda et al. [43]
Postow et al. [49]
Blattner et al. [187]
Golden et al. [52]Disease progression tracked by ALCs, white blood cells (WBCs) and absolute eosinophil counts (AECs)
Grimaldi et al. [54]ALC was increased before RT if AbE
Chen et al. [188]Correlation between increased ALC value after RT and AbE
T cell values Kuroda et al. [43]low CD8+ lymphocyte counts at the time of AbE
Postow et al. [49]IFNγ-producing CD8+ and CD4+ T cells decrease due to RT and increase slightly during AbE
Neutrophil-to-lymphocyte ratio Golden et al. [189]significantly lower baseline in non-responders
Tumor-infiltrating lymphocytes Joe et al. [31]Infiltration pattern of tumor reacting with AbE
Hori et al. [121]Infiltration of non-irradiated lesion by CD8+ T cells
Teulings et al. [25]Infiltration of abscopal reacting tumors with CD8+ T cells
Mampuya et al. [99]Moderate infiltration of CD3+ cells after occurrence of AbE
Cytokine profiles Ohba et al. [15]Monitoring serum concentrations of various cytokines in AbE, TNF-α serum levels increased in AbE
Exosomes Chen et al. [188]Circulating TEXs analysis could distinguish responders from non-responders
Table 5. Currently ongoing clinical trials.
Table 5. Currently ongoing clinical trials.
Study IdentifierStudy TitleStatusCondition or DiseaseTherapyRT SchemeCohort NPlanned Primary Outcome MeasuresPlanned Secondary Outcome Measures
NCT03480334Abscopal Effect of Radiotherapy and Nivolumab in Relapsed Hodgkin Lymphoma After Anti-PD-1 Therapy (AERN)RecruitingClassical Hodgkin lymphomaNivolumab + RT20 Gy29Abscopal response rate-
NCT03396471Study of Pembrolizumab and Concurrent Radiation in Patients With Previously Treated Carcinoma of Unknown PrimaryRecruitingCarcinoma, unspecified sitePembrolizumab + EBRT20-30 Gy,
5 fr
34Abscopal response rateResponse rate
Assess adverse events
Progression free survival
Overall survival
Time-to-progression
Disease control rate
NCT04238169Clinical Trial Assessing the Efficacy of Abscopal Effect Induced by SBRT and Immunotherapy in Advanced NSCLCRecruitingNon-Small-Cell Lung Cancer (NSCLC)(Bevacizumab +) Toripalimab + SBRT30-50 Gy, 5 fr60Objective response rateProgression free survival
Duration of response
Objective response of non-target lesionOverall survival
Incidence of adverse events
Quality of life
NCT04873440An Open-label, Phase I/II Study of Manganese Plus Radiotherapy in Patients With Metastatic Solid Tumors or LymphomaRecruitingSolid tumorManganese Chloride + RT (Chemo-immuno-therapy)standard-of-care RT or SBRT to one metastatic site10Proportion of subjects with an abscopal responseDisease control rate
Progression free survival
LymphomaNumber of treatment-related adverse eventsOverall survival
Number of participants with laboratory test abnormalities
NCT04168320SBRT-based PArtial Tumor Irradiation of HYpoxic Segment (SBRT-PATHY)RecruitingUnresectable malignant solid neoplasm bulky tumorsSBRTSBRT-based PArtial Tumor irradiation targeting HYpoxic segment30Bystander and abscopal effectsOverall survival
Progression free survival
Patient-reported outcome
Incidence of adverse events
Response evaluation criteria in solid tumors
Timing
NCT03449238Pembrolizumab And Stereotactic Radiosurgery (Srs) Of Selected Brain Metastases In Breast Cancer PatientsRecruitingMetastatic breast cancerPembrolizumab + Stereotactic Radiosurgeryn/a41Tumor response for non-irradiated brain lesionsNumber of participants with abscopal response
Brain metastases Correlation of abscopal responses with the radiation dose received
Overall survival
NCT04245514Multimodality Treatment in Stage III Non-small Cell Lung Cancer (NSCLC)RecruitingNon-Small-Cell Lung Cancer (NSCLC)Durvalumab + RT40 Gy, 20 fr90Event-free survivalRecurrence-free survival after resection
Overall survival
25 Gy, 5 frObjective response
Pathological complete response
24 Gy, 3 frMajor pathological response
Complete resection
NCT04530708Addition of Radiotherapy to Standard Medical Treatment for Stage IV NSCLC (MARS)RecruitingNon-Small-Cell Lung Cancer (NSCLC)Thoracic RT vs. standard of care36 Gy162Difference in quality of lifeOverall survival
Progression free survival
Toxicity of esophagitis, pneumonitis, dyspnea, fatigue, cough
NCT04212026Irreversible Electroporation (IRE) Followed by Nivolumab in Patients With Metastatic Pancreatic Cancer.RecruitingMetastatic pancreatic cancerNivolumabn/a15Overall response rate (ORR) of the reference liver metastasisORR of primary tumor site (pancreas)
ORR of IRE-treated liver metastasis
Progression free survival (PFS)
Overall survival
Adverse events
NCT03474497IL-2, Radiotherapy, and Pembrolizumab in Patients Refractory to Checkpoint BlockadeRecruitingNSCLC, Head and neck squamous cell carcinoma,IL-2 + Pembrolizumab + RT24 Gy,
3 fr, palliative regimen
45Abscopal response rateMaximum tolerated dose
Metastatic melanoma
Metastatic RCC
NCT03548428Stereotaxic Body Irradiation of Oligometastase in Sarcoma (Stereosarc)RecruitingSarcomaAtezolizumab + SBRT3 to 5 fr depending on tumor size103Progression-free survival (PFS) ratePFS by immune response criteria
Ratio PFS after RT/PFS during previous treatment
Objective response rate
Toxicity of treatment
Overall survival
Quality of life
Evaluation of the cost of treatment
Rate of PET-CT at inclusion
Impact of biomarkers on PFS or response rate
Developing mathematical models
NCT03927898Phase II Study of Toripalimab Plus Stereotactic Body Radiotherapy in Colorectal Cancer Patients With OligometastasisRecruitingMetastatic colorectal cancerToripalimab + SBRTSBRT (BED >80Gy) to oligometastatic lesions401 year progression free survival (PFS)Acute adverse events
Objective response rate
2 year local control rate
2 year overall survival
T cell receptor repertoire/T cell clones in blood
Expression of PD-1, Ki-67 on T cell
Expression of PD-L1 on exosomes in blood
Expression of PD-L1 on circulation tumor cell
NCT03774732PD-1 Inhibitors and Chemotherapy With Concurrent Irradiation at Varied Tumour Sites in Advanced Non-small Cell Lung Cancer (NIRVANA-LUNG)RecruitingNon-Small-Cell Lung Cancer (NSCLC)Pembrolizumab + Chemotherapy + Radiotherapyat least 18 Gy in 3 fr460Overall survivalTumour response
Progression free survival
Local and distant controls in irradiated patients
Pembrolizumab + ChemotherapyQuality of life
Acute/late toxicities
NCT04299646Study Assessing Stereotactic Radiotherapy in Therapeutic Strategy of Oligoprogressive Renal Cell Carcinoma Metastases (GETUG-StORM-01)RecruitingMetastatic RCCStereotactic RT + systemic treatmentn/a114Progression free survivalTreatment-related adverse events
Local control rate
Overall control rate
NCT03316872Study of Pembrolizumab and Radiotherapy in Liver CancerRecruitingHepatocellular carcinomaPembrolizumab + SBRTn/a30Overall response rateResponse rate in non-irradiated tumor lesions
Progression free survival rate
Overall survival rate
NCT03277482Durvalumab, Tremelimumab + Radiotherapy in Gynecologic CancerRecruitingRecurrent gyneco-logical cancerDurvalumab + Tremelimumab + RTn/a32Maximum tolerated doseOverall response rate
Metastatic cervical cancerLocal response rate
Metastatic ovarian cancerAbscopal response rate
Metastatic vaginal cancerResponse duration
Metastatic vulvar cancerProgression free survival
Metastatic endometrial cancer
NCT03085719Targeting PD-1 Therapy Resistance With Focused High or High and Low Dose Radiation in SCCHNRecruitingHead and neck cancerHigh dose radiation + Pembrolizumabn/a26Overall response rateOverall survival
Progression free survival
Treatment-related adverse events
Immune-related response
Local response
Clinical benefit rate
Abscopal response
NCT03176173Radical-Dose Image Guided Radiation Therapy in Treating Patients With Metastatic Non-small Cell Lung Cancer Undergoing ImmunotherapyRecruitingNon-Small-Cell Lung Cancer (NSCLC)Immunotherapy + Image Guided RTn/a85Progression free survivalChange in ctDNA levels
Immune marker levels from peripheral blood
Acute and late toxicity
Overall survival
Patterns of response and progression
Time to discontinuation of study
NCT04221893Radiation Therapy for the Treatment of Metastatic Gastrointestinal CancersRecruitingEsophageal adenocarcinomaRTn/a28Overall response rateProgression free survival
Esophageal squamous cell carcinomaOverall survival
Determine local control in radiated lesion(s)
Gastric cancerTumor measurement change
Adenocarcinoma of gastroesophageal junctionNew metastatic lesions
Adverse events
NCT03385226A Trial Assessing the Effect of Pembrolizumab Combined With Radiotherapy in Patients With Relapsed, Refractory, Specified Stages of Cutaneous T-cell Lymphoma (CTCL) Mycosis Fungoides (MF)/Sezary Syndrome (SS) (PORT)RecruitingCutaneous T cell lymphomaPembrolizumab + RT12 Gy, 3 fr46Overall response (global assessment)Response and duration
Safety and toxicity
Progression free survival
Overall survival
Abscopal effect rate
Changes in immune status
Plasma HMGB-1 levels
Functional analysis of isolated cell populations
Assessment of diversity and clonality of T cells
Immune signatures for responders and non-responders
Tumor-infiltrating lymphocyte-specific neo-antigens
Expression of immunological checkpoints
Investigation of tumor immune microenvironment
NCT02858869Pembrolizumab and Stereotactic Radiosurgery for Melanoma or Non-Small Cell Lung Cancer Brain MetastasesRecruitingMetastatic malignantPembrolizumab + Stereotactic RadiosurgerySRS 6 Gy,
9 Gy or
18–21 Gy
30Proportion of dose-limiting toxicitiesAbsolute cell counts for pre and post-treatment serum immune biomarkers
Neoplasm in the brain
Metastatic melanomaOverall response
Mucosal melanomaOverall survival
Ocular melanomaRate of anywhere intra-cranial failure
Non-Small Cell Lung CancerRate of leptomeningeal disease
Skin melanomaRate of local recurrence
Melanoma of unknown primaryRate of symptomatic radiation necrosis
NCT02830594Pembrolizumab and Palliative Radiation Therapy in Treating Patients With Metastatic Esophagus, Stomach, or Gastroesophageal Junction CancerActive,
not recruiting
Gastric and esophageal adeno-carcinomaEBRT + Pembrolizumabn/a14Tumor-infiltrating cytotoxic T cells and circulating cytotoxic T cellsIncidence of adverse events
Gastric and esophageal squamous cell carcinomaImmunosuppressive Tregs in non-irradiated sitesOverall response rate
Gastroesophageal junction adeno-carcinomaMDSCs in non-irradiated sitesProgression free survival
Metastatic malignant neoplasm in the stomachOverall survival
NCT03469713Nivolumab Plus Stereotactic Body Radiotherapy in II and III Line of Patients With Metastatic Renal Cell Carcinoma (NIVES)Active,
not recruiting
Metastatic renal cancerNivolumab + SBRT30 Gy in 3 consecutive fractions69Objective response rate (ORR)Progression free survival
Overall survival
ORR of irradiated and non-irradiated metastases and duration of response
Incidence, nature and severity of adverse event
Analysis of expression of PD-L1
Analysis of the genetic background of tumor
Analysis of the immuno-modulation
Identification of somatic mutations associated with acquired resistance to checkpoint inhibitors
NCT03453892Investigation of the Timely-coordinated Therapy of Patients With Metastatic Cancer by Radiotherapy Together With Immune Checkpoint Inhibition (ST-ICI)Active,
not recruiting
Metastatic cancerRT + Ipilimumabn/a150Systemic and local response of detected metastasesDetection of adverse events
Documentation of corticoid prescription
Nivolumab + Pembrolizumab + RTChange of circulating immune cells by deep immunophenotypingOverall survival
Progression free survival
NCT03322384Phase I/II Trial of Epacadostat, Intralesional SD101, Radiotherapy in Patients With LymphomaActive,
not recruiting
Advanced solid tumorsEpacadostat + SD-101 + RT24 Gy, 3 fr20Maximum tolerated doseAbscopal response rate
Lymphoma20 Gy, 5 frOncidence of treatment-emergent adverse events
NCT03539198Study of Proton SBRT and Immunotherapy for Recurrent/Progressive Locoregional or Metastatic Head and Neck CancerActive,
not recruiting
Head and neck cancerProton SBRT + Nivolumab35–45 Gy,
5 fr
91Objective response rateLocal control rate, overall survival
Progression free survival
Time to progression
New development of distant metastasis
Quality of Life
Predictive and prognostic biomarker
NCT03042156Immunotherapy And Palliative Radiotherapy Combined In Patients With Advanced MalignancyActive,
not recruiting
Advanced cancerPalliative RTn/a30Number of patients showing toxicityIn-field response on imaging and evidence of out of field (abscopal) response
Biomarkers analyses as indicator of abscopal response
Patient-reported outcome
Inflammatory and radiation sensitivity signatures
NCT03483012Atezolizumab + Stereotactic Radiation in Triple-negative Breast Cancer and Brain MetastasisActive,
not recruiting
Breast cancerAtezolizumab + stereotactic radiosurgery (SRS)n/a45Progression free survivalExtracranial objective response rate
Abscopal response rate
Clinical benefit rate
Overall survival
Patient-reported outcome
Development of radiation necrosis
Assessed neurological evaluation
Dose-limiting toxicity
NCT02888743Durvalumab and Tremelimumab With or Without High or Low-Dose Radiation Therapy in Treating Patients With Metastatic Colorectal or Non-small Cell Lung CancerActive,
not recruiting
Metastatic colorectalTremelimumab + Durvalumabn/a180Overall response rateProgression free survival
Overall survival
Objective response
Carcinoma metastatic lungIncidence of adverse events
Tremelimumab + Durvalumab + RTLocal control rate and abscopal response rates
Non-Small Cell CarcinomaPrognostic effect of PD-L1 expression
Prognostic effect of T-cell infiltration
Symptomatic adverse events
NCT02587455Pembrolizumab and Palliative Radiotherapy in Lung (PEAR)Active,
not recruiting
Thoracic tumoursPembrolizumab + RTn/a48Toxicity rateProgression free survival rates
Overall survival rates
Duration of clinical benefit
Maximum tolerated doseResponse rates
Assessing individual lesion response
Identify biomarkers and correlate with clinical benefit
NCT03601455Radiation Therapy and Durvalumab With or Without Tremelimumab in Treating Participants With Unresectable, Locally Advanced, or Metastatic Bladder CancerActive,
not recruiting
Bladder urothelial carcinomaRT + Durvalumabn/a13Incidence of adverse eventsLocal control at primary irradiate site
Pathologic complete rate of irradiated tumor
Overall response rate
Abscopal response
Duration of response
Disease-specific survival
RT + Durvalumab + TremelimumabProgression free survivalOverall survival
Incidence of adverse events
Immune cell subsets and PD-L1 in tumor biopsies
Gene signature biomarker
Circulating immune cell subsets
Circulating and tumor-infiltrating T-cell receptor repertoire
Abbreviations: RT: radiotherapy; NSCLC: Non-Small-Cell Lung Cancer; fr: fractions; SBRT: stereotactic body radiotherapy; n/a: not available; ORR: overall response rate; IRE: irreversible electroporation; PFS: progression free survival; RCC: renal cell carcinoma; PET-CT: positron emission tomography; CT: computed tomography; BED: radiation biologically effective dose; PD-1: programmed cell death protein 1; PD-L1: PD-1 receptor-ligand 1; ctDNA: circulating tumor DNA; HMGB1: high-mobility group box 1; SRS: stereotactic radiosurgery.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Link, B.; Torres Crigna, A.; Hölzel, M.; Giordano, F.A.; Golubnitschaja, O. Abscopal Effects in Metastatic Cancer: Is a Predictive Approach Possible to Improve Individual Outcomes? J. Clin. Med. 2021, 10, 5124. https://doi.org/10.3390/jcm10215124

AMA Style

Link B, Torres Crigna A, Hölzel M, Giordano FA, Golubnitschaja O. Abscopal Effects in Metastatic Cancer: Is a Predictive Approach Possible to Improve Individual Outcomes? Journal of Clinical Medicine. 2021; 10(21):5124. https://doi.org/10.3390/jcm10215124

Chicago/Turabian Style

Link, Barbara, Adriana Torres Crigna, Michael Hölzel, Frank A. Giordano, and Olga Golubnitschaja. 2021. "Abscopal Effects in Metastatic Cancer: Is a Predictive Approach Possible to Improve Individual Outcomes?" Journal of Clinical Medicine 10, no. 21: 5124. https://doi.org/10.3390/jcm10215124

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