Next Article in Journal
Meta-Analysis and Validation of a Colorectal Cancer Risk Prediction Model Using Deep Sequenced Fecal Metagenomes
Next Article in Special Issue
Alcohol as a Non-UV Social-Environmental Risk Factor for Melanoma
Previous Article in Journal
Genomic Landscape Alterations in Primary Tumor and Matched Lymph Node Metastasis in Hormone-Naïve Prostate Cancer Patients
Previous Article in Special Issue
Zebrafish Syndromic Albinism Models as Tools for Understanding and Treating Pigment Cell Disease in Humans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Abscopal Response in Metastatic Melanoma: Real-World Data of a Retrospective, Multicenter Study

by
Luc Ollivier
1,*,
Charles Orione
2,
Paul Bore
3,
Laurent Misery
4,5,
Delphine Legoupil
4,5,
Jean-Christophe Leclere
6,
Anne Coste
7,
Gilles Girault
8,
Iona Sicard-Cras
9,
Clemence Kacperek
10,
Francois Lucia
11,
Dinu Stefan
9,
François Thillays
1,
Emmanuel Rio
1,
Paul Lesueur
8,12,
Christian Berthou
13,
Dominique Heymann
14,15,
Stéphane Champiat
16,
Stéphane Supiot
1,
Loig Vaugier
1 and
William Kao
8
add Show full author list remove Hide full author list
1
Department of Radiation Oncology, Institut de Cancérologie de l’Ouest (ICO), 44800 Saint-Herblain, France
2
Département de Médecine Interne et Pneumologie, Centre Hospitalo-Universitaire de Brest, Université de Bretagne Occidentale, 29609 Brest, France
3
Oncology Department, Institut de Cancérologie de l’Ouest (ICO), 44800 Saint-Herblain, France
4
Service de Dermatologie, CHU, 2, Avenue Foch, 29200 Brest, France
5
Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale, 29200 Brest, France
6
Service d’ORL et de Chirurgie Cervico-Faciale, Hôpital Morvan, Centre Hospitalo-Universitaire de Brest, Université de Bretagne Occidentale, 5, Avenue Foch, 29200 Brest, France
7
Département de Maladie Infectieuses, Centre Hospitalo-Universitaire de Brest, Université de Bretagne Occidentale, 29609 Brest, France
8
Unicancer, Comprehensive Cancer Center F. Baclesse, Medical Library, F-14000 Caen, France
9
Service de Pédiatrie, Hôpital Morvan, Centre Hospitalo-Universitaire de Brest, Université de Bretagne Occidentale, 5, Avenue Foch, 29200 Brest, France
10
Département of Radiation Oncology, Centre Hospitalier de Cornouaille, 14 Avenue Yves Thepot, 29000 Quimper, France
11
Radiation Oncology Department, Centre Hospitalo-Universitaire de Brest, Université de Bretagne Occidentale-Hôpital Morvan-2 Avenue Foch, 29200 Brest, France
12
Centre Guillaume le Conquérant, Radiation Oncology Department, 76600 Le Havre, France
13
U1227 B Lymphocytes & Autoimmunity, University of Brest, INSERM, IBSAM, 29200 Brest, France
14
Nantes Université, CNRS, UMR6286, US2B, 44300 Nantes, France
15
Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
16
Département d’Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(17), 4213; https://doi.org/10.3390/cancers14174213
Submission received: 6 June 2022 / Revised: 2 August 2022 / Accepted: 11 August 2022 / Published: 30 August 2022
(This article belongs to the Special Issue Study on the Complex Melanoma)

Abstract

:

Simple Summary

An abscopal response (AR) is a rare phenomenon defined as a distant response outside of the radiation field. It opens up the perspective of “in situ” vaccination of cancer. This phenomenon is rare and its mechanisms are unknown. In metastatic melanoma (MM), the approach regarding the efficacy of immunotherapy is to not use immunotherapy as a tool for enhancing radiation response but rather as one that needs to be integrated into immunotherapy to potentiate the specific effects of immunotherapy. The aim of our retrospective study was to investigate the incidence of the AR and its impact on therapeutic outcomes in a homogeneous population of patients with MM and a control group, to identify the factors associated with the AR. AR in metastatic melanoma seems highly prognostic of overall survival although it is a rare phenomenon. Factors associated with AR have been identified.

Abstract

Objective: To evaluate the incidence of the abscopal response (AR) in patients with metastatic melanoma requiring palliative radiotherapy (RT). Patients and methods: Patients treated for metastatic melanoma between January 1998 and February 2020 in four oncology departments were screened. Patients with progression under immune checkpoint inhibitors or without ongoing systemic treatment, and requiring palliative RT were considered. The AR was defined as an objective response according to RECIST and/or iRECIST for at least one non-irradiated metastasis at distance (≥10 cm) from the irradiated lesion. Primary endpoint was the rate of AR. Secondary endpoints were overall survival (OS), progression-free survival (PFS), local control (LC) of the irradiated lesion, and toxicity as assessed by CTCAE v5. Results: Over the period considered, 118 patients were included and analyzed. Fifteen patients (12.7%) had an AR. With a median follow-up of 7.7 months (range, 0.2–242.2), median OS and PFS after RT were significantly longer in patients with an AR compared to those without: 28 vs. 6.6 months (p < 0.01) and not reached vs. 3.2 months, respectively. No grade ≥2 toxicity was reported. Patients who developed an AR were more likely to be treated with immunotherapy (93.3% vs. 55.9%, p = 0.02). In multivariate analysis, they had a higher number of irradiated metastases treated concomitantly (HR = 16.9, p < 0.01) and a higher rate of mild infections during RT (HR = 403.5, p < 0.01). Conclusions: AR in metastatic melanoma seems to be highly prognostic of overall survival, although it is a rare phenomenon. It may be promoted by multiple concomitant treatments with RT and immunotherapy and by acute inflammatory events such as infection.

1. Introduction

Radiotherapy can be used to convert a tumor into an “in situ’’ vaccine [1]. The mechanisms of this concept are related to the immune system: when focal radiation therapy is applied to the tumor and the tumor microenvironment, it induces immunogenic cell death, a type of cell death which can be recognized by the immune system. This process can generate an immune response that contributes not only to the in-field response, but also to the out of field response and in occasional situations it has been described as an abscopal response. However, the abscopal effect is very rare because tumors are established, with metastases and many mechanisms of immune evasion, and it is extremely difficult for radiotherapy to one site to induce an immune response. However, in combination with immunotherapy, the situation can be different [2]. The problem is shifting the balance of the mixed signals from immunosuppressive to pro-immunogenic and trying to either mitigate the negative effect of radiation or enhance the positive effect of radiation [3]. This challenge has been explored in numerous preclinical models, with some translated to clinical practice. In metastatic melanoma, the approach regarding the efficacy of immunotherapy is to not use immunotherapy as a tool for enhancing the radiation response, but rather as a tool that needs to be integrated into immunotherapy to potentiate the specific effects of immunotherapy. With the emergence of immune checkpoint inhibitors of programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA 4), the 5-year overall survival (OS) rate of patients with metastatic melanoma (MM) has dramatically improved in the last decade, reaching 50%, as well as with 20% of long responder patients [4,5,6]. Large tumor mutational burden and/or the immune-mediated patient-dependent environment may partly explain such significant outcomes.
Simultaneously, the interest in one of the holy grails of radiation oncology was reborn. Does the abscopal response (AR) exist, i.e., could the local radiation-induced tumor response generate a distant, out-of-field tumor response? Since the first description of the AR in 1953 by Dr. RJ Mole [7], only 46 cases of AR have been reported between 1953 and 2016 [8] and 24 cases in combination with immunotherapy [9]. A patient-level data meta-analysis for the predictors of response was performed in order to report data on progression free-survival (PFS), distant metastases, and overall survival with the hypothesis that certain clinical covariates may predict the survival of patients with an abscopal response. Among 67% of abscopal responses in non-squamous cell lung cancer, kidney cancer, melanoma, lymphoma, and hepatobiliary cancer, there were no clinical predictors of the duration of response or survival, as well as a lack of control group [10]. Additionally, many trials have been conducted to reproduce this phenomenon with disappointing results. Nowadays, the AR remains one of the most active areas of research in oncology [11]. Great interest is given to the mechanisms and trigger factors relative to the AR, especially regarding specific radiotherapy (RT) modalities.
MM is recognized as one of the most immune-sensitive oncologic diseases and patients with MM may thus be the most likely to show an AR [12]. The aim of our study was to investigate the incidence of AR and its impact on therapeutic outcomes in a homogeneous cohort of patients with MM. Factors associated with AR were also identified.

2. Patients and Methods

2.1. Patient Population

Retrospective data extraction was performed between January 1998 and February 2020 at the University Hospital in Brest (France), the Regional Hospital in Quimper (France), the Francois Baclesse Cancer Center in Caen (France), and the Institut de Cancérologie de l’Ouest (Saint-Herblain, France). All consecutive patients with MM and requiring palliative RT were considered. Inclusion criteria were: histologically proven MM, disease progression under immunotherapy according to the Immune Response Evaluation Criteria in Solid Tumors (iRECIST) or disease progression without systemic treatment (i.e., patients unfit for immunotherapy), RT for at least one primary or metastatic site, and the presence of at least one non-irradiated metastasis which was (i) ≥10 cm away from the irradiated target volume and (ii) assessable according to iRECIST criteria. A cut-off of 10 cm was arbitrarily set to avoid biases arising from potential low-dose radiation responses. Exclusion criteria were: initiation of a new systemic treatment, irradiation of brain metastases, and previous or concomitant treatment with BRAF inhibitors.
All RT schedules were accepted. The RT-target irradiation volume was defined as the planning target volume (PTV) covered by at least 95% of the prescribed dose. The following patient characteristics were recorded at the beginning of the RT: Eastern Cooperative Oncology Group Performance Status (PS), American Joint Committee on Cancer 8th edition (AJCC 8th) TNM [13], RT total dose, RT dose per fraction, RT duration, the localization of target volumes, number of irradiated sites treated simultaneously, modalities and date of the first day of the last systemic treatment, the continuation of the last systemic treatment, and primary and secondary resistance status. Primary resistance was defined as the absence of clinical response after initial PD-1/PD-L1 blockade. Secondary resistance refers to progression while on therapy despite an initial response, or unresponsive to re-initiation of checkpoint blockade with the need for local symptomatic treatment such as RT [14].
Individual immune-related events (irE) were defined as acute events occurring during RT only: documented infectious episodes, clinical symptoms of acute inflammation (redness, heat, swelling, pain), and/or biological inflammatory syndrome with elevation of inflammation parameters including at least C-reactive protein (CRP) > 30 mg/L). Clinical acute inflammation was classified as (i) light clinical inflammation and CRP <40 mg/L, (ii) moderate clinical inflammation and CRP <40 mg/L, and (iii) severe clinical inflammation and CRP >50 mg/L [15].
Usual follow-up after RT in participating centers is performed by an onco-dermatologist every 3 months and a radiation-oncologist every 6 months. Clinical examination, blood samples, and either a computed tomography (CT) or a 18F-Fluorodeoxyglucose positron emission tomography with CT (18F-FDG-PET/CT) are performed.

2.2. Outcome

The primary endpoint was the rate of patients developing an AR following RT. An AR was defined as an objective response rate for at least one non-irradiated metastasis, namely, either complete response (CR) or partial response (PR) according to iRECIST [16,17]. All patients with a suspected AR were confirmed with a double reading by two radiation oncologists. Secondary outcomes were local control (LC) of the irradiated lesion, overall survival (OS), progression-free survival (PFS), and toxicity as assessed by the Common Terminology Criteria for Adverse Events version 5 (CTCAE v5) classification [18]. LC was defined as complete response (CR) or partial response (PR) in the irradiated lesion as per iRECIST. In specific cases of new or re-growth of existing lesions within or at the margin of the PTV, the lesion was considered locally progressive. If progression was confirmed at the next assessment, the date of progression assigned was the earlier date when progression was first suspected. Distant progression was defined as the appearance of new lesions outside the PTV.

2.3. Statistical Analysis

The chi-square test or Fisher’s exact test was used to analyze and compare patients’ clinical characteristics at baseline between groups for categorical variables. Student t-test or the Wilcoxon test was used for continuous variables, according to their distributions. PFS and OS were defined as the time between the first day of RT and either local, regional, or distant progression or death, respectively. Survival (PFS, OS) was described by means of Kaplan–Meier curves and compared using log-rank tests in univariate analysis.
OS and PFS were analyzed with a multivariable Cox model considering AR as a time-dependent binary variable, since its value changed over time among patients who experienced an abscopal effect.
A secondary analysis used another Cox model to identify risk factors for developing AR over time. For the dosimetric variables, the ROC curve was built to identify the best value associated with the occurrence of AR within the first 30 days after the beginning of RT.
For each variable and each model, the proportional hazard hypothesis and the log-linearity for continuous variables were assessed using Schoenfeld residuals and graphical visualization, respectively.
Statistical analyses were performed using R software-© 2009–2019 RStudio, Inc.

3. Results

Overall, 118 patients treated with RT for melanoma between January 1998 and February 2020 in four French oncology institutions were included. Median age at the beginning of RT was 66.5 (range, 23.5–100.1). PS was ≤1 in 70.4% patients (Table 1).

3.1. Population Characteristics

Sixty-seven percent of patients (n = 80) received concomitant antiPD1 +/− antiCTLA4 immunotherapy during RT, including nivolumab (n = 47), pembrolizumab (n = 16), ipilimumab (n = 11), and nivolumab + ipilimumab (n = 6). Median time between the first injection of immunotherapy and RT was 2.9 months (range, 0.5–47 months). Patients had primary and secondary resistance to immunotherapy before RT started in 42.5% and 57.5% of cases, respectively (Table 1). The median delivered dose of RT was 30 Gy (range, 6.5–60 Gy). Median dose per fraction was 4 Gy (range, 2–20 Gy). Fifteen patients were treated with stereotactic body RT (SBRT) and the median dose per fraction was 9 Gy (range, 8–20). Median duration of RT was 10 days (range, 1–63). Twenty-two patients (18.6%) had ≥2 simultaneously irradiated metastases. The metastatic sites irradiated were bone lesions (37.3% of patients), lymph nodes (23.7%), and skin metastases (19.5%). Conformal RT with linear accelerators was used for all patients. No cases of severe grade 2+ acute or late toxicity were reported. IrE occurred in 16 patients (13.5%) during RT and were mainly mild (Table 2).

3.2. Survival Analysis

An AR was observed after RT in 15 patients (12.7%) after a median time following RT of 1.7 months (range, 0.2–3.6) and persisted over a median time of 11.9 months (range, 4.2–68.4) (Table 2). With median follow-up of 7.7 months (range, 0.2–242.2), LC or stable disease for the irradiated lesions was obtained in 103 patients (87.3%) (Table 3). Median PFS after RT was 3.6 months (95% CI: 2.7–4.4). Median PFS was 3.2 months for patients without AR, whereas it was not reached in cases of AR (Figure 1). In multivariate analyses, the occurrence of AR was associated with increased PFS (p < 0.01) (Table 4). Median OS after RT was 8.5 months (95% CI: 4.3–12.6).
Among the variables tested in multivariate analysis, the occurrence of an AR (HR = 0.19, p = 0.007) and concomitant immunotherapy (HR = 0.51, p = 0.01) significantly improved OS (Table 5), whereas a poor performance status (WHO 2–3) was negatively correlated with OS (p < 0.01). OS was significantly longer in patients who received immunotherapy [12.6 (95%CI: 5.8–19.3) versus 2.9 months (95%CI: 1.1–4.8)] (Figure 2) and in those who developed an AR compared to those who did not (28 months (95% CI: 27.4–28.7) versus 6.6 months (95% CI: 4.2–8.9), (p < 0.01)) (Figure 3).

3.3. Abscopal Risk Factors

In multivariate analysis, multiple (≥2) irradiated metastases (HR 16.85 (95% CI 2.16–131.49) p < 0.01) and concomitant irE (HR 403.45 (95% CI 13.83–11769.39), p < 0.01) were significantly associated with the occurrence of an AR (Table 6).
For dosimetric parameters, the optimal values associated with a higher chance of developing an abscopal response at 30 days was estimated at 19 Gy (AUC = 0.706) delivered in 3.5 fractions (AUC = 0.779) of 5.25 Gy (AUC = 0.796) over 4.5 days (AUC = 0.849) (Table 7,Figure 4).

4. Discussion

In our multicenter cohort including 118 patients with MM treated with palliative RT, 15 patients (12.7%) developed an AR. In comparison, other studies found evidence for AR in 34.3% (range, 18–63%) of cases in an analysis of 10 nonrandomized studies of metastatic melanoma patients treated with RT and ICI between 2014 and 2019 [19]. Our study is of moderate size compared to others given the eligibility criteria and the strict definition of an abscopal response.
The occurrence of AR in our study was associated with significantly improved oncologic outcomes, including OS rates in multivariate analysis. By definition, an AR correlates with effective distant control; interestingly, it also resulted in better OS rates. Our results are in accordance with the literature: in a prospective study that aimed to induce an abscopal effect in 41 stable or progressing patients treated with systemic treatment for solid tumors, the median OS was 20.98 months (95% CI 11.05–30.96) in abscopal responders versus 8.33 months in others (95% CI 5.03–13.29). This study, however, did not include patients with metastatic melanoma, but mostly lung and breast cancer [20].
With a median follow-up of 7.7 months, median OS (10.2 months) after the occurrence of antiPD1 +/− antiCTLA4 immunotherapy resistance was poor in our cohort, which is in agreement with the literature [21]. Immunotherapy was continued concomitantly with RT in 63% of patients despite progression under such treatment. Although this attitude may appear contradictory, it is often justified by i) the addition of RT for the most aggressive or painful lesions, ii) the possibility of a delayed response under immunotherapy, and iii) the potential synergy between immunotherapy and RT [22]. Patients treated with RT and immunotherapy had significantly better survival outcomes compared to RT alone, with a median OS of 12.6 months (95%CI: 5.8–19.3) versus 2.9 months (95%CI: 1.1–4.8) (p < 0.01). These observations—also coherent with the literature—corroborate such a frequently adopted strategy [22]. The use of ipilimumab [23] has been associated with the AR, but the immunotherapy sequence with respect to RT needs to be discussed in further prospective studies to promote an abscopal response. The administration of antiPD1 before irradiation seems to abrogate systemic immunity and increase the radiosensitivity of CD8+ T cells, while tumor control is improved when antiPD1 is given after RT [24].
Such AR-associated improved outcomes justify investigating how AR might be provoked. The interplay between immune environment and AR has already been described [25] and numerous protocols have already been employed, such as the use of granulocyte-macrophage colony-stimulating factor (GM-CSF) concomitantly with RT and chemo- or hormonotherapy, SBRT, and immunotherapy [20,26]. In the present study, the occurrence of mild infections was observed more frequently in patients with an AR. To our knowledge, the potential role of infection-driven inflammation as an AR-promoting factor has never been described. Polynuclear neutrophils—whose recruitment and type may have significant antitumor activity as shown in the proof-of-principle trial of GM-CSF and radiotherapy [20,27,28]—could be part of the explanation. However, previous studies (but not in cases of MM) have shown an inverse relationship between RT efficacy and hyperleukocytosis [29]. The role of neutrophils with RT thus remains controversial [30]. One possible explanation may be the induction by RT of type I interferon (IFN-I), which is a critical factor. In irradiated tumors and viral infections, IFN-I is induced by cytosolic DNA that stimulates cGAS to produce cGAMP, leading to STING activation. IFN-I activates conventional dendritic type 1 cells that cross-present the antigens to CD8 T-cells to eliminate the infected or tumoral cells [31].
Activation of the interferon pathway may also come from the dose of radiotherapy and fractionation dose. In fact, in preclinical data regarding 8 Gy, there is more induction of IFN than at higher doses, such as 20 Gy or 30 Gy. Repeating the dose of 8 Gy three times seems to be the optimal formula for activating a type I IFN response and upregulates the expression of MHC-I. [32,33]. At higher doses, further stimulation of DNA damage leads to negative feedback expression of 3-prime repair endonuclease 1 (TREX1), which digests cytosolic DNA and thus reduces the activation of the IFN I response [34]. In a subset analysis of a multicenter clinical trial with matched biopsied pre/post RT plus pembrolizumab, a complete response and partial response were found in patients with a low level of TREX1 and a high level of DNASE1, which is the opposite of TREX1 [35]. In our study, optimal dosimetric parameters for inducing an abscopal response at 30 days may be 19 Gy delivered in 3.5 fractions of 5.25 Gy over 4.5 days. This analysis should be accepted with caution given the small number of patients and the heterogeneity in practices. However, it seems to concur with preclinical findings where hypo-fractionated RT was superior to a single dose of RT in promoting an antitumor immune response with a dose/fraction between 5–7.5 Gy [36] and a number of fractions between 3 and 5 [31].
Among other agents involved in the immune response against external injuries, lymphocytes are less likely to be involved as they are known to be radiosensitive and thus strongly depleted within radiation fields. This understanding has led to the idea that blood should be considered an organ at risk because RT-induced lymphopenia is associated with poorer survival in multiple tumor types [37]. Lung cancer radiotherapy with 60 Gy/30 fractions exposes 99% of circulating blood to more than 0.5 Gy [38] and that has an impact on the efficacy of the patient’s immune system and seems to be associated with low survival. Then, when combining radiotherapy with immunotherapy, it seems to be preferable to use hypo-fractionation, a small field size, and the integral dose.
In contrast, dendritic cells, while particularly stimulated during infections [39], are capable of surviving common radiation doses, making them potential vectors for the distance antitumor response of the RT targets [40,41].
All these hypotheses merit further exploration but are currently not robust enough to be reproducible and need to be documented as accurately as possible. Infection-driven immune-related events can barely be distinguished from treatment-related immune events. As our study was retrospective, the parameters used to monitor inflammation have limitations. Although CRP has relatively good sensitivity for detecting inflammatory processes, it remains non-specific. Concentrations of 10–40 mg/l are generally found in cases of moderate inflammation or viral infections, and typically rise to 50–200 mg/l in cases of severe inflammation or bacterial infections [15], without any clear cut-off [42]. Small increases, between 3 and 10 mg/l, are also found in cases of obesity, smoking, diabetes, and high blood pressure [43]. It is nevertheless interesting to note that in current practice, any infection—from mild to severe—causes RT delay because of the potentially harmful effects of radiotherapy in this context [44], whereas in our study, such events did not seem to induce any increased toxicity and may have even promoted an AR with RT.
A number of irradiated sites ≥ 2 at the same time also appeared to be a favorable factor for the AR in our study, as suggested in the literature [45], but not the type of irradiated anatomical structure. Amplifying the number of tumor antigens and the improved response to immunotherapy following multisite RT has already been reported [46]. In this study, all patients were metastatic, meaning that multi-site irradiation would make possible a higher probability of recognition of neo-antigens in the drainage nodes. In cases of primary tumors, it has been proposed that the draining node should be excluded from the radiation field and only the primary tumor should be irradiated [47], with the hypothesis of being immunized against the primary and obtaining a positive node response. This raises the question of excluding drainage nodes when radiotherapy is performed in combination with checkpoint inhibitors. This is relevant in head and neck cancer regarding the negative results of the JAVELIN trial, in which avelumab plus the standard of care chemoradiotherapy versus chemoradiotherapy alone did not improve PFS [48]. To investigate the importance of lymph nodes, preclinical data show that in immune competent mice with tongue tumors, receiving antiCTLA4 reduces tumor volume, but in the case of sham surgery of the draining node, the effect of the antiCTLA4 is abrogated. Similarly, when the nodes are included in the radiation field treating the tongue tumor, the effect of antiCTLA 4 is abrogated [49].
Our study has several limitations, with a possible selection bias inherent to its retrospective design. First, clinicians do not always report infections occurring during radiation therapy. Second, the small proportion of AR patients makes the analysis less robust but remains consistent with the rarity of the phenomenon. Lastly, the follow-up modalities and retrospective data recording may have influenced the results of the analysis.

5. Conclusions

The abscopal response in metastatic melanoma patients seems highly prognostic. Although a rare phenomenon, it may be provoked with multiple concomitant radiation treatments with an acute immune-related event during radiotherapy. A biological substrate is urgently needed to corroborate our hypothesis.

Author Contributions

Conceptualization, L.O., P.B. and W.K.; methodology, C.O. and J.-C.L.; software, C.O. and J.-C.L.; validation, C.K., F.L., P.L. and S.S.; formal analysis, A.C. and L.V.; investigation, A.C., L.M., D.L., D.S., F.T., E.R. and E.R.; resources, G.G.; data curation, L.O. and C.O.; writing—original draft preparation, L.O., P.B., I.S.-C., S.C., D.H. and W.K.; writing—review and editing, all authors; visualization, L.O. and W.K.; supervision, S.S.; project administration, C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was approved by the Ethics Committee of the Centre Francois Baclesse in Caen, FR (CNIL-MR004: 2214228V0) and by the other centers consecutively.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AJCC 8thAmerican Joint Committee on Cancer 8th edition AJCC 8th
AUCArea Under the curve
ARAbscopal Response
CRcomplete response
CRPC-Reactive Protein
CTCAE v5Common Terminology Criteria for Adverse Events version 5
CTLA-4cytotoxic T lymphocyte antigen
EBRTexternal beam radiation therapy
18F-FDG-PET/CT18F-Fluorodeoxyglucose positron emission tomography with CT
HRhazard ratio
ICIimmune checkpoint inhibitors
LClocal control of the irradiated lesion
MMMetastatic melanoma
OSOverall survival
PD-1programmed cell death 1
PD-L1programmed cell death-ligand 1
PFSprogression-free survival
PTVplanning target volume
PRpartial response
RECISTResponse Evaluation Criteria in Solid Tumors
RTRadiotherapy
SBRTStereotactic body radiation therapy

References

  1. 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]
  2. Demaria, S.; Bhardwaj, N.; McBride, W.H.; Formenti, S.C. Combining radiotherapy and immunotherapy: A revived partnership. Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 655–666. [Google Scholar] [CrossRef]
  3. Formenti, S.C.; Demaria, S. Combining radiotherapy and cancer immunotherapy: A paradigm shift. J. Natl. Cancer Inst. 2013, 105, 256–265. [Google Scholar] [CrossRef]
  4. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef]
  5. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. Available online: https://www.nejm.org/doi/10.1056/NEJMoa1910836 (accessed on 24 August 2020). [CrossRef]
  6. 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]
  7. Mole, R.H. Whole body irradiation; radiobiology or medicine? Br. J. Radiol. 1953, 26, 234–241. [Google Scholar] [CrossRef]
  8. 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]
  9. 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]
  10. Hatten, S.J.; Lehrer, E.J.; Liao, J.; Sha, C.M.; Trifiletti, D.M.; Siva, S.; McBride, S.M.; Palma, D.; Holder, S.L.; Zaorsky, N.G. A Patient-Level Data Meta-analysis of the Abscopal Effect. Adv. Radiat. Oncol. 2022, 7, 100909. [Google Scholar] [CrossRef]
  11. 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]
  12. Update on Tumor Neoantigens and Their Utility: Why It Is Good to Be Different—PubMed [Internet]. Available online: https://pubmed.ncbi.nlm.nih.gov/29751996/ (accessed on 24 October 2020).
  13. Keung, E.Z.; Gershenwald, J.E. The eighth edition American Joint Committee on Cancer (AJCC) melanoma staging system: Implications for melanoma treatment and care. Expert Rev. Anticancer Ther. 2018, 18, 775–784. [Google Scholar] [CrossRef] [PubMed]
  14. Kluger, H.M.; Tawbi, H.A.; Ascierto, M.L.; Bowden, M.; Callahan, M.K.; Cha, E.; Chen, H.X.; Drake, C.G.; Feltquate, D.M.; Ferris, R.L.; et al. Defining tumor resistance to PD-1 pathway blockade: Recommendations from the first meeting of the SITC Immuno-therapy Resistance Taskforce. J. Immunother. Cancer 2020, 8, e000398. [Google Scholar] [CrossRef]
  15. Vermeire, S.; Van Assche, G.; Rutgeerts, P. Laboratory markers in IBD: Useful, magic, or unnecessary toys? Gut 2006, 55, 426–431. [Google Scholar] [CrossRef]
  16. Mulkey, F.; Theoret, M.R.; Keegan, P.; Pazdur, R.; Sridhara, R. Comparison of iRECIST versus RECIST V.1.1 in patients treated with an anti-PD-1 or PD-L1 antibody: Pooled FDA analysis. J. Immunother. Cancer 2020, 8, e000146. [Google Scholar] [CrossRef]
  17. Seymour, L.; Bogaerts, J.; Perrone, A.; Ford, R.; Schwartz, L.H.; Mandrekar, S.; Lin, N.U.; Litière, S.; Dancey, J.; Chen, A.; et al. iRECIST: Guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017, 18, e143–e152. [Google Scholar] [CrossRef]
  18. National Cancer Institute (US). Cancer Therapy Evaluation Program. In Common Terminology Criteria for Adverse Events (CTCAE); Cancer Therapy Evaluation Program: Rockville, MD, USA, 2017; p. 155. [Google Scholar]
  19. D’Andrea, M.A.; Reddy, G.K. Systemic Antitumor Effects and Abscopal Responses in Melanoma Patients Receiving Radiation Therapy. Oncology 2020, 98, 202–215. [Google Scholar] [CrossRef]
  20. 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]
  21. Ugurel, S.; Röhmel, J.; Ascierto, P.A.; Flaherty, K.T.; Grob, J.J.; Hauschild, A.; Larkin, J.; Long, G.V.; Lorigan, P.; McArthur, G.A.; et al. Survival of patients with advanced metastatic melanoma: The impact of novel therapies-update 2017. Eur. J. Cancer 2017, 83, 247–257. [Google Scholar] [CrossRef]
  22. Borcoman, E.; Kanjanapan, Y.; Champiat, S.; Kato, S.; Servois, V.; Kurzrock, R.; Goel, S.; Bedard, P.; Le Tourneau, C. Novel patterns of response under immunotherapy. Ann. Oncol. 2019, 30, 385–396. [Google Scholar] [CrossRef]
  23. Chicas-Sett, R.; Morales-Orue, I.; Rodriguez-Abreu, D.; Lara-Jimenez, P. Combining radiotherapy and ipilimumab induces clinically relevant radiation-induced abscopal effects in metastatic melanoma patients: A systematic review. Clin. Transl. Radiat. Oncol. 2017, 9, 5–11. [Google Scholar] [CrossRef] [PubMed]
  24. Wei, J.; Montalvo-Ortiz, W.; Yu, L.; Krasco, A.; Ebstein, S.; Cortez, C.; Lowy, I.; Murphy, A.J.; Sleeman, M.A.; Skokos, D. Sequence of αPD-1 relative to local tumor irradiation determines the induction of abscopal antitumor immune responses. Sci. Immunol. 2021, 6, eabg0117. [Google Scholar] [CrossRef]
  25. Lippert, T.P.; Greenberg, R.A. The abscopal effect: A sense of DNA damage is in the air. J. Clin. Investig. 2021, 131, 148274. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Souto, J.C.; Vila, L.; Brú, A. Polymorphonuclear neutrophils and cancer: Intense and sustained neutrophilia as a treatment against solid tumors. Med. Res. Rev. 2011, 31, 311–363. [Google Scholar] [CrossRef]
  28. Takeshima, T.; Pop, L.M.; Laine, A.; Iyengar, P.; Vitetta, E.S.; Hannan, R. Key role for neutrophils in radiation-induced antitumor immune responses: Potentiation with G-CSF. Proc. Natl. Acad. Sci. USA 2016, 113, 11300–11305. [Google Scholar] [CrossRef] [PubMed]
  29. Schernberg, A.; Blanchard, P.; Chargari, C.; Deutsch, E. Neutrophils, a candidate biomarker and target for radiation therapy? Acta Oncol. 2017, 56, 1522–1530. [Google Scholar] [CrossRef]
  30. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef]
  31. Demaria, S.; Guha, C.; Schoenfeld, J.; Morris, Z.; Monjazeb, A.; Sikora, A.; Crittenden, M.; Shiao, S.; Khleif, S.; Gupta, S.; et al. Radiation dose and fraction in immunotherapy: One-size regimen does not fit all settings, so how does one choose? J. Immunother. Cancer 2021, 9, e002038. [Google Scholar] [CrossRef]
  32. Yamazaki, T.; Kirchmair, A.; Sato, A.; Buqué, A.; Rybstein, M.; Petroni, G.; Bloy, N.; Finotello, F.; Stafford, L.; Manzano, E.N.; et al. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat. Immunol. 2020, 21, 1160–1171. [Google Scholar] [CrossRef]
  33. Formenti, S.C.; Rudqvist, N.P.; Golden, E.; Cooper, B.; Wennerberg, E.; Lhuillier, C.; Vanpouille-Box, C.; Friedman, K.; Ferrari de Andrade, L.; Wucherpfennig, K.W.; et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat. Med. 2018, 24, 1845–1851. [Google Scholar] [CrossRef] [PubMed]
  34. Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; DeMaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef] [PubMed]
  35. Luke, J.J.; Onderdonk, B.E.; Bhave, S.R.; Karrison, T.; Lemons, J.M.; Chang, P.; Zha, Y.; Carll, T.; Krausz, T.; Huang, L.; et al. Improved Survival Associated with Local Tumor Response Following Multisite Radiotherapy and Pembrolizumab: Secondary Analysis of a Phase I Trial. Clin. Cancer Res. 2020, 26, 6437–6444. [Google Scholar] [CrossRef] [PubMed]
  36. de la Cruz-Merino, L.; Illescas-Vacas, A.; Grueso-López, A.; Barco-Sánchez, A.; Míguez-Sánchez, C. Radiation for Awakening the Dormant Immune System, a Promising Challenge to be Explored. Front. Immunol. 2014, 5, 102. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3953712/ (accessed on 14 September 2020). [CrossRef]
  37. Grassberger, C.; Ellsworth, S.G.; Wilks, M.Q.; Keane, F.K.; Loeffler, J.S. Assessing the interactions between radiotherapy and antitumour immunity. Nat. Rev. Clin. Oncol. 2019, 16, 729–745. [Google Scholar] [CrossRef] [PubMed]
  38. Yovino, S.; Kleinberg, L.; Grossman, S.A.; Narayanan, M.; Ford, E. The etiology of treatment-related lymphopenia in patients with malignant gliomas: Modeling radiation dose to circulating lymphocytes explains clinical observations and suggests methods of modifying the impact of radiation on immune cells. Cancer Investig. 2013, 31, 140–144. [Google Scholar] [CrossRef]
  39. Bockel, S.; Durand, B.; Deutsch, E. Combining radiation therapy and cancer immune therapies: From preclinical findings to clinical applications. Cancer Radiother. 2018, 22, 567–580. [Google Scholar] [CrossRef]
  40. Merrick, A.; Errington, F.; Milward, K.; O’Donnell, D.; Harrington, K.; Bateman, A.; Pandha, H.; Vile, R.; Morrison, E.; Selby, P.; et al. Immunosuppressive effects of radiation on human dendritic cells: Reduced IL-12 production on activation and impairment of naïve T-cell priming. Br. J. Cancer 2005, 92, 1450–1458. [Google Scholar] [CrossRef]
  41. 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]
  42. Woloshin, S.; Schwartz, L.M. Distribution of C-reactive protein values in the United States. N. Engl. J. Med. 2005, 352, 1611–1613. [Google Scholar] [CrossRef]
  43. Kushner, I.; Rzewnicki, D.; Samols, D. What does minor elevation of C-reactive protein signify? Am. J. Med. 2006, 119, 166.e17–166.e28. [Google Scholar] [CrossRef]
  44. McKelvey, K.J.; Hudson, A.L.; Back, M.; Eade, T.; Diakos, C.I. Radiation, inflammation and the immune response in cancer. Mamm. Genome 2018, 29, 843–865. [Google Scholar] [CrossRef]
  45. Brooks, E.D.; Chang, J.Y. Time to abandon single-site irradiation for inducing abscopal effects. Nat. Rev. Clin. Oncol. 2019, 16, 123–135. [Google Scholar] [CrossRef]
  46. Formenti, S.C. Optimizing Dose Per Fraction: A New Chapter in the Story of the Abscopal Effect? Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 677–679. [Google Scholar] [CrossRef]
  47. Altorki, N.K.; McGraw, T.E.; Borczuk, A.C.; Saxena, A.; Port, J.L.; Stiles, B.M.; Lee, B.E.; Sanfilippo, N.J.; Scheff, R.J.; Pua, B.B.; et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: A single-centre, randomised phase 2 trial. Lancet Oncol. 2021, 22, 824–835. [Google Scholar] [CrossRef]
  48. Lee, N.Y.; Ferris, R.L.; Psyrri, A.; Haddad, R.I.; Tahara, M.; Bourhis, J.; Harrington, K.; Chang, P.M.H.; Lin, J.C.; Razaq, M.A.; et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: A randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 2021, 22, 450–462. [Google Scholar] [CrossRef]
  49. Saddawi-Konefka, R.; O’Farrell, A.; Faraji, F.; Clubb, L.; Allevato, M.; Jensen, S.M.; Yung, B.S.; Wang, Z.; Wu, V.H.; Anang, N.A.; et al. Lymphatic-Preserving Treatment Sequencing with Immune Checkpoint Inhibition Unleashes cDC1-Dependent Antitumor Immunity in HNSC. Nat. Commun. 2022, 13, 4298. [Google Scholar] [CrossRef]
Figure 1. Kaplan- Meier estimates of progression-free survival at 24 months depending on the occurrence of an abscopal response.
Figure 1. Kaplan- Meier estimates of progression-free survival at 24 months depending on the occurrence of an abscopal response.
Cancers 14 04213 g001
Figure 2. Kaplan–Meier estimates of overall survival at 24 months depending on the administration of immunotherapy.
Figure 2. Kaplan–Meier estimates of overall survival at 24 months depending on the administration of immunotherapy.
Cancers 14 04213 g002
Figure 3. Kaplan–Meier estimates of overall survival at 24 months depending on the occurrence of abscopal response.
Figure 3. Kaplan–Meier estimates of overall survival at 24 months depending on the occurrence of abscopal response.
Cancers 14 04213 g003
Figure 4. ROC curves for the dosimetric parameters. AUC = Area under the curve.
Figure 4. ROC curves for the dosimetric parameters. AUC = Area under the curve.
Cancers 14 04213 g004
Table 1. Baseline and patient characteristics.
Table 1. Baseline and patient characteristics.
Baseline CharacteristicsOverall
(n = 118)
Age, median (range) years66.5 (23.5–100.1)
Sex, men (%)64 (54.2)
Performance status (%)
036 (30.6)
147 (39.8)
228 (23.7)
37 (5.9)
AJCC 8th stage (%)
M1A/IV29 (24.6)
M1B/IV5 (4.2)
M1C/IV68 (57.6)
M1D/IV16 (13.6)
Irradiated metastases (%)
Abdomen (spleen, liver)13 (11.0)
Lymph node28 (23.7)
Skin 23 (19.5)
Bone44 (37.3)
Lung4 (3.4)
Breast2 (1.7)
Muscle4 (3.4)
RT total dose, median (range) Gy30 (6.5–60)
RT dose per fraction, median (range) Gy4 (2–20)
SBRT (n = 15), median (range) Gy9 (8–20)
RT duration, median (range) days10 (1–63)
Multiple (≥2) irradiated metastases22(18.6)
Concomitant IT (%)80 (67.8)
Ipilimumab11 (13.7)
Nivolumab47 (58.8)
Nivolumab + Ipilimumab6 (7.5)
Pembrolizumab16 (20.0)
Time between the first injection of IT and RT, median (range) months.2.9 (0.5–47)
Immune-related events 16 (13.5)
Resistance to immunotherapy (%)
Primary34 (42.5)
Secondary46 (57.5)
AJCC 8th stage = American Joint Committee on Cancer 8th edition, Gy = Gray, immune-related events = episodes of infection and symptoms of acute clinical inflammation (redness, heat, swelling, pain, loss of function, and/or biological inflammatory syndrome with the elevation of inflammation parameters including at least C-reactive protein). IT = immunotherapy, RT = radiotherapy, SBRT = stereotactic body radiotherapy.
Table 2. Summary of the characteristics of abscopal responders.
Table 2. Summary of the characteristics of abscopal responders.
Sex Age1r or 2r to IT or noneSingle (1)
Or
Multiple (≥2)
Co RT plans
Site of
RT
RT schedulesRT irECo-RT anti-infectious agents 0 no 1 yesLT
Before RT
Timing of irEiRE categoryITOS
(month)
Time between the start of RT and AR detection
(days)
F8421Splenic metastasis3*6GyGastritis Helicobacter Pylori0 concomitantlightPembrolizumab2017
F7702Skin metastases of the leg7*6GyErysipelas on the same leg1 concomitantmoderate-1299
M5312Pulmonary nodules5*12Gy
3*6Gy
infectious pneumonia1 beforemoderateNivolumab1622
F6111Left axillary lymph node 3*6GyLeft Elbow adenitis. rise of CRP: 30 mg/L1630concomitantlightNivolumab1812
F7522Skin metastases of the right leg3*5.5Gyerysipelas and urinary infection1 concomitantmoderateNivolumab3224
F5612Right axillary lymph node + Skin metastases3*6GyUnknown biological inflammatory syndrome. Leucocytes: 18.35 G/L. Neutrophil count: 15.38 G/L
CRP: 130 mg/L
01080
concomitantsevereNivolumab1641
F7311Skin metastases of the left leg4*5GyInflammation axillary node
Thyroiditis
1 concomitantlightNivolumab1244
F6611Left axillary lymphnode 4*5GySubcutaneous inflammatory hyper metabolism0 before lightNivolumab6325
M6522Retrocava lymphnode 4*5GySigmoiditis0950beforemoderateNivolumab and Ipilimumab3226
M5822Oral floor+bone metastasis10*3GyOral infection1 concomitantmoderateNivolumab and Ipilimumab3116
W8621Left inguinal adenopathy 3*6Gybullous pemphigoid0900concomitantlightPembrolizumab1321
M7212Pulmonary nodules5*12GyRhinopharyngitis +follicular acnea01300concomitantmoderateNivolumab2328
M6721Skin metastases 13*3Gy-- - Ipilimumab6719
M8521Cervical lymph node15*3Gysarcoidosis- -moderateNivolumab1857
M8821Cervical lymph node30*2Gy-- - Nivolumab2845
IT = Immunotherapy; irE = immune-related events (immune-related events* = episodes of infection and symptoms of acute clinical inflammation (redness. heat. swelling. pain. loss of function and/or biological inflammatory syndrome with elevation of inflammation parameters including at least C-reactive protein); LT = lymphocytes; M = male W = female; 1r or 2r = primary or secondary resistance; RT = radiotherapy.
Table 3. Characteristics for the patients with an abscopal response (AR).
Table 3. Characteristics for the patients with an abscopal response (AR).
Patient CharacteristicsNo ARARp
n = 103n = 15
Age, median (range) years66.4 (23.3-100.1)66.8 (53.5- 87.8)0.09
Sex, Women (%)46 (44.7)8 (53.3)0.72
PS (%) 0.45
032 (31.1)4 (26.7)
140 (38.8)7 (46.7)
226 (25.2)2 (13.3)
35 (4.9)2 (13.3)
AJCC 8th stage (%) 0.10
M1A/IV24 (23.3)5 (33.4)
M1B/IV3 (2.9)2 (13.3)
M1C/IV60 (58.3)8 (53.3)
M1D/IV16 (15.5)0 (0.0)
Irradiated metastases (%) 0.02
Abdomen (spleen, liver)11 (10.7)2 (13.3)
Lymph node22 (21.4)6 (40.0)
Skin metastases19 (18.4)4 (26.7)
Bone43 (41.8)1 (6.7)
Lung2 (1.9)2 (13.3)
Breast2 (1.9)0 (0.0)
Muscle4 (3.9)0 (0.0)
RT total dose, median(range) Gy30 (6.5-60)30(16.5-60)0.17
RT dose per fraction, median (range) Gy4(2.4-20)4 (2-18)0.09
10 (1-63)11 (2-22)0.49
RT duration, median (range) days
Multiple (≥2) irradiated metastases (%)16 (15.5)6 (40.0)0.05
Concomitant immunotherapy (%)66 (55.9)14 (93.3)0.02
Resistance to immunotherapy 0.96
Primary28 (42.4)6 (42.9)
Secondary38 (57.6)8 (57.1)
RT immune-related events *3 (3.2)13 (86.7)<0.01
Local response for the RT-targets ** <0.01
Progressive15 (14.6)0 (0.0)
Stable disease58 (56.3)2 (13.3)
Tumor regression30 (29.1)13 (86.7)
AJCC 8th stage = American Joint Committee on Cancer 8th edition; RT = radiotherapy; immune-related events* = episodes of infection and symptoms of acute clinical inflammation (redness, heat, swelling, pain, loss of function and/or biological inflammatory syndrome with elevation of inflammation parameters including at least C-reactive protein); ** RECIST 1.1 and/or iRECIST in case of immunotherapy.
Table 4. Univariate and multivariate analyses for progression-free survival.
Table 4. Univariate and multivariate analyses for progression-free survival.
DemographicsUnivariate AnalysisMultivariate Analysis
HRp ValueHRp Value
Age (per 1 year increase)0.99 [0.98; 1.01]0.41
Sex, female
male
Reference
1.55 [1.03; 2.33]
0.031.89 [1.21; 2.93]<0.01
Clinical characteristics
Performance status
0
Reference
11.09 [0.67; 1.78]0.72
21.82 [1.06; 3.13]0.032.09 [1.21; 3.63]<0.01
31.19 [0.42; 3.40]0.74
AJCC 8th stage
M1A/IV
Reference
M1B/IV0.45 [0.13; 1.49]0.19
M1C/IV0.88 [0.55; 1.42]0.62
M1D/IV1.30 [0.67; 2.51]0.43
Number of irradiated metastases
One metastasis
Multiple metastasis (≥2)
Reference
0.56 [0.32; 0.98]
0.040.83 [0.47; 1.49]0.53
RT dose0.98 [0.97; 1.01]0.09
Immunotherapy
None
Concomitant with RT
Reference
0.51 [0.33; 0.78]
<0.010.64 [0.40; 1.02]0.06
Abscopal response
No AR
AR

Reference
0.08 [0.02; 0.24]
<0.010.1 [0.03; 0.32]<0.01
AJCC 8th stage = American Joint Committee on Cancer 8th edition; RT = radiotherapy, immune-related events* = episodes of infection and symptoms of acute clinical inflammation (redness. heat. swelling. pain. loss of function and/or biological inflammatory syndrome with elevation of inflammation parameters including at least C-reactive protein).
Table 5. Univariate and multivariate analyses for overall survival.
Table 5. Univariate and multivariate analyses for overall survival.
DemographicsUnivariate AnalysisMultivariate Analysis
HRp-ValueHRp-Value
Age (per 1 year increase)0.99 [0.98; 1.01]0.64
Gender female
male
Reference
1.31 [0.84; 2.05]
0.23
Clinical characteristics
Performance status
0

Reference
11.41 [0.79; 2.51]0.24
23.05 [1.65; 5.63]<0.012.73 [1.46; 5.09]<0.01
34.20 [1.44; 12.27]0.014.70 [1.59; 13.94]<0.01
AJCC 8th stage
M1A/IV

Reference
M1B/IV0.63 [0.14; 2.72]0.53
M1C/IV1.57 [0.90; 2.75]0.11
M1D/IV2.20 [1.04; 4.67]0.042.45 [0.164; 3.97]0.63
Number of irradiated metastases
One metastasis
Multiple metastasis (≥2)

Reference
0.38 [0.18; 0.78]
0.01



0.48 [0.23; 1.02]
0.05

RT dose0.98 [0.96; 0.99]0.040.98 [0.96; 1.01]0.11
Immunotherapy
None
Concomitant with RT

Reference
0.49 [0.31; 0.77]
<0.01



0.51 [0.30; 0.87]
0.01

Abscopal response0.19 [0.04; 0.46]<0.010.25 [0.07; 0.88]0.03
AJCC 8th stage = American Joint Committee on Cancer 8th edition; RT = radiotherapy, immune-related events = episodes of infection and symptoms of acute clinical inflammation (redness, heat, swelling, pain, loss of function, and/or biological inflammatory syndrome with the elevation of inflammation parameters, including, at least, C-reactive protein).
Table 6. Risk factors of abscopal response.
Table 6. Risk factors of abscopal response.
DemographicsUnivariate AnalysisMultivariate Analysis
HRp-ValueHRp-Value
Age (per 1 year increase)1.03 [0.99; 1.06]0.09
Gender
Female
Male

Reference
1.43 [0.52; 3.96]
0.48
Clinical characteristics
Performance status
0

Reference
11.45 [0.42; 4.95]0.55
20.89 [0.16; 4.85]0.89
34.67 [0.85; 25.69]0.07
AJCC 8th stage
M1A/IV

Reference
M1B/IV2.80 [0.54; 14.47]0.22
M1C/IV0.73 [0.24; 2.22]0.57
M1D/IV0.00 [0.00; Inf]0.99
Total RT dose1.02 [0.98; 1.06]0.27
RT dose per fraction1.10 [0.98; 1.24]0.11
RT duration0.96 [0.90; 1.04]0.34
Number of irradiated metastases
One metastasis
Multiple metastasis (≥2)

* Reference
3.16 [1.12; 8.89]
0.03



16.85 [2.16; 131.49]
<0.01

Irradiated metastases
Abdomen (spleen, liver)0.20 [0.01; 3.27]0.26
Lymph node0.52 [0.06; 4.35]0.55
Skin0.56 [0.06; 4.83]0.60
Bone0.00 [0.00; Inf]0.99
Lung1.56 [0.14; 17.29]0.72
Breast0.00 [0.00; Inf]0.99
Muscle0.00 [0.00; Inf]0.99
Immunotherapy
None
Concomitant with RT

Reference
7.56 [0.99; 57.57]
0.05

IpilimumabReference
Nivolumab2.05 [0.26; 16.21]0.49
Nivolumab + ipilimumab3.79 [0.34; 41.86]0.28
Pembrolizumab1.32 [0.12; 14.59]0.82
Occurrence of immune-related events *
64.83 [14.48; 290.18]
<0.01

403.45 [13.83; 11769.39]
<0.01
AJCC 8th stage = American Joint Committee on Cancer 8th edition; RT = radiotherapy; immune-related events * = episodes of infection and symptoms of acute clinical inflammation (redness, heat, swelling, pain, loss of function, and/or biological inflammatory syndrome with the elevation of inflammation parameters, including, at least, C-reactive protein).
Table 7. ROC analysis.
Table 7. ROC analysis.
Total Radiation Dose (Gray)Dose per Session (Gray)Fractionation of Radiation Total Treatment Time, Including Breaks (Days)
Optimal value195.253.54.5
Specificity0.7650.6790.6910.864
Sensitivity0.810.80.8
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ollivier, L.; Orione, C.; Bore, P.; Misery, L.; Legoupil, D.; Leclere, J.-C.; Coste, A.; Girault, G.; Sicard-Cras, I.; Kacperek, C.; et al. Abscopal Response in Metastatic Melanoma: Real-World Data of a Retrospective, Multicenter Study. Cancers 2022, 14, 4213. https://doi.org/10.3390/cancers14174213

AMA Style

Ollivier L, Orione C, Bore P, Misery L, Legoupil D, Leclere J-C, Coste A, Girault G, Sicard-Cras I, Kacperek C, et al. Abscopal Response in Metastatic Melanoma: Real-World Data of a Retrospective, Multicenter Study. Cancers. 2022; 14(17):4213. https://doi.org/10.3390/cancers14174213

Chicago/Turabian Style

Ollivier, Luc, Charles Orione, Paul Bore, Laurent Misery, Delphine Legoupil, Jean-Christophe Leclere, Anne Coste, Gilles Girault, Iona Sicard-Cras, Clemence Kacperek, and et al. 2022. "Abscopal Response in Metastatic Melanoma: Real-World Data of a Retrospective, Multicenter Study" Cancers 14, no. 17: 4213. https://doi.org/10.3390/cancers14174213

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