Radiotherapy and immunotherapy benefit subsets of patients with metastatic cancer. Here, we review selected laboratory and clinical studies investigating the utility of combining radiotherapy and immunotherapy in metastatic patients. We examine potential approaches to increase the therapeutic ratio of radioimmunotherapy in the treatment of metastatic cancers moving forward.

A growing body of literature supports the hypothesis that metastasis-directed radiotherapy delays progression and confers a survival benefit to a select group of patients with metastatic cancer (1–5). Immunotherapy has also been demonstrated to improve outcomes among subsets of patients with metastatic disease and has changed the standard of care for many malignancies (6). Immunomodulatory effects of ionizing radiation are currently under investigation and therefore, combined treatment with radioimmunotherapy has been proposed as a method of optimizing outcomes in patients with metastatic disease. Here, we review clinical studies that examine the treatment of metastatic disease with radiotherapy and immunotherapy, individually and in combination, and suggest future avenues for investigating the role of radioimmunotherapy.

Role of radiotherapy

Radiotherapy has historically been used with palliative intent in patients with metastatic cancer; however, mounting evidence suggests that radiotherapy may play a role in the curative management of certain patients with metastasis. Several recently completed randomized phase II trials support the hypothesis that metastasis-directed radiotherapy delays progression and may confer a survival benefit in select groups of patients (1–5). Nonrandomized prospective series with extended follow-up suggest that durable disease control can be achieved following ablative radiotherapy to all sites of metastatic disease (7–9). Clinical studies supporting the benefit of metastasis-directed radiotherapy are heterogeneous, and exclusively focus on patients with few sites of disease (typically ≤ 5 metastases). Ongoing phase III trials are investigating the role of metastasis-directed radiotherapy in patients with 1–3 metastases (NCT03862911) and 4–10 metastases (NCT0372134) separately and additional phase III trials of metastasis-directed radiotherapy include burden of disease as a stratification factor (NCT02364557, NCT03137771).

Role of immunotherapy

Immunotherapy plays an important role in the management of a subset of patients with metastatic cancer, including patients with metastatic melanoma and non–small cell lung cancer (NSCLC), among other histologies. Among patients with metastatic melanoma treated with pembrolizumab on KEYNOTE-001, 5-year overall survival (OS) was 34% in all patients and 41% in treatment-naïve patients (10), which represents a dramatic improvement in outcomes as compared with previous systemic therapies for this disease (11). More recent data from patients with advanced melanoma treated with nivolumab and ipilimumab resulting in a 5-year OS of 52% (12) support the conclusion that immunotherapy can provide long-term disease control for a subset of metastatic patients. Likewise, in patients with metastatic NSCLC treated on KEYNOTE-189, the addition of pembrolizumab to chemotherapy (pemetrexed and platinum) resulted in a 2-year OS of 46%, compared with 30% in patients treated with chemotherapy alone [HR, 0.56 (95% confidence interval, CI: 0.45–0.70)] (13). Nonrandomized data from patients treated with pembrolizumab for metastatic NSCLC with a PD-L1 tumor proportion score ≥ 50% demonstrate 5-year OS of 30% and 25% in treatment-naïve and previously treated patients, respectively (14), further supporting the potential for durable disease control in a subset of metastatic patients treated with immunotherapy.

Analogous to the impact of metastasis-directed radiotherapy, the efficacy of immunotherapy is greatest in the setting of low-volume disease. On secondary analysis of KEYNOTE-001, patients with less than the median baseline tumor size (summed diameter < 10.2 cm) were noted to have an improved overall objective response rate (ORR; 44% vs. 23%; P < 0.001) and OS (HR, 0.38; P < 0.001; ref. 15). Further evidence that immunotherapy is most effective in low-volume disease is observed in the adjuvant setting after local therapy for nonmetastatic disease. Among patients who underwent complete resection of high-risk stage III cutaneous melanoma on KEYNOTE-054, patients treated with adjuvant ipilimumab had a significantly improved 5-year OS compared with patients receiving placebo (65% vs. 54%; P = 0.001; ref. 16). Moreover, in the IMMUNED trial, patients with advanced melanoma with no evidence of residual disease following local therapy had a significantly improved 2-year recurrence-free survival following treatment with ipilimumab and nivolumab [70% (95% CI: 61–85)] compared with patients treated with placebo [14% (95% CI: 6–26)] or nivolumab alone [42% (95% CI: 29–55)] (17). Likewise, the PACIFIC trial demonstrated that 3-year OS was significantly greater with the addition of durvalumab among patients without evidence of progression of disease following definitive chemoradiotherapy for locally advanced NSCLC [57% (95% CI: 52–61)] compared with those receiving placebo [44% (37–50)] (18). Similar results have also been seen in patients with esophageal/gastroesophageal junction cancer treated with neoadjuvant chemoradiotherapy followed by surgery on CheckMate 577, where the addition of adjuvant nivolumab significantly improved disease-free survival compared with placebo [HR, 0.69 (96.4% CI: 0.56–0.86); P < 0.001] (19). These data from the adjuvant setting represent some of the most successful demonstrated applications of immunotherapy, further supporting the idea that the efficacy of immunotherapy is highest in the setting of low-volume disease. Thus, although data supporting the treatment of patients with metastatic disease using either radiotherapy or immunotherapy are seemingly disparate, both therapies have increased efficacy in patients with low disease burden. These results are analogous to those observed with chemotherapy and radiotherapy generally.

Given the individual efficacies of radiotherapy and immunotherapy and the proposed immunomodulatory effects of radiotherapy, the combination of radiotherapy and immunotherapy has been suggested as a strategy to improve outcomes in patients with metastatic disease. Initial studies investigated whether the combination of radiotherapy and immunotherapy impacts unirradiated (abscopal) or local irradiated tumor responses. Unlike the adjuvant setting, in which immunotherapy is used as a means to increase immune surveillance to eliminate subclinical residual disease and prevent disease recurrence, in this setting immunotherapy is aimed at potentiating distant responses. Demaria and colleagues initially proposed that abscopal effects following radiotherapy are mediated by T cells (20) and accordingly later demonstrated in murine mammary carcinoma models that the combination of radiotherapy and CTLA-4 blockade inhibited development of lung metastases (21). A number of preclinical data have subsequently demonstrated the potential for abscopal responses following radioimmunotherapy. For instance, in melanoma and renal cell carcinoma mouse models, the combination of radiotherapy with programmed cell death protein 1 (PD-1) blockade resulted in a 66% reduction in the size of tumors outside of the irradiated field (22). Other studies utilizing murine carcinoma models have similarly demonstrated the potential for radioimmunotherapy to induce significant out-of-field responses (23, 24).

In contrast to preclinical models, robust abscopal responses are rarely observed in clinical metastases (25). Postow and colleagues described a case in which a patient being treated with ipilimumab for metastatic melanoma received radiotherapy to a progressing paraspinal metastasis and subsequently had an impressive response in unirradiated metastases (26). A number of subsequent prospective studies have sought to replicate these findings by combining immunotherapy with radiotherapy directed at a single metastatic site. In a study reported by Golden and colleagues, patients with solid metastatic tumors with ≥ 3 metastases were treated with radiotherapy (35 Gy in 10 fractions) to one metastasis in combination with GMCSF (27). Responses in unirradiated metastases were noted in 11 of 41 enrolled patients, with 8 of the 11 patients with out-of-field responses having three lesions at baseline and the additional 3 patients having 4–6 lesions at baseline (27). [Of note, there is no consensus definition regarding what constitutes an abscopal response though in this study a decrease in the longest diameter of any measurable nonirradiated lesion by ≥ 30% was considered an abscopal response; this differs from the summed change in index lesion diameter used for RECIST (28) or the modified response criteria used to capture later responses that are common in patients treated with immunotherapy, immune-related response criteria (29)]. As described in Table 1, comparative studies reported by McBride and colleagues (30), Moreno and colleagues (31), and Theelen and colleagues (32) similarly investigated radiotherapy targeted at one metastasis in combination with immunotherapy. In each of these of these phase II studies, patients received PD-1 blockade alone or in combination with metastasis-directed radiotherapy. Notably, none of these studies demonstrated significant improvements in ORR, progression-free survival (PFS), or OS with the addition of radiotherapy to immunotherapy. Considered together, these studies suggest that laboratory studies may not faithfully reproduce aspects of clinical interactions between radiotherapy and immunotherapy and that abscopal effects may not be as common in humans as preclinical models suggest.

Table 1.

Reported phase I/II studies of immunotherapy with and without radiotherapy in patients with metastatic disease.

First authorPrimaryTreatmentRandomly assigned, nToxicityORR (RT±ICB vs. ICB)a
Curti (35) Melanoma IL2 ± 20 Gy × 1–2 to 1–3 lesions; RT given 3 days prior to IL2 44 G5 respiratory failure in 1 patient in RT+ICB arm 54% vs. 35% (NS) 
Mahmood (36) ACC Pembrolizumab ± 6 Gy × 5 to 1–5 leions; RT given within 1 week of ICB 20 G3 transaminitis in 1 patient; no additional G3+; NS difference between arms 50% vs. 70% (p = 0.65) 
McBride (30) HNSCC Nivolumab ± 9 Gy × 3 to 1 lesion; RT given between cycles 1 and 2 of ICB 62 G3–5 in 10% (RT+ICB) vs. 13% (ICB) (P = 0.70) 29% vs. 35% (p = 0.86) 
Moreno (31) NSCLC Cemiplimab ± 9 Gy × 3 to 1 lesion; RT given within 1 week of ICB 20/33b ICB EC: G3 pneumonia (10%); ICB+RT EC: G3 anemia (12%), G3 hypophosphatemia (6%), G3 UTI (6%), G5 pneumonitis (6%) 18% vs. 40% (NS) 
Theelenc (32) NSCLC Pembrolizumab ± 8 Gy × 3 to 1 lesion; RT given within 1 week of ICB 76 17% G3+; NS difference between arms 36% vs. 18% (p = 0.07) 
Welshc (37) NSCLC Pembrolizumab ± RTd to 1–4 lesions; RT given concurrent with cycle 1 of ICB 20/80e Phase 1: 30% G3+, no G4+; Phase 2: 11% G3+, two G4 cardiac AEs in 1 patient (all in RT+ICB arm) 22% vs. 25% (p = 0.99) 
First authorPrimaryTreatmentRandomly assigned, nToxicityORR (RT±ICB vs. ICB)a
Curti (35) Melanoma IL2 ± 20 Gy × 1–2 to 1–3 lesions; RT given 3 days prior to IL2 44 G5 respiratory failure in 1 patient in RT+ICB arm 54% vs. 35% (NS) 
Mahmood (36) ACC Pembrolizumab ± 6 Gy × 5 to 1–5 leions; RT given within 1 week of ICB 20 G3 transaminitis in 1 patient; no additional G3+; NS difference between arms 50% vs. 70% (p = 0.65) 
McBride (30) HNSCC Nivolumab ± 9 Gy × 3 to 1 lesion; RT given between cycles 1 and 2 of ICB 62 G3–5 in 10% (RT+ICB) vs. 13% (ICB) (P = 0.70) 29% vs. 35% (p = 0.86) 
Moreno (31) NSCLC Cemiplimab ± 9 Gy × 3 to 1 lesion; RT given within 1 week of ICB 20/33b ICB EC: G3 pneumonia (10%); ICB+RT EC: G3 anemia (12%), G3 hypophosphatemia (6%), G3 UTI (6%), G5 pneumonitis (6%) 18% vs. 40% (NS) 
Theelenc (32) NSCLC Pembrolizumab ± 8 Gy × 3 to 1 lesion; RT given within 1 week of ICB 76 17% G3+; NS difference between arms 36% vs. 18% (p = 0.07) 
Welshc (37) NSCLC Pembrolizumab ± RTd to 1–4 lesions; RT given concurrent with cycle 1 of ICB 20/80e Phase 1: 30% G3+, no G4+; Phase 2: 11% G3+, two G4 cardiac AEs in 1 patient (all in RT+ICB arm) 22% vs. 25% (p = 0.99) 

Abbreviations: ACC, adenoid cystic carcinoma; EC, expansion cohort; Gy, Gray; G3, grade 3 adverse event; G4, grade 4 adverse event; G5, grade 5 adverse event; HNSCC, head and neck squamous cell carcinoma; ICB, immune checkpoint blockade, IL2, interleukin-2; NS, nonsignificant; NSCLC, non–small cell lung cancer; RT, radiotherapy.

aOut-of-field objective response rate in patients treated with RT+ICB and ICB alone in each of the reported studies.

bThe comparative study reported by Moreno and colleagues consists of the interim report of two separate nonrandomized phase I expansion cohorts in which patients were treated with ICB alone and RT+ICB, respectively.

cIt is of note that an unplanned pooled analysis of the studies by Theelen and colleagues and Welsh and colleagues demonstrated improved PFS (4.4 vs. 9.0 months, P = 0.045) and OS (8.7 vs. 19.2 months, P < 0.001) with the addition of RT to ICB (38).

dIn the study by Welsh and colleagues, patients randomized to RT+ICB were treated with an RT dose of 12.5 Gy × 4 if deemed clinically feasible (n = 19) and otherwise were treated with 3 Gy × 15 (n = 21).

eThe study reported by Welsh and colleagues included 20 patients in the initial phase I component, with 80 patients randomly assigned on the subsequent phase II portion.

In contrast to studies targeting a single metastasis with radiotherapy, a number of prospective studies have combined multi-site metastasis-directed radiotherapy with immunotherapy. In a study reported by Luke and colleagues, 73 patients with a variety of advanced solid tumors were treated with 30–50 Gy in 3–5 fractions to 2–4 metastases prior to pembrolizumab, yielding an out-of-field response (≥ 30%) in one or more metastases in 27% of patients (33). Although the authors were encouraged by the 2-year OS of patients in this study of nearly 20% (34), it is possible that these results may have been achieved with treatment with immunotherapy alone. Randomized studies by reported by Curti and colleagues (35), Mahmood and colleagues (36), and Welsh and colleagues (37) also investigated the use of multi-site metastasis-directed radiotherapy in combination with immunotherapy, as detailed in Table 1. In these studies, the addition of metastasis-directed radiotherapy did not increase ORR, PFS, or OS compared with systemic therapy with IL2 (35) or pembrolizumab alone (36, 37). It is of note that an unplanned pooled analysis of the studies by Theelen and colleagues and Welsh and colleagues demonstrated improved PFS (4.4 vs. 9.0 months, P = 0.045) and OS (8.7 vs. 19.2 months, P < 0.001) with the addition of radiotherapy to immune checkpoint blockade (ICB; ref. 38). Radioimmunotherapy has also been investigated in the context of multi-site metastasis-directed radiotherapy targeting all sites of metastatic disease. In a nonrandomized phase II study reported by Bauml and colleagues, locally ablative therapy (surgery, radiotherapy, chemoradiotherapy, or interventional ablation) to all sites of disease (≤ 4 metastases) was combined with pembrolizumab in 45 patients with metastatic NSCLC yielding 2-year PFS of 56% and 17% in patients with metachronous and synchronous metastatic disease, respectively [HR, 1.98 (95% CI: 0.87–4.52)], and a significant improvement in the primary endpoint of the study, median PFS from time of locally ablative therapy compared with historical outcomes (19.1 months vs. 6.6 months, P = 0.005), with several patients surviving greater than 3 years following the completion of locally ablative therapy (39).

Overall, the results of prospective studies investigating the use of radioimmunotherapy in patients with metastatic disease are not conclusive. Beyond the number of metastases targeted with radiotherapy, several additional variables exist in the design of currently reported studies of radioimmunotherapy in metastatic patients. Thus, data examining radioimmunotherapy in patients with metastatic cancer should be further considered in the context of a number of tumor and treatment-related factors.

Radiotherapy treatment volume

Although the majority of studies of radioimmunotherapy in metastatic patients have required at least one metastasis to be unirradiated (27, 30–33, 35–37), a number of preclinical and translational data support the rationale for treating all sites of metastatic disease with radiotherapy, similar to the approach in the study reported by Bauml and colleagues (39). High doses of radiotherapy have been demonstrated to kill resistant tumor clones and increase tumor T-cell infiltration and priming without killing radioresistant tumor-reprogrammed resident T cells (40–42). In addition, radiotherapy has been demonstrated to enhance MHC class I (MHC-I) expression (43) and induce and/or upregulate the expression of genes containing immunogenic mutations and nonmutated genes that are immunogenic when overexpressed (44). Given that downregulation of MHC-I is known to be an important mechanism of tumor immune evasion (45) and that tumor neoantigens have been demonstrated to drive response to ICB (46), it is plausible that antitumor immune response may be enhanced in irradiated metastases. An interesting observation noted by Luke and colleagues was that partial irradiation of large metastases did not result in inferior local control compared with targeting the entire tumor (33). Thus, is possible that the potential benefits of using radiotherapy targeted at all sites of disease in combination with immunotherapy may be attainable even if treating the entire volume of each metastasis is not feasible. However, it should be noted that given that approximately 80% of the patients in this study had expired within 2 years of treatment, the definitive conclusions that can be derived from this observation are limited. Nonetheless, it is hoped that future technical advances in radiotherapy will increase the number of tumors that can be treated to a curative dose.

In contrast to tumor-reprogramed resident T cells, T cells in lymphoid organs are radiosensitive (41). Draining lymph nodes have been demonstrated to be essential for the accumulation and priming of T cells induced in response to ionizing radiation (47) and thus it is feasible that nodal irradiation may decrease antitumor immune response. Consistent with this finding, a study in which mice were treated with concurrent ICB and radiotherapy to either tumor alone or tumor and draining lymph nodes found that inclusion of the draining lymph nodes in the irradiated field resulted in lower levels of immunostimulatory chemokines and tumor-specific CD8+ T cells, as well as worse OS (48). In addition, in contrast to randomized trials that have demonstrated improved outcomes with the addition of ICB to chemoradiotherapy in patients with locally advanced NSCLC (18) and esophageal cancer (19), similar trials for patients with locally advanced head and neck cancer have not been successful (49, 50). While a number of differences in trial design and populations examined in these studies could potentially explain their disparate results, it is conceivable that this discrepancy could be explained by the immunosuppressive effects of elective nodal irradiation in the head and neck cancer studies. Similarly, given that radiotherapy-induced lymphopenia is associated with a poor prognosis and decreased response to ICB (51, 52), minimizing the volume of bone marrow irradiated may be an important consideration in patients treated with radioimmunotherapy (53).

Radiotherapy dose and fractionation

Although several comparative studies have utilized radiotherapy doses of 24–30 Gy given in 3–5 fractions (30–32, 36) a number of other dose/fractionation schemes have been utilized in randomized studies of radioimmunotherapy in metastatic patients. Notably, the study reported by Curti and colleagues, utilized either 1 or 2 fractions of 20 Gy (35), while patients in the study reported by Welsh and colleagues were treated with 48 Gy in 4 fractions if deemed clinically feasible (n = 19) and otherwise were treated with 45 Gy in 15 fractions (n = 21; ref. 37). Dose and fractionation are similarly heterogeneous in the reported noncomparative studies of radioimmunotherapy in patients with metastatic disease (27, 33). Consistent with these varied approaches, the radiotherapy dose and fractionation scheme most likely to optimize the benefit of radioimmunotherapy remains unclear. In patients treated with metastasis-directed radiotherapy alone, ablative doses have been demonstrated to be crucial to achieve high rates of local control (54). In addition, it has been demonstrated that type 1 IFN produced in response to high doses of ionizing radiation is important for radiotherapy-mediated tumor control (40, 55). However, it is of note that large radiotherapy doses may attenuate radiotherapy-induced tumor immunogenicity via induction of the DNA exonuclease TREX1 (56), which in some instances has been correlated with decreased expression of cytolytic T-cell genes and inferior irradiated tumor local control following radiotherapy and ICB (34). Thus, further investigation is required to better delineate the ideal radiotherapy dose and fractionation scheme for metastatic patients treated with radioimmunotherapy.

Immunotherapy modality and timing

Most of studies of radioimmunotherapy in metastatic patients have utilized ICB agents targeting either PD-1 (30–33, 36, 37, 39) or CTL-associated protein 4 (CTLA-4; refs. 57, 58). Among studies investigating combined radiotherapy and ICB, the sequencing of these therapies is variable. Nonrandomized series have suggested that treatment response and OS are improved with concurrent radiotherapy and ICB compared with sequential therapy (59, 60). In addition, preliminary data from a phase I trial in which patients with metastatic NSCLC treated with multi-site ablative radiotherapy were randomized to receive either concurrent or sequential ipilimumab and nivolumab suggest that concurrent ICB results in fewer dose-limiting toxicities and improved cytoreduction (61). However, the ideal sequencing of radioimmunotherapy remains largely unclear and likely depends upon a number of factors including the immunotherapy agent utilized, radiotherapy dose/fractionation, and the clinical scenario. Thus, future studies are necessary to better delineate the ideal immunotherapy modality and timing relative to radiotherapy in metastatic patients treated with radioimmunotherapy.

Antagonistic effects of radiotherapy and immunotherapy

Antagonistic effects of radiotherapy and immunotherapy may, at least in part, account for the heterogeneous results of trials of radioimmunotherapy in patients with metastatic disease. Despite the rationale for combining radiotherapy and immunotherapy, which has been detailed previously (62), a number of potentially antagonistic effects may prevent these therapies from acting synergistically. IFN induced in response to high-dose radiotherapy has been demonstrated to result in an immune cascade that is important for tumor control (55); however, prolonged IFN signaling has also been shown to result in resistance to T cell–mediated cytotoxicity (63). Factors secreted into the tumor microenvironment following radiotherapy can also result in an efflux of immunostimulatory cells and attract immunosuppressive cell populations (20, 40). In particular, high levels of infiltrating regulatory T cells (Treg) following radiotherapy have been demonstrated to suppress antitumor immune response, resulting in poor clinical outcomes (64, 65). Moreover, Tregs enhance the function of tumor-associated myeloid cells including M2-like tumor-associated macrophages and myeloid-derived suppressor cells, both of which have been shown to negatively impact disease control following radiotherapy (66, 67). One hypothesis is that immunomodulatory effects of ionizing radiation are complex and likely simultaneously important for tumor control, but also potentially detrimental if prolonged, much the same way that acute inflammation in response to a foreign pathogen is adaptive but can also become disastrous if unabated. Thus, additional strategies aimed at negating the immunosuppressive effects of radiotherapy while preserving antitumor response require further investigation and may be important to maximize the therapeutic ratio of radioimmunotherapy in metastatic patients.

Radiotherapy and immunotherapy have been demonstrated to be effective when used together, most convincingly so in the adjuvant setting. However, the role of immunotherapy in this setting, differs from its application in the metastatic setting and these data do not necessarily imply a positive interaction between radiotherapy and immunotherapy. Some trials to date do suggest an interaction occurs in a subset of patients; however, this population seems relatively small and more work needs to be done to allow consistent identification of these patients. Radiotherapy and immunotherapy seemingly provide the largest benefit to metastatic patients with low-volume disease. Although a variety questions remain regarding the optimal strategies for combining radiotherapy and immunotherapy in metastatic patients, data seemingly support the benefit of targeting all sites of metastasis to reduce tumor burden. Given that patients with oligometastatic disease inherently have a low disease burden that is likely to be amenable to metastasis-directed radiotherapy to all sites of disease, patients with oligometastatic disease may be best suited for future studies of radioimmunotherapy.

Initially proposed by Hellman and Weichselbaum in 1995, the oligometastatic hypothesis postulates the existence of a state characterized by limited disease burden that has metastasized distantly and a slow rate of progression (68). Pending the results of ongoing phase III trials, preliminary data suggest that patients with oligometastatic disease may benefit from metastasis-directed radiotherapy. Although durable disease control can be achieved following ablative radiotherapy to all sites of metastatic disease in a subset of patients, immunotherapy may serve as a strategy to eradicate subclinical disease and improve outcomes in the remaining patients. Thus, future studies investigating the role of radioimmunotherapy in the management of metastatic patients should focus upon patients with oligometastatic disease.

In addition to the volume and rate of metastatic progression, timing of metastasis, histology, and lymph node status, among other factors, have been demonstrated to be prognostic in patients with purported oligometastatic disease (69–71). Recent work has also identified molecular factors associated with oligometastatic versus polymetastatic progression (71) and an integrated molecular classification scheme has been demonstrated to successfully stratify patients with hepatic colorectal cancer metastases by risk of failure following metastasectomy (72). Thus, these molecular and clinicopathologic factors will be important considerations in the design and interpretation of future studies of radioimmunotherapy in patients with oligometastatic disease, as will be the identification of novel biomarkers that more clearly delineate the oligometastatic state. Moreover, future work will need to delineate the ideal radiotherapy dose/fractionation, immunotherapy agents, and sequencing of radiotherapy and immunotherapy in patients with oligometastatic disease treated with radioimmunotherapy. In addition, studies are needed to investigate methods to overcome known antagonistic effects of radiotherapy and immunotherapy as well as novel targets to increase the efficacy of these therapies, such as the microbiome, which has been shown to impact both response to ICB (73) and radiotherapy-induced antitumor immune response (74, 75).

In conclusion, both radiotherapy and immunotherapy have been shown to improve outcomes in select patients with metastatic cancer. Despite rationale supporting the potential utility of treating metastatic patients with radioimmunotherapy, the design and results of preliminary investigations of this therapeutic approach have been heterogeneous. Future studies investigating the role of radioimmunotherapy in the management of metastatic patients should focus upon patients with oligometastatic disease.

R.R. Weichselbaum reports other support from Boost Therapeutics, Immvira LLC, Reflexion Pharmaceuticals, Coordination Pharmaceuticals Inc., Magi Therapeutics, Oncosenescence, Aettis Inc., AstraZeneca, Coordination Pharmaceuticals, Genus, Merck Serono S.A., Nano proteagen, NKMax America Inc, Shuttle Pharmaceuticals, and Highlight Therapeutics; grants from Varian and Regeneron; and personal fees from AstraZeneca, Boehringer Ingelheim LTD, and Merck Serono S.A. outside the submitted work. No disclosures were reported by the other authors.

We would like to acknowledge the Ludwig Cancer Research Foundation and the Foglia Family Foundation for their funding support.

1.
Gomez
DR
,
Tang
C
,
Zhang
J
,
Blumenschein
GR
 Jr
,
Hernandez
M
,
Lee
JJ
, et al
Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study
.
J Clin Oncol
2019
;
37
:
1558
65
.
2.
Iyengar
P
,
Wardak
Z
,
Gerber
DE
,
Tumati
V
,
Ahn
C
,
Hughes
RS
, et al
Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial
.
JAMA Oncol
2018
;
4
:
e173501
.
3.
Ost
P
,
Reynders
D
,
Decaestecker
K
,
Fonteyne
V
,
Lumen
N
,
De Bruycker
A
, et al
Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: a prospective, randomized, multicenter phase II trial
.
J Clin Oncol
2018
;
36
:
446
53
.
4.
Palma
DA
,
Olson
R
,
Harrow
S
,
Gaede
S
,
Louie
AV
,
Haasbeek
C
, et al
Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: long-term results of the SABR-COMET phase II randomized trial
.
J Clin Oncol
2020
;
38
:
2830
8
.
5.
Phillips
R
,
Shi
WY
,
Deek
M
,
Radwan
N
,
Lim
SJ
,
Antonarakis
ES
, et al
Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial
.
JAMA Oncol
2020
;
6
:
650
9
.
6.
Waldman
AD
,
Fritz
JM
,
Lenardo
MJ
. 
A guide to cancer immunotherapy: from T cell basic science to clinical practice
.
Nat Rev Immunol
2020
;
20
:
651
68
.
7.
Milano
MT
,
Katz
AW
,
Zhang
H
,
Huggins
CF
,
Aujla
KS
,
Okunieff
P
. 
Oligometastatic breast cancer treated with hypofractionated stereotactic radiotherapy: Some patients survive longer than a decade
.
Radiother Oncol
2019
;
131
:
45
51
.
8.
Horner-Rieber
J
,
Bernhardt
D
,
Blanck
O
,
Duma
M
,
Eich
HT
,
Gerum
S
, et al
Long-term follow-up and patterns of recurrence of patients with oligometastatic NSCLC treated with pulmonary SBRT
.
Clin Lung Cancer
2019
;
20
:
e667
e77
.
9.
Weichselbaum
RR
. 
The 46th david a. karnofsky memorial award lecture: oligometastasis—from conception to treatment
.
J Clin Oncol
2018
;
36
:
3240
50
.
10.
Hamid
O
,
Robert
C
,
Daud
A
,
Hodi
FS
,
Hwu
WJ
,
Kefford
R
, et al
Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001
.
Ann Oncol
2019
;
30
:
582
8
.
11.
Ugurel
S
,
Röhmel
J
,
Ascierto
PA
,
Flaherty
KT
,
Grob
JJ
,
Hauschild
A
, et al
Survival of patients with advanced metastatic melanoma: the impact of novel therapies–update 2017
.
Eur J Cancer
2017
;
83
:
247
57
.
12.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Rutkowski
P
,
Lao
CD
, et al
Five-year survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2019
;
381
:
1535
46
.
13.
Gadgeel
S
,
Rodriguez-Abreu
D
,
Speranza
G
,
Esteban
E
,
Felip
E
,
Domine
M
, et al
Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer
.
J Clin Oncol
2020
;
38
:
1505
17
.
14.
Garon
EB
,
Hellmann
MD
,
Rizvi
NA
,
Carcereny
E
,
Leighl
NB
,
Ahn
MJ
, et al
Five-year overall survival for patients with advanced nonsmall-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study
.
J Clin Oncol
2019
;
37
:
2518
27
.
15.
Joseph
RW
,
Elassaiss-Schaap
J
,
Kefford
R
,
Hwu
W-J
,
Wolchok
JD
,
Joshua
AM
, et al
Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab
.
Clin Cancer Res
2018
;
24
:
4960
7
.
16.
Eggermont
AM
,
Chiarion-Sileni
V
,
Grob
JJ
,
Dummer
R
,
Wolchok
JD
,
Schmidt
H
, et al
Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy
.
N Engl J Med
2016
;
375
:
1845
55
.
17.
Zimmer
L
,
Livingstone
E
,
Hassel
JC
,
Fluck
M
,
Eigentler
T
,
Loquai
C
, et al
Adjuvant nivolumab plus ipilimumab or nivolumab monotherapy versus placebo in patients with resected stage IV melanoma with no evidence of disease (IMMUNED): a randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet
2020
;
395
:
1558
68
.
18.
Gray
JE
,
Villegas
A
,
Daniel
D
,
Vicente
D
,
Murakami
S
,
Hui
R
, et al
Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC-update from PACIFIC
.
J Thorac Oncol
2020
;
15
:
288
93
.
19.
Kelly
RJ
,
Ajani
JA
,
Kuzdzal
J
,
Zander
T
,
Van Cutsem
E
,
Piessen
G
, et al
Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer
.
N Engl J Med
2021
;
384
:
1191
203
.
20.
Demaria
S
,
Ng
B
,
Devitt
ML
,
Babb
JS
,
Kawashima
N
,
Liebes
L
, et al
Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated
.
Int J Radiat Oncol Biol Phys
2004
;
58
:
862
70
.
21.
Demaria
S
,
Kawashima
N
,
Yang
AM
,
Devitt
ML
,
Babb
JS
,
Allison
JP
, et al
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
34
.
22.
Park
SS
,
Dong
H
,
Liu
X
,
Harrington
SM
,
Krco
CJ
,
Grams
MP
, et al
PD-1 restrains radiotherapy-induced abscopal effect
.
Cancer Immunol Res
2015
;
3
:
610
9
.
23.
Deng
L
,
Liang
H
,
Burnette
B
,
Beckett
M
,
Darga
T
,
Weichselbaum
RR
, et al
Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice
.
J Clin Invest
2014
;
124
:
687
95
.
24.
Vanpouille-Box
C
,
Diamond
JM
,
Pilones
KA
,
Zavadil
J
,
Babb
JS
,
Formenti
SC
, et al
TGFbeta is a master regulator of radiation therapy-induced antitumor immunity
.
Cancer Res
2015
;
75
:
2232
42
.
25.
Abuodeh
Y
,
Venkat
P
,
Kim
S
. 
Systematic review of case reports on the abscopal effect
.
Curr Probl Cancer
2016
;
40
:
25
37
.
26.
Postow
MA
,
Callahan
MK
,
Barker
CA
,
Yamada
Y
,
Yuan
J
,
Kitano
S
, et al
Immunologic correlates of the abscopal effect in a patient with melanoma
.
N Engl J Med
2012
;
366
:
925
31
.
27.
Golden
EB
,
Chhabra
A
,
Chachoua
A
,
Adams
S
,
Donach
M
,
Fenton-Kerimian
M
, 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
.
28.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
29.
Wolchok
JD
,
Hoos
A
,
Day
S
,
Weber
JS
,
Hamid
O
,
Lebbé
C
, et al
Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria
.
Clin Cancer Res
2009
;
15
:
7412
20
.
30.
McBride
S
,
Sherman
E
,
Tsai
CJ
,
Baxi
S
,
Aghalar
J
,
Eng
J
, 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
2020
;
39
:
30
7
.
31.
Moreno
V
,
Gil-Martin
M
,
Johnson
M
,
Aljumaily
R
,
Lopez-Criado
MP
,
Northfelt
D
, et al
MA04.01 cemiplimab, a human monoclonal anti-PD-1, alone or in combination with radiotherapy: phase 1 NSCLC expansion cohorts
.
J Thorac Oncol
2018
;
13
:
S366
.
32.
Theelen
W
,
Peulen
HMU
,
Lalezari
F
,
van der Noort
V
,
de Vries
JF
,
Aerts
J
, et al
Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial
.
JAMA Oncol
2019
;
5
:
1276
82
.
33.
Luke
JJ
,
Lemons
JM
,
Karrison
TG
,
Pitroda
SP
,
Melotek
JM
,
Zha
Y
, et al
Safety and clinical activity of pembrolizumab and multisite stereotactic body radiotherapy in patients with advanced solid tumors
.
J Clin Oncol
2018
;
36
:
1611
8
.
34.
Luke
JJ
,
Onderdonk
BE
,
Bhave
SR
,
Karrison
T
,
Lemons
JM
,
Chang
P
, 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
44
.
35.
Curti
B
,
Crittenden
M
,
Seung
SK
,
Fountain
CB
,
Payne
R
,
Chang
S
, et al
Randomized phase II study of stereotactic body radiotherapy and interleukin-2 versus interleukin-2 in patients with metastatic melanoma
.
J Immunother Cancer
2020
;
8
;
e000773
.
36.
Mahmood
U
,
Bang
A
,
Chen
YH
,
Mak
RH
,
Lorch
JH
,
Hanna
GJ
, et al
A randomized phase 2 study of pembrolizumab with or without radiation in patients with recurrent or metastatic adenoid cystic carcinoma
.
Int J Radiat Oncol Biol Phys
2021
;
109
:
134
44
.
37.
Welsh
J
,
Menon
H
,
Chen
D
,
Verma
V
,
Tang
C
,
Altan
M
, et al
Pembrolizumab with or without radiation therapy for metastatic non-small cell lung cancer: a randomized phase I/II trial
.
J Immunother Cancer
2020
;
8
:
e001001
.
38.
Theelen
W
,
Chen
D
,
Verma
V
,
Hobbs
BP
,
Peulen
HMU
,
Aerts
J
, et al
Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials
.
Lancet Respir Med
2021
;
9
:
467
75
-X.
39.
Bauml
JM
,
Mick
R
,
Ciunci
C
,
Aggarwal
C
,
Davis
C
,
Evans
T
, et al
Pembrolizumab after completion of locally ablative therapy for oligometastatic non-small cell lung cancer: a phase 2 trial
.
JAMA Oncol
2019
;
5
:
1283
90
.
40.
Deng
L
,
Liang
H
,
Xu
M
,
Yang
X
,
Burnette
B
,
Arina
A
, et al
STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors
.
Immunity
2014
;
41
:
843
52
.
41.
Arina
A
,
Beckett
M
,
Fernandez
C
,
Zheng
W
,
Pitroda
S
,
Chmura
SJ
, et al
Tumor-reprogrammed resident T cells resist radiation to control tumors
.
Nat Commun
2019
;
10
:
3959
.
42.
Wray
J
,
Hawamdeh
RF
,
Hasija
N
,
Dagan
R
,
Yeung
AR
,
Lightsey
JL
, et al
Stereotactic body radiation therapy for oligoprogression of metastatic disease from gastrointestinal cancers: a novel approach to extend chemotherapy efficacy
.
Oncol Lett
2017
;
13
:
1087
94
.
43.
Reits
EA
,
Hodge
JW
,
Herberts
CA
,
Groothuis
TA
,
Chakraborty
M
,
Wansley
EK
, et al
Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy
.
J Exp Med
2006
;
203
:
1259
71
.
44.
Lhuillier
C
,
Rudqvist
N-P
,
Yamazaki
T
,
Zhang
T
,
Charpentier
M
,
Galluzzi
L
, et al
Radiotherapy-exposed CD8+ and CD4+ neoantigens enhance tumor control
.
J Clin Invest
2021
;
131
:
e138740
.
45.
Dhatchinamoorthy
K
,
Colbert
JD
,
Rock
KL
. 
Cancer immune evasion through loss of MHC class I antigen presentation
.
Front Immunol
2021
;
12
:
636568
.
46.
Schumacher
TN
,
Schreiber
RD
. 
Neoantigens in cancer immunotherapy
.
Science
2015
;
348
:
69
74
.
47.
Takeshima
T
,
Chamoto
K
,
Wakita
D
,
Ohkuri
T
,
Togashi
Y
,
Shirato
H
, et al
Local radiation therapy inhibits tumor growth through the generation of tumor-specific CTL: its potentiation by combination with Th1 cell therapy
.
Cancer Res
2010
;
70
:
2697
706
.
48.
Marciscano
AE
,
Ghasemzadeh
A
,
Nirschl
TR
,
Theodros
D
,
Kochel
CM
,
Francica
BJ
, et al
Elective nodal irradiation attenuates the combinatorial efficacy of stereotactic radiation therapy and immunotherapy
.
Clin Cancer Res
2018
;
24
:
5058
71
.
49.
Bourhis
J
,
Sire
C
,
Tao
Y
,
Martin
L
,
Alfonsi
M
,
Prevost
JB
, et al
LBA38 Pembrolizumab versus cetuximab, concomitant with radiotherapy (RT) in locally advanced head and neck squamous cell carcinoma (LA-HNSCC): results of the GORTEC 2015–01 “PembroRad” randomized trial
.
Ann Oncol
2020
;
31
:
S1142
1215
.
50.
Lee
NY
,
Ferris
RL
,
Psyrri
A
,
Haddad
RI
,
Tahara
M
,
Bourhis
J
, 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
62
.
51.
Ellsworth
SG
. 
Field size effects on the risk and severity of treatment-induced lymphopenia in patients undergoing radiation therapy for solid tumors
.
Adv Radiat Oncol
2018
;
3
:
512
9
.
52.
Chen
D
,
Verma
V
,
Patel
RR
,
Barsoumian
HB
,
Cortez
MA
,
Welsh
JW
. 
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
.
53.
Mell
LK
,
Schomas
DA
,
Salama
JK
,
Devisetty
K
,
Aydogan
B
,
Miller
RC
, et al
Association between bone marrow dosimetric parameters and acute hematologic toxicity in anal cancer patients treated with concurrent chemotherapy and intensity-modulated radiotherapy
.
Int J Radiat Oncol Biol Phys
2008
;
70
:
1431
7
.
54.
Park
S
,
Urm
S
,
Cho
H
. 
Analysis of biologically equivalent dose of stereotactic body radiotherapy for primary and metastatic lung tumors
.
Cancer Res Treat
2014
;
46
:
403
10
.
55.
Burnette
BC
,
Liang
H
,
Lee
Y
,
Chlewicki
L
,
Khodarev
NN
,
Weichselbaum
RR
, et al
The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity
.
Cancer Res
2011
;
71
:
2488
96
.
56.
Vanpouille-Box
C
,
Alard
A
,
Aryankalayil
MJ
,
Sarfraz
Y
,
Diamond
JM
,
Schneider
RJ
, et al
DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity
.
Nat Commun
2017
;
8
:
15618
.
57.
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
2018
;
9
:
5
11
.
58.
Formenti
SC
,
Rudqvist
NP
,
Golden
E
,
Cooper
B
,
Wennerberg
E
,
Lhuillier
C
, et al
Radiotherapy induces responses of lung cancer to CTLA-4 blockade
.
Nat Med
2018
;
24
:
1845
51
.
59.
Qian
JM
,
Yu
JB
,
Kluger
HM
,
Chiang
VL
. 
Timing and type of immune checkpoint therapy affect the early radiographic response of melanoma brain metastases to stereotactic radiosurgery
.
Cancer
2016
;
122
:
3051
8
.
60.
Samstein
R
,
Rimner
A
,
Barker
CA
,
Yamada
Y
. 
Combined immune checkpoint blockade and radiation therapy: timing and dose fractionation associated with greatest survival duration among over 750 treated patients
.
Int J Radiat Oncol Biol Phys
2017
;
99
:
S129
S30
.
61.
Chmura
SJ
,
Bestvina
CM
,
Karrison
TG
,
Jelinek
M
,
Juloori
A
,
Pointer
KB
, et al
Safety and efficacy of a randomized phase I trial to evaluate concurrent or sequential ipilimumab, nivolumab, and stereotactic body radiotherapy in patients with stage IV non-small cell lung cancer (COSINR study)
.
Int J Radiat Oncol Biol Phys
2020
;
108
:
S72
.
62.
Spiotto
M
,
Fu
YX
,
Weichselbaum
RR
. 
The intersection of radiotherapy and immunotherapy: mechanisms and clinical implications
.
Sci Immunol
2016
;
1
:
EAAG1266
.
63.
Chen
J
,
Cao
Y
,
Markelc
B
,
Kaeppler
J
,
Vermeer
JA
,
Muschel
RJ
. 
Type I IFN protects cancer cells from CD8+ T cell-mediated cytotoxicity after radiation
.
J Clin Invest
2019
;
129
:
4224
38
.
64.
Burnette
B
,
Weichselbaum
RR
. 
Radiation as an immune modulator
.
Semin Radiat Oncol
2013
;
23
:
273
80
.
65.
Facciabene
A
,
Motz
GT
,
Coukos
G
. 
T-regulatory cells: key players in tumor immune escape and angiogenesis
.
Cancer Res
2012
;
72
:
2162
71
.
66.
Medrek
C
,
Ponten
F
,
Jirstrom
K
,
Leandersson
K
. 
The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients
.
BMC Cancer
2012
;
12
:
306
.
67.
Bergenfelz
C
,
Larsson
AM
,
von Stedingk
K
,
Gruvberger-Saal
S
,
Aaltonen
K
,
Jansson
S
, et al
Systemic monocytic-MDSCs are generated from monocytes and correlate with disease progression in breast cancer patients
.
PLoS One
2015
;
10
:
e0127028
.
68.
Hellman
S
,
Weichselbaum
RR
. 
Oligometastases
.
J Clin Oncol
1995
;
13
:
8
10
.
69.
Ashworth
AB
,
Senan
S
,
Palma
DA
,
Riquet
M
,
Ahn
YC
,
Ricardi
U
, et al
An individual patient data metaanalysis of outcomes and prognostic factors after treatment of oligometastatic non-small-cell lung cancer
.
Clin Lung Cancer
2014
;
15
:
346
55
.
70.
Fong
Y
,
Fortner
J
,
Sun
RL
,
Brennan
MF
,
Blumgart
LH
. 
Clinical score for predicting recurrence after hepatic resection for metastatic colorectal cancer: analysis of 1001 consecutive cases
.
Ann Surg
1999
;
230
:
309
18
.
71.
Lussier
YA
,
Khodarev
NN
,
Regan
K
,
Corbin
K
,
Li
H
,
Ganai
S
, et al
Oligo- and polymetastatic progression in lung metastasis(es) patients is associated with specific microRNAs
.
PLoS One
2012
;
7
:
e50141
.
72.
Pitroda
SP
,
Khodarev
NN
,
Huang
L
,
Uppal
A
,
Wightman
SC
,
Ganai
S
, et al
Integrated molecular subtyping defines a curable oligometastatic state in colorectal liver metastasis
.
Nat Commun
2018
;
9
:
1793
.
73.
Matson
V
,
Fessler
J
,
Bao
R
,
Chongsuwat
T
,
Zha
Y
,
Alegre
ML
, et al
The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients
.
Science
2018
;
359
:
104
8
.
74.
Uribe-Herranz
M
,
Rafail
S
,
Beghi
S
,
Gil-de-Gómez
L
,
Verginadis
I
,
Bittinger
K
, et al
Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response
.
J Clin Invest
2019
;
130
:
466
79
.
75.
Yang
K
,
Hou
Y
,
Zhang
Y
,
Liang
H
,
Sharma
A
,
Zheng
W
, et al
Suppression of local type I interferon by gut microbiota-derived butyrate impairs antitumor effects of ionizing radiation
.
J Exp Med
2021
;
218
:
e20201915
.