Purpose:

Multisite stereotactic body radiotherapy followed by pembrolizumab (SBRT+P) has demonstrated safety in advanced solid tumors (ASTs). However, no studies have examined the relationships between irradiated tumor response, SBRT-induced tumor gene expression, and overall survival (OS).

Patients and Methods:

Patients with AST received SBRT (30–50 Gy in 3–5 fractions) to two to four metastases followed by pembrolizumab (200 mg i.v. every 3 weeks). SBRT was prescribed to a maximum tumor volume of 65 mL. Small metastases received the complete prescribed coverage (complete-Rx), while larger metastases received partial coverage (partial-Rx). Treated metastasis control (TMC) was defined as a lack of progression for an irradiated metastasis. Landmark analysis was used to assess the relationship between TMC and OS. Thirty-five biopsies were obtained from 24 patients: 19 pre-SBRT and 16 post-SBRT (11 matched) prior to pembrolizumab and were analyzed via RNA microarray.

Results:

Sixty-eight patients (139 metastases) were enrolled with a median follow-up of 10.4 months. One-year TMC was 89.5% with no difference between complete-Rx or partial-Rx. On multivariable analysis, TMC was independently associated with a reduced risk for death (HR, 0.36; 95% confidence interval, 0.17–0.75; P = 0.006). SBRT increased expression of innate and adaptive immune genes and concomitantly decreased expression of cell cycle and DNA repair genes in the irradiated tumors. Elevated post-SBRT expression of DNASE1 correlated with increased expression of cytolytic T-cell genes and irradiated tumor response.

Conclusions:

In the context of SBRT+P, TMC independently correlates with OS. SBRT impacts intratumoral immune gene expression associated with TMC. Randomized trials are needed to validate these findings.

This article is featured in Highlights of This Issue, p. 6399

Translational Relevance

In this phase I trial, irradiated tumor response was associated with overall survival. Large tumors treated with a partial radiation prescription dose achieved similar local control to smaller, completely irradiated tumors. The primary effect of combination stereotactic body radiotherapy (SBRT) with pembrolizumab was localized to irradiated metastases and not to systemic, nonirradiated metastases. SBRT induced innate and adaptive immune responses in tumors, and post-SBRT gene expression correlated with tumor control. Survival after multisite SBRT and pembrolizumab was associated with irradiated tumor response, and SBRT induced immune gene expression within the tumor microenvironment. These results support a randomized trial examining this approach.

Tumors with sustained responses to immunotherapy demonstrate a T cell–inflamed tumor microenvironment characterized by tumor-infiltrating lymphocytes and type I/II IFN gene expression profiles (1, 2). In contrast, tumors that fail to respond to immunotherapy frequently exhibit a non–T-cell-inflamed tumor phenotype. High-dose stereotactic body radiotherapy (SBRT) has been reported to promote immunogenic cell death through activation of innate and adaptive immunity orchestrated by IFN signaling (3, 4). These observations have led to several investigations exploring the use of radiation to enhance response to immunotherapy.

We previously reported the initial results of a clinical trial investigating the safety of treating metastases in multiple body sites with SBRT followed by pembrolizumab (5). Dose-limiting toxicities (DLTs) were seen in <10% of enrolled patients with advanced solid tumors. We observed a RECIST objective response rate (ORR) of 13% in a population of predominately programmed death ligand-1 (PD-L1)-low tumor types, which was similar to the ORR of PD-L1–positive patients with cancer treated with pembrolizumab alone on KEYNOTE-28 (6). Our finding indicates that the primary effect of combination SBRT+P was localized to irradiated metastases and not to systemic, nonirradiated metastases. In addition, we observed >90% control rates of irradiated metastases at 4 months even though many metastases received the prescription dose to only a portion of the tumor volume to minimize potential toxicity, which has historically resulted in poor local control. Therefore, a secondary analysis of this trial was performed to investigate the biological effects of SBRT as they relate to pembrolizumab response, we analyzed gene expression profiles of irradiated tumors and their impact on treated metastasis control (TMC). Here, we report outcomes based on local tumor response and tumor gene expression patterns.

Study design and participants

Patients with advanced solid tumors were enrolled between January 2016 and March 2017 (clinicaltrials.gov: NCT02608385; ref. 5). Study enrollment began after the institution internal review board approved the study, written informed consent was obtained, and pursued per Declaration of Helsinki. Data lock occurred on May 1, 2018. Patients received SBRT to at least two measurable metastases with each receiving 30–50 Gy over 3–5 fractions (5, 7). Metastases were prioritized on the basis of the largest size and/or those causing the most morbidity. The SBRT prescription dose was limited to a maximum of 65 mL such that tumors >65 mL received the prescription dose to part of the metastasis (partial-Rx). Otherwise, tumors received SBRT to the entire metastasis (complete-Rx). Pembrolizumab (200 mg i.v. every 3 weeks) began within 7 days following SBRT (SBRT+P). Tumor assessments were performed every 2–3 months using Response Evaluation for Solid Tumors 1.1 (RECIST) principles. Osseous and spinal metastases were considered controlled as part of the TMC measurement as per MD Anderson response criteria (8). Under FDA guidance, we attributed DLTs to the combination of SBRT+P therapy by anatomic organ system, and not to each specific therapy.

Tumor tissue analysis

Thirty-five tumor biopsies were collected prior to and/or within 7 days following SBRT, but prior to pembrolizumab, from 24 patients: 19 pre-SBRT and 16 post-SBRT (11 matched samples from the same tumor; Supplementary Table S1). RNA was extracted and analyzed using Affymetrix Human Clariom D microarrays. Computational gene expression deconvolution methods were used for genome-wide expression analyses following SBRT and correlated with irradiated tumor response. We utilized Ingenuity Pathway Analysis to predict the upstream regulators of pathways involved in the gene expression changes. See Supplementary Data for details regarding baseline PD-L1 expression, microarray data preprocessing, and detection of treatment-responsive gene expression changes. A TaqMan RT-PCR assay (Thermo Fisher Scientific) was used to validate DNASE1 gene expression utilizing an exon-spanning probe (Hs00173736_m1) overlapping the Clariom D transcript cluster TC1600011345.hg. Expression values for DNASE1 were normalized to the geometric mean of β-actin and 18S rRNA.

Statistical analysis

Baseline characteristics were compared between patients who received complete-Rx (to all treated lesions) and partial-Rx (to at least one of the lesions) using t tests and Pearson χ2 test for continuous and discrete variables, respectively. TMC was defined as a lack of progression for an irradiated metastasis (8–10), and duration of control was estimated using the Kaplan–Meier method. Death without irradiated tumor progression led to censoring at time of death (10). TMC and overall treatment response (i.e., CR, PR, SD, or PD) were evaluated using shared frailty Cox regression models and generalized estimating equations to account for correlation of individual metastasis responses within patients. At the first scan, patients who had a 30% or greater decrease (50% for osseous/spine) in the maximum transverse diameter of each irradiated metastasis were classified as responders, and those who had a 20% or greater growth (25% for osseous/spine) in the maximum transverse diameter of one or more irradiated metastases were nonresponders. Patients who did not fit either category were deemed to have stable disease (mixed responders). We used the Kaplan–Meier method to estimate progression-free survival (PFS) utilizing the time from SBRT to either progression or death from any cause and overall survival (OS) using the time from SBRT to death. Landmark analysis at 2 months was used to avoid a guarantee time bias when examining the association of early irradiated metastasis response with OS (11). We stratified outcomes using clinical, prognostic, and pathologic variables and compared differences using the log-rank test and Cox regression modeling. Baseline PD-L1 staining was available from 51 analyzable patients, and a cut-off of ≥1% was used to define PD-L1 positivity. P values <0.05 were considered statistically significant. Multivariable Cox regression analysis was performed for covariates with univariate P values <0.10. Data were analyzed using Stata software version 15.0 and JMP version 14 (SAS Institute Inc.).

Baseline characteristics and toxicity

Seventy-nine patients were enrolled and 11 patients were excluded from the analysis due to: not receiving SBRT (n = 3); refusing pembrolizumab (n = 3); or lost to follow-up (n = 5). The overall median follow-up time (n = 68) at data lock was 10.4 months (range, 2.3–24.1), and was 33.4 months (range, 15.3–42.5) for the 17 patients alive past the data lock. Sixty-eight patients with 26 different, nonradiosensitive and/or generally non-PD-1 responsive, solid tumor types who received SBRT to 139 metastases were analyzed (Supplementary Table S2). Patients had previously received a median of 5 lines of systemic therapy, and 3 had received checkpoint therapy. Table 1 summarizes baseline patient-level (n = 68) characteristics stratified on the basis of the receipt of partial-Rx to at least one metastasis. A representative example of partial-Rx versus complete-Rx treatment is shown in Supplementary Fig. S1. In 50 patients (73%), 102 metastases received the radiation prescription dose to the entire tumor volume (complete-Rx), whereas in 18 patients (26%) at least one metastasis (21 of 37 metastases) received the prescription dose to less than the entire tumor volume (partial-Rx). There were no differences in tumor histology (P = 0.19), baseline PD-L1 status (P = 0.92), or prior lines of therapy (P = 0.54) between patients stratified by who received all complete-Rx versus at least one partial-Rx. There were only six DLTs: six (all metastases complete-Rx), 0 (at least one metastasis partial-Rx), for an overall rate of 8.8%.

Table 1.

Patient-level baseline and treatment characteristics.

Complete-RxPartial-Rx
50 Pts18 Pts
Patient characteristic102 Mets37 Mets
Partial-tumor SBRT (metastasis-level) 0% (0/102 Mets) 56.8% (21/37 Mets) 
Follow up (mo) mean (SD) 10.8 (6.5) 8.9 (6.0) 
Age (yrs), mean (SD) 60.5 (13.25) 61.0 (15.9) 
Gender 
 F 28 (55%) 13 (72%) 
 M 22 (44%) 5 (28%) 
ECOG performance status 
 0 25 (50%) 7 (39%) 
 1 25 (50%) 11 (61%) 
Smoking status 
 Current 2 (4%) 1 (6%) 
 Former 23 (46%) 7 (39%) 
 Never 25 (50%) 10 (56%) 
Baseline albumin, median (range) 4.0 (2.4–4.5) 4.0 (3.2–4.4) 
Primary cancer histology 
 Other 18 (36%) 9 (50%) 
 Ovarian/fallopian tube 7 (14%) 2 (11%) 
 Non–small cell lung 5 (10%) 1 (6%) 
 Breast 5 (10%) 1 (6%) 
 Cholangiocarcinoma 2 (4%) 4 (22%) 
 Endometrial 6 (12%) 0 (0%) 
 Colorectal 3 (6%) 1 (6%) 
 Head and neck 4 (8%) 0 (0%) 
PD-L1 status   
 <1% 19 (39%) 7 (39%) 
 >1% 19 (39%) 6 (33%) 
 Missing 12 (24%) 5 (28%) 
No. of prior treatments, median (range) 5 (0–13) 3 (0–10) 
Prior immunotherapy? 
 No 47 (94%) 18 (100%) 
 Yes 3 (6%) 0 (0%) 
Total RECIST lesions followed (mean, SD) 3.4 (1.4) 3.2 (1.1) 
Sites treated with SBRT 
 2 metastases 48 pts (96%) 17 pts (94%) 
 3 metastases 2 pts (4%) 1 pts (6%) 
Cycles of pembrolizumab, median (range) 4 (1–27) 5 (1–34) 
Complete-RxPartial-Rx
50 Pts18 Pts
Patient characteristic102 Mets37 Mets
Partial-tumor SBRT (metastasis-level) 0% (0/102 Mets) 56.8% (21/37 Mets) 
Follow up (mo) mean (SD) 10.8 (6.5) 8.9 (6.0) 
Age (yrs), mean (SD) 60.5 (13.25) 61.0 (15.9) 
Gender 
 F 28 (55%) 13 (72%) 
 M 22 (44%) 5 (28%) 
ECOG performance status 
 0 25 (50%) 7 (39%) 
 1 25 (50%) 11 (61%) 
Smoking status 
 Current 2 (4%) 1 (6%) 
 Former 23 (46%) 7 (39%) 
 Never 25 (50%) 10 (56%) 
Baseline albumin, median (range) 4.0 (2.4–4.5) 4.0 (3.2–4.4) 
Primary cancer histology 
 Other 18 (36%) 9 (50%) 
 Ovarian/fallopian tube 7 (14%) 2 (11%) 
 Non–small cell lung 5 (10%) 1 (6%) 
 Breast 5 (10%) 1 (6%) 
 Cholangiocarcinoma 2 (4%) 4 (22%) 
 Endometrial 6 (12%) 0 (0%) 
 Colorectal 3 (6%) 1 (6%) 
 Head and neck 4 (8%) 0 (0%) 
PD-L1 status   
 <1% 19 (39%) 7 (39%) 
 >1% 19 (39%) 6 (33%) 
 Missing 12 (24%) 5 (28%) 
No. of prior treatments, median (range) 5 (0–13) 3 (0–10) 
Prior immunotherapy? 
 No 47 (94%) 18 (100%) 
 Yes 3 (6%) 0 (0%) 
Total RECIST lesions followed (mean, SD) 3.4 (1.4) 3.2 (1.1) 
Sites treated with SBRT 
 2 metastases 48 pts (96%) 17 pts (94%) 
 3 metastases 2 pts (4%) 1 pts (6%) 
Cycles of pembrolizumab, median (range) 4 (1–27) 5 (1–34) 

Note: Data are n (%) unless otherwise stated.

Abbreviations: Complete-Rx, prescription SBRT dose to the entire metastasis volume; Mets, treated metastases; Partial-Rx, prescription SBRT dose to part of the metastasis volume; pts, patients; SBRT, stereotactic body radiotherapy; SD, standard deviation.

TMC

The 1-year Kaplan–Meier TMC rate was 89.5% (Fig. 1A) consistent with other reports following complete volumetric tumor coverage with high-dose SBRT (12–14). However, 21 of the 139 treated metastases received partial-Rx SBRT+P. The partial-Rx treated metastases (Supplementary Table S4) were larger at baseline (mean volume: 157.6 mL vs. 12.8 mL, P < 0.001) and more likely to be located in the abdomen or pelvis (P < 0.001; Supplementary Table S3). We observed no significant difference in TMC between complete-Rx versus partial-Rx SBRT+P (Fig. 2) despite differences in the metastasis target volume receiving at least 95% of prescribed radiation dose (median volume for partial-Rx vs. complete-Rx: 67.2% vs. 100%); however, the low number of events limits statistical power to draw firm conclusions. Moreover, there were no significant differences in TMC when stratified by baseline PD-L1 status (P = 0.81; Fig. 1B), or tumor volume irradiated (P = 0.86; Fig. 1C). In contrast to typical SBRT plans (15), we observed similar TMC rates independent of the metastasis size or volume receiving the complete prescription dose when delivered prior to pembrolizumab.

Figure 1.

TMC was independent of PD-L1 status, tumor size, and tumor histology. Kaplan–Meier estimates of overall TMC (n = 139; A). B, TMC by PD-L1 status (n = 105). C, TMC by treated metastasis quartile volume prior to SBRT. In B and C, P values were determined using shared-frailty Cox and then fit to a flexible parametric model to generate figures. Values along the x-axis are the number of metastases at risk among 68 evaluable patients. NSCLC, non–small cell lung cancer; cholangio, cholangiocarcinoma.

Figure 1.

TMC was independent of PD-L1 status, tumor size, and tumor histology. Kaplan–Meier estimates of overall TMC (n = 139; A). B, TMC by PD-L1 status (n = 105). C, TMC by treated metastasis quartile volume prior to SBRT. In B and C, P values were determined using shared-frailty Cox and then fit to a flexible parametric model to generate figures. Values along the x-axis are the number of metastases at risk among 68 evaluable patients. NSCLC, non–small cell lung cancer; cholangio, cholangiocarcinoma.

Close modal
Figure 2.

Treatment outcomes stratified by prescription radiotherapy dose volume. TMC by prescription SBRT dose covering the entire metastasis (cRx) compared with partial metastasis coverage (pRx; n = 139). P values were determined using shared-frailty Cox regression and then fit to a flexible parametric model. Values along the x-axis are the number of metastases at risk among 68 evaluable patients.

Figure 2.

Treatment outcomes stratified by prescription radiotherapy dose volume. TMC by prescription SBRT dose covering the entire metastasis (cRx) compared with partial metastasis coverage (pRx; n = 139). P values were determined using shared-frailty Cox regression and then fit to a flexible parametric model. Values along the x-axis are the number of metastases at risk among 68 evaluable patients.

Close modal

Association of SBRT+P response and survival

We analyzed treatment responses of individual metastases and at the patient level (Supplementary Fig. S2). Seven metastases (5%) had a complete response and 42 (30.2%) metastases had a partial response for an overall treated metastasis response rate of 35.2%. Eighty-one metastases (58.3%) were stable with nine (6.5%) demonstrating immediate progression following SBRT+P. No differences in SBRT+P response were observed between patients who received complete-Rx versus partial-Rx SBRT+P (P = 0.15). Partial-Rx SBRT+P did not affect local treatment response (P = 0.35; Supplementary Fig. S3A), nor did tumor volume (P = 0.44; Supplementary Fig. S3B), baseline PD-L1 status (P = 0.12; Supplementary Fig. S3C), or histology (P = 0.41; Supplementary Fig. S3D). Supplementary Table S5 summarizes all factors analyzed in this regression model.

The ORR of the nonirradiated metastases was 13.2% as reported previously. The median PFS was 3.1 months (95% CI 2.9–3.5; Supplementary Fig. S4A) and the median OS was 9.9 months (95% CI, 7.0–13.5; Supplementary Fig. S4B). No covariates were predictive of OS on univariate Cox regression (Supplementary Table S6). Also, no significant differences in OS were observed between patients stratified by partial-Rx status (Supplementary Fig. S5A) or baseline PD-L1 expression (Supplementary Fig. S5B). These findings demonstrated that TMC to SBRT+P occurred independent of baseline or treatment characteristics.

We analyzed OS based on irradiated tumor response (landmark analysis). This is supported by a direct relationship between the change in size of unirradiated lesions as a function of response of irradiated lesions (Supplementary Fig. S6). Only one patient died prior to their first scan, and of the 67 patients who survived beyond the 2-month landmark time, the median OS was 3.5 months for nonresponders (95% CI, 2.3–9.6 months), 9.0 months (95% CI, 6.5–12.9 months) for mixed responders, and 17.8 months (95% CI, 7.7–37.7 months) for responders (P = 0.0007; Fig. 3). Moreover, on post-landmark Cox regression, a larger irradiated tumor response, coded as an ordinal variable (0 = no response, 1 = mixed response, 2 = response), decreased the risk for death (HR, 0.44; 95% CI, 0.23–0.85; P = 0.015). These results demonstrate that patients whose tumors exhibited a local response to the combination of SBRT+P were more likely to experience extended survival.

Figure 3.

Kaplan–Meier OS stratified by SBRT response. OS stratified by irradiated tumor response (responders, mixed responders, and nonresponders) from those that survived to the 2-month landmark time (n = 67). HR 0.44; 95% CI, 0.23–0.85; P = 0.015. SBRT, stereotactic body radiation therapy; HR, hazard ratio; mo, month.

Figure 3.

Kaplan–Meier OS stratified by SBRT response. OS stratified by irradiated tumor response (responders, mixed responders, and nonresponders) from those that survived to the 2-month landmark time (n = 67). HR 0.44; 95% CI, 0.23–0.85; P = 0.015. SBRT, stereotactic body radiation therapy; HR, hazard ratio; mo, month.

Close modal

Gene expression analysis

We investigated radiation-induced gene expression changes associated with local response in serial biopsies acquired from irradiated metastases. First, we performed a genome-wide analysis to characterize gene expression changes following SBRT from 11 patients with both pre- and post-SBRT biopsies (matched samples). SBRT increased the expression of pathways enriched for innate and adaptive immunity, such as B-cell development, antigen presentation, and dendritic cell maturation, while concomitantly reducing the expression of pathways involved in G2–M cell-cycle progression and DNA damage repair processes (Fig. 4A). Immune genes in the activated pathways were predicted to be regulated by type I/II IFN signaling, whereas downregulation of cell cycle and DNA repair processes were predicted to be in response to RAD21 and CDKN2A signaling pathways (Fig. 4A; ref. 16). Furthermore, the response to SBRT was associated with suppression of histone subunits and genes mediating mitosis and cell division, such as CDC20, CENPM, MCM5, and PRC1 (Supplementary Fig. S7). In contrast, SBRT led to the induction of CXCR3, which has shown to regulate leukocyte trafficking and to promote Th1 and CD8+ effector cell migration into inflamed tissues (17, 18). These results are consistent with preclinical models demonstrating that high-dose SBRT impacts pathways associated with cell cycle, DNA repair, and antitumor immunity.

Figure 4.

Gene expression analysis of irradiated tumors following SBRT and association with irradiated tumor response. A, Enriched canonical pathways and predicted upstream activators following SBRT. P values were determined using Fisher exact tests. B, Association of irradiated tumor response with post-SBRT gene expression. Pearson correlation coefficients plotted against statistical significance of correlation for all protein-coding genes. C, Change in intratumoral cytolytic gene expression (PRF1 + GZMA expression) in relation to change in DNASE1 (top) or TREX1 (bottom) expression after SBRT. Δ > 0 indicates increased expression, while Δ < 0 indicates decreased expression. Tumors were dichotomized as low or high expressors based on the median expression value of DNASE1 and TREX1. P values were determined using Mann–Whitney U tests. D, Change in irradiated tumor size ([(posttreatment largest diameter/pretreatment largest diameter) -1] * 100%) after SBRT + pembrolizumab as a function of post-SBRT DNASE1 and TREX1 expressions in matched tumor specimens. Tumors were dichotomized as low or high expressors based on the median expression value of DNASE1 and TREX1. P values were determined using Mann–Whitney U tests. PD, progressive disease; SD, stable disease; CR/PR, complete response/partial response based on RECIST criteria.

Figure 4.

Gene expression analysis of irradiated tumors following SBRT and association with irradiated tumor response. A, Enriched canonical pathways and predicted upstream activators following SBRT. P values were determined using Fisher exact tests. B, Association of irradiated tumor response with post-SBRT gene expression. Pearson correlation coefficients plotted against statistical significance of correlation for all protein-coding genes. C, Change in intratumoral cytolytic gene expression (PRF1 + GZMA expression) in relation to change in DNASE1 (top) or TREX1 (bottom) expression after SBRT. Δ > 0 indicates increased expression, while Δ < 0 indicates decreased expression. Tumors were dichotomized as low or high expressors based on the median expression value of DNASE1 and TREX1. P values were determined using Mann–Whitney U tests. D, Change in irradiated tumor size ([(posttreatment largest diameter/pretreatment largest diameter) -1] * 100%) after SBRT + pembrolizumab as a function of post-SBRT DNASE1 and TREX1 expressions in matched tumor specimens. Tumors were dichotomized as low or high expressors based on the median expression value of DNASE1 and TREX1. P values were determined using Mann–Whitney U tests. PD, progressive disease; SD, stable disease; CR/PR, complete response/partial response based on RECIST criteria.

Close modal

We then examined 16 patients with post-SBRT (pre-pembrolizumab) biopsies to determine whether the same changes in gene expression were associated with irradiated tumor response. Among the 30 top-ranked genes in terms of correlation with irradiated tumor response, we found that elevated expression of immune genes, including DNASE1 (P = 0.00029) and CCR10 (P = 0.00069), were associated with increased irradiated tumor response, while elevated expression of TGFBR3L (P = 0.0014) and SIGLEC15 (P < 0.0001) were strongly associated with decreased tumor response (Fig. 4B; refs. 19–21).

DNASE1 has been shown to promote DNA degradation in the response to ionizing radiation, which may influence activation of cGAS-STING signaling in tumor cells (3, 22–24). Increased DNASE1 levels following SBRT were associated with concomitant increases in the expression of cytolytic T-cell genes (granzyme A and perforin; ref. 25; Fig. 4C) and a 2.1-fold improved local tumor response (Fig. 4D). RT-PCR analysis showed that increased DNASE1 expression following SBRT was associated with a reduction in size of irradiated metastases in the response to SBRT+P. Furthermore, the patient with the highest DNASE1 expression exhibited the greatest reduction in tumor burden (Supplementary Fig. S8). In contrast, increased TREX1 (DNase III) expression levels following SBRT were associated with decreased cytolytic gene expression (Fig. 4C) in concert with a 2.4-fold reduced local tumor response (Fig. 4D; ref. 26). These findings indicated that SBRT induces gene expression changes in immune pathways, which are associated with local tumor response in the presence of adjuvant pembrolizumab.

We initially reported the safety of multisite SBRT+P in patients with advanced solid malignancies (5). Here, we report that ORR, PFS, and OS did not differ between patients who received complete-Rx versus partial-Rx SBRT to multiple sites in the context of pembrolizumab. Also, clinical responses at the irradiated site could be induced without irradiation of an entire metastasis. Moreover, gene expression changes correlating with SBRT+P treatment response were enriched for innate and adaptive immune pathways. Finally, we found that the local response of irradiated metastases to SBRT+P was associated with OS.

Because TMC following radiotherapy in the absence of immunotherapy is a function of the overall dose, the dose per fraction, and the overall time during which the radiation is given (12–15, 27–29), we examined the relationship between irradiated tumor volume and radiation dose. Previous studies have established the minimum dose required for local control following SBRT alone is 36 Gy in 3 fractions (10, 30). While in our partial-Rx cohort, the median value for tumor minimum dose was 9 Gy in 3 fractions, which is expected to have poor rates of local control (31–33). Moreover, partial-Rx treatment increased normal tissue sparing compared with the complete-Rx group (see Supplementary Methods for biological effective dose calculations). Thus, partial-Rx treatment in combination with pembrolizumab is feasible and should be examined further.

The correlation of survival with the local response of the irradiated tumors, and not with baseline patient characteristics suggesting that multisite SBRT can limit progression of existing metastases while also augmenting antitumor immune responses to potentially improve outcomes in metastatic patients treated with pembrolizumab. Cytoreduction through radiation may be important, particularly given the clinical observation indicating that patients with lower disease burden treated on prospective immune checkpoint therapy trials have improved outcomes (34).

We also found that clinical responses could be induced, regardless of tumor PD-L1 status, without irradiation of an entire metastasis and also resulted in less toxicity. Despite the partial treatment of these metastases, their inherently larger size, and the reduced overall dose to the metastasis, we did not detect any difference in TMC when compared with tumors that received the prescribed radiation dose to the entirety of the lesion. Specifically, 15/16 (94%) metastases in our partial-Rx group responded despite receiving lower radiation doses. On the basis of prior studies using single modality SBRT prescribed to the entire metastasis volume in which 25.5 Gy was prescribed in 3 fractions, we expected few metastases receiving partial-Rx to have radiographic response (35). We propose the response seen in our study is likely multifactorial and related to both cytoreduction and local immune enhancement as previously demonstrated in preclinical models when stereotactic radiation was combined with immune checkpoint therapy (36). However, we await further prospective evidence to further understand the contribution of these two potential mechanisms. Of note, the high TMC rate seen was not associated with increased toxicities given that no DLTs occurred in the partial-Rx group. Collectively, these data suggest that a multisite SBRT approach, including partial-Rx, followed by immunotherapy may have broad applicability by enhancing response and limiting adjacent organ toxicity in patients with advanced cancer.

Consistent with preclinical evidence and the recent ORIOLE phase II randomized trial, we observed that SBRT induced both innate and adaptive immune pathways, while concomitantly decreasing genes involved in cell cycle and DNA damage repair pathways (3, 4, 37). We found overexpression of the DNase I (DNASE1) endonuclease following SBRT was associated with favorable irradiated tumor response to SBRT+P, whereas overexpression of the DNase III (TREX1) exonuclease was associated with unfavorable local tumor response. These findings suggest that differential DNA degradation in the response to SBRT might contribute to distinct treatment responses to SBRT+P. Moreover, overexpression of SIGLEC15, recently described as an important immune suppressor in the context of checkpoint blockade (38), following SBRT emerged as potential negative regulator of SBRT+P response in our analysis. Taken together, our finding that there is no difference in tumor control whether patients receive partial-Rx or complete-Rx, our work suggests that the ablative function of radiation may not be the only aspect of radiation, which should be considered. In fact, our work is novel in suggesting that the other aspects of the biological response to radiation (which are not accounted for in the α/β ratio) may be just as important.

While there is generally a lack of consensus regarding the optimal radiation dose (thought to be in the range of 8–12 Gy per fraction; refs. 10, 26, 30) and the degree of tumor coverage required to potentiate systemic immunity (39). In our study, we used higher radiation doses per treatment, and although we found an inverse relationship between treatment response and the post-SBRT expression of TREX1, we also observed that the majority of irradiated tumors responded to treatment and exhibited high levels of post-SBRT immune gene expression. We recognize that even in the context of higher fraction irradiation some portion of the tumor receives a lower dose, especially in partial-Rx tumors. However, when we investigated the percentage of the tumor that received 8–12 Gy we did not find an association with irradiated tumor or overall patient response. These findings suggest that, in humans, the optimal radiation dose and volume to synergize with immunotherapy remains undefined.

While these analyses were generated from a prospective clinical trial, there are several limitations of our investigation. For example, only 18 patients and 21 (of 37) metastases were treated with partial-Rx SBRT+P. Additional prospectively treated patients are needed to confirm the favorable local tumor response with partial-Rx SBRT+P. The single-arm nature and heterogeneous patient population within the phase I trial also have limitations and randomized evaluation within a specific disease setting is required. Regarding gene expression analysis, RNA isolation was prioritized from tumor biopsies limiting the availability of additional tissue for exploratory analyses.

Multisite SBRT, including partial-Rx SBRT to large lesions, in combination with immunotherapy may provide a benefit to the irradiated metastases, irrespective of PD-L1 status. This treatment paradigm is feasible and clinically available to patients in the standard-of-care setting. Furthermore, the association of treatment response to radiation (TMC) with OS suggests a potential clinical and radiographic biomarker that is easily evaluable. Finally, gene expression profiling changes should similarly be assessed in other studies using a range of radiation doses to identify the optimal dose and schedule for immune potentiation.

J.J. Luke reports grants from Merck (Merck Investigator Sponsored Program) during the conduct of the study; scientific advisory board relationships (no stock) with 7 Hills, Spring bank (stock) Actym, Alphamab Oncology, Arch Oncology, Kanaph, Mavu, Onc.AI, Pyxis, Tempest; consultation with compensation for Abbvie, Aligos, Array, Bayer, Bristol-Myers Squibb, Checkmate, Cstone, Eisai, EMD Serono, KSQ, Janssen, Merck, Mersana, Novartis, Partner, Pfizer, RefleXion, Regeneron, Ribon, Rubius, Silicon, Tesaro, Werewolf, Xilio, Xencor; research support from (all to institution for clinical trials unless noted) AbbVie, Agios (IIT), Array (IIT), Astellas, Bristol-Myers Squibb (IIT & industry), Corvus, EMD Serono, Immatics, Incyte, Kadmon, Macrogenics, Merck, Spring bank, Tizona, Xencor; travel compensation from Bristol-Myers Squibb, Janssen, Mersana, Pyxis; and patents from (both provisional) Serial #15/612,657 (Cancer Immunotherapy), PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof). J.W. Moroney reports other from Clovis (honoraria) outside the submitted work. J.D. Patel reports personal fees from AstraZeneca (advisor) and Takeda (advisor) and grants and personal fees from BMS (advisor; institution) outside the submitted work. P.A. Ott reports grants and personal fees from Neon Therapeutics, Merck, CytomX, Roche/Genentech, Celldex; grants from BMS, ArmoBiosciences, Pfizer, AstraZeneca/MedImmune; and personal fees from Novartis and Array outside the submitted work. G.F. Fleming reports other from Merck during the conduct of the study; personal fees from GSK (also site PI for industry sponsored study); other from corcept (supplied drug for mouse studies. Also am site PI for industry sponsored study); Abbvie (site PI for industry sponsored study), Roche (site PI for industry sponsored study), 47 inc (site PI for industry sponsored study), Iovance (site PI for industry sponsored study), Syros (site PI for industry sponsored study), Merck (site PI for industry sponsored study), Sanofi (site PI for industry sponsored study), Sermonix (site PI for industry sponsored study), Compugen (site PI for industry sponsored study), Incyte (site PI for industry sponsored study), Eisai (site PI for industry sponsored study), and plexxicon (site PI for industry sponsored study) outside the submitted work. T.F. Gajewski reports grants from Merck during the conduct of the study and personal fees from Merck (advisory board) outside the submitted work. R.R. Weichselbaum reports other from Boost Therapeutics (stock and other ownership interests), Immvira LLC (stock and other ownership interests), RefleXion Pharmaceuticals (stock and other ownership interests), Coordination Pharmaceuticals Inc. (stock and other ownership interests), Magi Therapeutics (stock and other ownership interests), and Oncosenescence (stock and other ownership interests) during the conduct of the study; grants from Varian, Regeneron, AstraZeneca (travel), Boehringer Ingelheim LTD (travel), and Merck Serono S.A. (travel) outside the submitted work; in addition, R.R. Weichselbaum has a patent for Methods and Kits for Diagnosis and Triage of Patients with Colorectal Liver Metastases pending and has served in a consulting or advisory role for Aettis Inc., Astrazeneca, Coordination Pharmaceuticals, Genus, Merck Serono S.A., Nano proteagen, NKMax America Inc, Shuttle Pharmaceuticals, Highlight Therapeutics, S.L. S.J. Chmura reports grants from Merck during the conduct of the study, as well as other from AStellas Pharma (wife employment) and grants from RefleXion (trials support) and BMS (trial support) outside the submitted work. No potential conflicts of interest were disclosed by the other authors.

J.J. Luke: Conceptualization, methodology, writing-review and editing. B.E. Onderdonk: Software, formal analysis, investigation, writing-original draft, writing-review and editing. S.R. Bhave: Conceptualization, software, writing-original draft. T. Karrison: Data curation, software, formal analysis, supervision, writing-review and editing. J.M. Lemons: Conceptualization, writing-review and editing. P. Chang: Data curation, methodology. Y. Zha: Data curation, methodology. T. Carll: Data curation, methodology. T. Krausz: Data curation, methodology. L. Huang: Data curation, methodology. C. Martinez: Data curation, software, methodology. L.A. Janisch: Methodology, project administration. R.D. Hseu: Methodology, project administration. J.W. Moroney: Methodology, writing-original draft. J.D. Patel: Conceptualization, supervision, investigation, writing-original draft, writing-review and editing. N.N. Khodarev: Methodology. J.K. Salama: Methodology, writing-original draft. P.A. Ott: Methodology, writing-original draft. G.F. Fleming: Supervision, writing-original draft, writing-review and editing. T.F. Gajewski: Methodology, writing-original draft. R.R. Weichselbaum: Supervision, methodology, writing-original draft, writing-review and editing. S.P. Pitroda: Conceptualization, formal analysis, supervision, methodology, writing-original draft, writing-review and editing. S.J. Chmura: Conceptualization, formal analysis, investigation, methodology, writing-original draft, writing-review and editing.

We thank the patients and their families who participated in this study. We thank Bernadette Libao, Jill Stetkevych, and Lauren Wall for logistical support of the trial. We thank Merck & Co., Inc. for drug and grant support. J.J. Luke and S.J. Chmura had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Trujillo
JA
,
Sweis
RF
,
Bao
R
,
Luke
JJ
. 
T cell-inflamed versus non-T cell-inflamed tumors: a conceptual framework for cancer immunotherapy drug development and combination therapy selection
.
Cancer Immunol Res
2018
;
6
:
990
1000
.
2.
Ayers
M
,
Lunceford
J
,
Nebozhyn
M
,
Murphy
E
,
Loboda
A
,
Kaufman
DR
, et al
IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade
.
J Clin Invest
2017
;
127
:
2930
40
.
3.
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
.
4.
Liang
H
,
Deng
L
,
Hou
Y
,
Meng
X
,
Huang
X
,
Rao
E
, et al
Host STING-dependent MDSC mobilization drives extrinsic radiation resistance
.
Nat Commun
2017
;
8
:
1736
.
5.
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
.
6.
Ott
PA
,
Bang
Y-J
,
Berton-Rigaud
D
,
Elez
E
,
Pishvaian
MJ
,
Rugo
HS
, et al
Safety and antitumor activity of pembrolizumab in advanced programmed death ligand 1-positive endometrial cancer: results from the KEYNOTE-028 study
.
J Clin Oncol
2017
;
35
:
2535
41
.
7.
Salama
JK
,
Hasselle
MD
,
Chmura
SJ
,
Malik
R
,
Mehta
N
,
Yenice
KM
, et al
Stereotactic body radiotherapy for multisite extracranial oligometastases: final report of a dose escalation trial in patients with 1 to 5 sites of metastatic disease
.
Cancer
2012
;
118
:
2962
70
.
8.
Costelloe
CM
,
Chuang
HH
,
Madewell
JE
,
Ueno
NT
. 
Cancer response criteria and bone metastases: RECIST 1.1, MDA and PERCIST
.
J Cancer
2010
;
1
:
80
92
.
9.
Milano
MT
,
Katz
AW
,
Zhang
H
,
Okunieff
P
. 
Oligometastases treated with stereotactic body radiotherapy: long-term follow-up of prospective study
.
Int J Radiat Oncol Biol Phys
2012
;
83
:
878
86
.
10.
Wong
AC
,
Watson
SP
,
Pitroda
SP
,
Son
CH
,
Das
LC
,
Stack
ME
, et al
Clinical and molecular markers of long-term survival after oligometastasis-directed stereotactic body radiotherapy (SBRT)
.
Cancer
2016
;
122
:
2242
50
.
11.
Anderson
JR
,
Cain
KC
,
Gelber
RD
. 
Analysis of survival by tumor response
.
J Clin Oncol
1983
;
1
:
710
9
.
12.
Mahadevan
A
,
Blanck
O
,
Lanciano
R
,
Peddada
A
,
Sundararaman
S
,
D'Ambrosio
D
, et al
Stereotactic Body Radiotherapy (SBRT) for liver metastasis - clinical outcomes from the international multi-institutional RSSearch® Patient Registry
.
Radiat Oncol
2018
;
13
:
26
.
13.
McCammon
R
,
Schefter
TE
,
Gaspar
LE
,
Zaemisch
R
,
Gravdahl
D
,
Kavanagh
B
. 
Observation of a dose-control relationship for lung and liver tumors after stereotactic body radiation therapy
.
Int J Radiat Oncol Biol Phys
2009
;
73
:
112
18
.
14.
Ohri
N
,
Tomé
WA
,
Méndez Romero
A
,
Miften
M
,
Ten Haken
RK
,
Dawson
LA
, et al
Local control after stereotactic body radiation therapy for liver tumors
.
Int J Radiat Oncol Biol Phys
2018
;
S0360-3016(17)34525-X
.
15.
Yamada
Y
,
Katsoulakis
E
,
Laufer
I
,
Lovelock
M
,
Barzilai
O
,
McLaughlin
LA
, et al
The impact of histology and delivered dose on local control of spinal metastases treated with stereotactic radiosurgery
.
Neurosurg Focus
2017
;
42
:
E6
.
16.
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
.
17.
Harlin
H
,
Meng
Y
,
Peterson
AC
,
Zha
Y
,
Tretiakova
M
,
Slingluff
C
, et al
Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment
.
Cancer Res
2009
;
69
:
3077
85
.
18.
Spranger
S
,
Dai
D
,
Horton
B
,
Gajewski
TF
. 
Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy
.
Cancer Cell
2017
;
31
:
711
23.e4
.
19.
Formenti
SC
,
Lee
P
,
Adams
S
,
Goldberg
JD
,
Li
X
,
Xie
MW
, et al
Focal irradiation and systemic TGFβ blockade in metastatic breast cancer
.
Clin Cancer Res
2018
;
24
:
2493
504
.
20.
Barcellos-Hoff
MH
. 
Radiation-induced transforming growth factor beta and subsequent extracellular matrix reorganization in murine mammary gland
.
Cancer Res
1993
;
53
:
3880
6
.
21.
Barcellos-Hoff
MH
,
Cucinotta
FA
. 
New tricks for an old fox: impact of TGFβ on the DNA damage response and genomic stability
.
Sci Signal
2014
;
7
:
re5
.
22.
Woo
S-R
,
Fuertes
MB
,
Corrales
L
,
Spranger
S
,
Furdyna
MJ
,
Leung
MYK
, et al
STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors
.
Immunity
2014
;
41
:
830
42
.
23.
Ahn
J
,
Gutman
D
,
Saijo
S
,
Barber
GN
. 
STING manifests self DNA-dependent inflammatory disease
.
Proc Natl Acad Sci U S A
2012
;
109
:
19386
91
.
24.
Weichselbaum
RR
,
Liang
H
,
Deng
L
,
Fu
Y-X
. 
Radiotherapy and immunotherapy: a beneficial liaison?
Nat Rev Clin Oncol
2017
;
14
:
365
79
.
25.
Rooney
MS
,
Shukla
SA
,
Wu
CJ
,
Getz
G
,
Hacohen
N
. 
Molecular and genetic properties of tumors associated with local immune cytolytic activity
.
Cell
2015
;
160
:
48
61
.
26.
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
.
27.
Duijm
M
,
Nuyttens
J
,
Schillemans
W
,
Antonisse
I
,
Hoogeman
MS
,
Verhoef
C
, et al
The contribution of underdosage to the local control for central tumors in the lung treated with stereotactic radiation therapy
.
Int J Radiat Oncol Biol Phys
2016
;
96
:
E465
.
28.
Onishi
H
,
Shirato
H
,
Nagata
Y
,
Hiraoka
M
,
Fujino
M
,
Gomi
K
, et al
Hypofractionated stereotactic radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study
.
J Thorac Oncol
2007
;
2
:
S94
100
.
29.
Ball
D
,
Mai
GT
,
Vinod
S
,
Babington
S
,
Ruben
J
,
Kron
T
, et al
Stereotactic ablative radiotherapy versus standard radiotherapy in stage 1 non-small-cell lung cancer (TROG 09.02 CHISEL): a phase 3, open-label, randomised controlled trial
.
Lancet Oncol
2019
;
20
:
494
503
.
30.
Rusthoven
KE
,
Kavanagh
BD
,
Burri
SH
,
Chen
C
,
Cardenes
H
,
Chidel
MA
, et al
Multi-institutional phase I/II trial of stereotactic body radiation therapy for lung metastases
.
J Clin Oncol
2009
;
27
:
1579
84
.
31.
Liu
F
,
Tai
An
,
Lee
P
,
Biswas
T
,
Ding
GX
,
El Naqa
I
, et al
Tumor control probability modeling for stereotactic body radiation therapy of early-stage lung cancer using multiple bio-physical models
.
Radiother Oncol
2017
;
122
:
286
94
.
32.
Laufer
I
,
Iorgulescu
JB
,
Chapman
T
,
Lis
E
,
Shi
W
,
Zhang
Z
, et al
Local disease control for spinal metastases following “separation surgery” and adjuvant hypofractionated or high-dose single-fraction stereotactic radiosurgery: outcome analysis in 186 patients
.
J Neurosurg Spine
2013
;
18
:
207
14
.
33.
Alghamdi
M
,
Tseng
C-L
,
Myrehaug
S
,
Maralani
P
,
Heyn
C
,
Soliman
H
, et al
Postoperative stereotactic body radiotherapy for spinal metastases
.
Chin Clin Oncol
2017
;
6
:
S18
.
34.
Huang
AC
,
Postow
MA
,
Orlowski
RJ
,
Mick
R
,
Bengsch
B
,
Manne
S
, et al
T-cell invigoration to tumour burden ratio associated with anti-PD-1 response
.
Nature
2017
;
545
:
60
5
.
35.
Bondiau
P-Y
,
Courdi
A
,
Bahadoran
P
,
Chamorey
E
,
Queille-Roussel
C
,
Lallement
M
, et al
Phase 1 clinical trial of stereotactic body radiation therapy concomitant with neoadjuvant chemotherapy for breast cancer
.
Int J Radiat Oncol Biol Phys
2013
;
85
:
1193
9
.
36.
Zeng
J
,
See
AP
,
Phallen
J
,
Jackson
CM
,
Belcaid
Z
,
Ruzevick
J
, et al
Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas
.
Int J Radiat Oncol Biol Phys
2013
;
86
:
343
9
.
37.
Phillips
R
,
Shi
WY
,
Deek
M
,
Radwan
N
,
Lim
SuJ
,
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
.
38.
Wang
J
,
Sun
J
,
Liu
LN
,
Flies
DB
,
Nie
X
,
Toki
M
, et al
Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy
.
Nat Med
2019
;
25
:
656
66
.
39.
Markovsky
E
,
Budhu
S
,
Samstein
RM
,
Li
H
,
Russell
J
,
Zhang
Z
, et al
An antitumor immune response is evoked by partial-volume single-dose radiation in 2 murine models
.
Int J Radiat Oncol Biol Phys
2019
;
103
:
697
708
.