Main

Radiation therapy has the potential to enhance systemic immune responses in the context of immune checkpoint inhibition (ICI) through a variety of mechanisms1,2,3,4. Radiotherapy elicits immunogenic cell death that is an immunostimulatory program encompassing tumor-derived antigen processing and presentation, T cell priming, trafficking and migration and the induction of a permissive tumor microenvironment (TME)1,2,3,4. As such, the immunostimulatory effects of radiotherapy on priming and effector phases of antitumor immunity have been shown to mediate rejection of the irradiated tumor and nonirradiated metastatic sites, with the latter known as the abscopal effect1. The potential for therapeutic synergy between radiotherapy and ICI has been evidenced by numerous preclinical studies1,5,6,7 and ICI after local tumor irradiation has been shown to induce the expansion of both intratumoral CD8+ T cells and potent abscopal responses8. Combined radioimmunotherapy can decrease regulatory CD4+ T cells while increasing effector memory, early activation and precursor-exhausted CD8+ T cells9. While the immunomodulatory effects of radiotherapy have been described in the TME of irradiated tumors, much less is known about the biology of the abscopal effect that may be mediated by cytokine release and migration of activated effector T cells10.

In tandem, despite the compelling rationale for combination radioimmunotherapy11,12, the clinical efficacy of such approaches has not been consistently demonstrated, highlighting the unmet need to better understand the immunomodulatory effects of radiotherapy in the context of clinical trials and ultimately identify the subset of persons that may most benefit from these approaches. This is particularly timely for individuals with non-small cell lung cancer (NSCLC), where a sizable fraction develops primary or acquired resistance to ICI and where radioimmunotherapy has shown clinical efficacy in the metastatic13,14, locally advanced15 and resectable16 settings. To address these questions, capture systemic antitumor immune responses and pinpoint the landscape of response to sequential radioimmunotherapy for individuals with metastatic NSCLC, especially in the context of immunologically cold tumors, we performed serial comprehensive multiomic analyses of nonirradiated tumors and their TME together with dynamic temporal investigation of the intratumoral and peripheral T cell repertoire, leveraging the randomized, phase 2 PEMBRO-RT trial of pembrolizumab after stereotactic body radiation therapy (SBRT) compared with pembrolizumab monotherapy (NCT02492568)13. We focused on putatively immunologically cold tumors (hereafter referred to as immunologically cold tumors), defined by a low tumor mutation burden (TMB < 300 mutations per exome), null programmed death ligand 1 (PDL1) expression or presence of mutations in the Wnt pathway, and linked our findings with mutation-associated neoantigen (MANA)-reactive T cell responses, ultimately suggesting that radioimmunotherapy may circumvent immunotherapy primary resistance of these tumors.

Results

Cohort and analysis overview

We examined serial nonirradiated tumor and peripheral blood samples collected from participants treated with pembrolizumab (200 mg kg−1 every 3 weeks) either alone (control arm) or in combination with SBRT (three doses of 8 Gy to a single tumor site before pembrolizumab initiation; SBRT arm) in the multicenter randomized phase 2 PEMBRO-RT clinical trial13 (NCT02492568). We leveraged 293 serial peripheral blood and nonirradiated tumor samples collected at baseline and after 3–6 weeks of treatment (2 cycles of pembrolizumab) from 72 participants in the control (n = 37) and SBRT (n = 35) arms. Matched baseline and on-therapy nonirradiated tumor and blood samples were used for genomic, transcriptomic and T cell repertoire analyses (Methods and Supplementary Tables 14). In tandem, ex vivo autologous T cell cultures were pulsed with MANA-derived peptides to identify MANA-reactive T cell clones (Methods). Immunologically cold tumors defined as TMB-low (n = 43), PDL1-null (n = 41) or Wnt-mutated (n = 10) were separately studied (Methods).

Genomic features and PDL1 expression are differentially associated with response to radioimmunotherapy

We hypothesized that clinical responses with radioimmunotherapy would be encountered across the spectrum of TMB and PDL1 expression; to this end, we first assessed differential correlations between these biomarkers (indicative of immunologically hot tumors) and clinical outcomes in the SBRT and control groups. We computed TMB estimates from whole-exome sequencing (WES) (Methods and Supplementary Table 2) and, in line with previous studies17, we found a correlation between TMB and radiographic response in participants in the control arm (Mann–Whitney U-test, P = 0.023). Consistent with our hypothesis, high TMB was not associated with therapeutic response in the SBRT arm (Mann–Whitney U-test, P = 0.53) (Fig. 1 and Supplementary Table 5). As reported in the PEMBRO-RT study13, high PDL1 expression was associated with radiographic response in the control arm (Mann–Whitney U-test, P = 0.00041), with a trend noted in the SBRT arm (Mann–Whitney U-test, P = 0.07) (Fig. 1 and Supplementary Table 5).

Fig. 1: Genomic and molecular features of differential responses to immunotherapy and radioimmunotherapy.
figure 1

Participants are stratified by control versus SBRT arm and therapy response within each arm (CR + PR versus SD + PD); rows represent distinct features and columns represent individual participants. TMB correlated with radiographic response in the control arm (Mann–Whitney U-test, P = 0.023) but not the SBRT arm (Mann–Whitney U-test, P = 0.53). Similarly, PDL1 expression was associated with therapy response in the control arm (Mann–Whitney U-test, P = 0.00041), with a trend noted in the SBRT arm (Mann–Whitney U-test, P = 0.07). In line with the TMB findings, a mutational smoking signature was enriched in responding tumors in the control arm (Mann–Whitney U-test, P = 0.019) but not the SBRT arm (Mann–Whitney U-test, P = 0.12). Tumor aneuploidy (represented as the fraction of genome with allelic imbalance) was not correlated with response in the control or SBRT arms (Mann–Whitney U-test, P = 0.43 and P = 0.87, respectively). Key NSCLC driver genes are shown together with annotations for hotspot mutations. We did not identify a differential enrichment in the overall number or in oncogenic mutations in STK11, KRAS or TP53 by treatment arm; however, KRAS;TP53 comutations were enriched in responding tumors in the control arm. A total of 16 tumors harbored STK11 mutations, 13 of which are characterized as oncogenic in the literature (10 in the control arm and 3 in the SBRT arm). Of these 13 participants, there was 1 responding participant with an STK11-mutant tumor in the control arm and 1 responding participant with an STK11-mutant tumor in the SBRT arm (1/10, 10% versus 1/3, 33%; Fisher’s exact test, P = 0.42). Notably, we observed an enrichment of Wnt pathway mutations in participants with tumors responding to SBRT (OS > 12 months; Fisher’s exact test, P = 0.047).

Source data

In evaluating differences in the mutational spectra and consistent with the TMB findings, we found an enrichment of the mutational smoking signature in responding tumors in the control arm (Mann–Whitney U-test, P = 0.019) but not the SBRT arm (Mann–Whitney U-test, P = 0.12) (Fig. 1 and Supplementary Table 5). Given the potential association between aneuploidy and response to combined radioimmunotherapy18, we evaluated differences in aneuploidy among tumors with differential therapy responses (Methods, Fig. 1, Supplementary Tables 5 and 6, and Extended Data Fig. 1a,b). We did not detect a correlation between aneuploidy and clinical response in the SBRT arm (Supplementary Tables 5 and 6 and Extended Data Fig. 1b–d). In stratifying tumors in the SBRT arm by degree of aneuploidy, we noted that participants with highly aneuploid tumors had a numerically shorter progression-free survival (PFS) and overall survival (OS) (median PFS: 4.39 versus 15.56 months; log-rank test, P = 0.29; median OS: 9.89 versus 40.50 months; log-rank test, P = 0.13) (Extended Data Fig. 1c,d).

Next, we evaluated differential single-gene, pathway and comutation patterns, particularly focusing on capturing clinical responses with radioimmunotherapy in tumors harboring genomic features of resistance to ICI. We did not identify an enrichment in driver alterations by therapeutic response in either arm (Supplementary Table 7). Focusing on KRAS comutations, we did not identify an enrichment in KRAS;STK11, KRAS;KEAP1, KRAS;KEAP1;STK11 or KRAS;TP53 comutations in the SBRT arm. Similarly, in evaluating the differential enrichment of STK11 mutations by treatment arm, we did not detect an association between STK11 oncogenic mutations and therapy response (Fisher’s exact test, P = 0.42) (Fig. 1) nor did we observe transcriptomic or T cell receptor (TCR) reshaping differences in these tumors (Extended Data Fig. 2a–d). Consistent with our previous studies19, KRAS;TP53 comutations were enriched in responding tumors in the control arm (Fisher’s exact test, P = 0.05) (Fig. 1 and Supplementary Table 7). Notably, we found a numerically higher frequency of mutations in genes in the canonical Wnt–β-catenin pathway in responding tumors in the SBRT arm, which was particularly interesting given the expected T cell exclusion of such tumors20 (Fig. 1 and Supplementary Table 8). To orthogonally assess Wnt pathway status in tumors harboring Wnt mutations, we evaluated Wnt pathway signaling leveraging transcriptomic data and indeed found an upregulation in expression of Wnt signaling-associated gene sets in Wnt-mutated tumors (false discovery rate (FDR)-adjusted P = 0.046) (Supplementary Table 8). These findings indicated that clinical responses with radioimmunotherapy are encountered across the spectrum of immunologically cold and hot tumors, including tumors harboring features of immunotherapy resistance.

Serial transcriptomic analyses point to upregulation of adaptive immunity programs at nonirradiated metastatic sites after radioimmunotherapy

We next asked the question whether the synergistic therapeutic effect of ICI and SBRT, supported by the clinical outcomes in the PEMBRO-RT trial13, was reflected in the induction of systemic inflammatory responses and remodeling of the TME of nonirradiated tumor sites. To this end, we assessed changes in the expression of immune-related pathways by RNA sequencing (RNA-seq) of serial nonirradiated tumor samples, collected at baseline and after two cycles of pembrolizumab ± SBRT (Methods and Supplementary Table 3). Gene set enrichment analysis (GSEA) was performed to evaluate differential expression of inflammatory and adaptive immunity programs (Methods). These analyses revealed significant post-SBRT and on-therapy upregulation of interferon-γ (IFNγ; normalized effect size (NES) = 2.60, FDR-adjusted P = 1.03 × 10−25), IFNα (NES = 2.43, FDR-adjusted P = 1.64 × 10−12), chemokine signaling (NES = 2.32, FDR-adjusted P = 2.78 × 10−14), antigen processing and presentation (NES = 2.38, FDR-adjusted P = 2.28 × 10−10), natural killer (NK) cell cytotoxicity (NES = 2.50, FDR-adjusted P = 9.13 × 10−16), B cell receptor (BCR) signaling (NES = 2.48, FDR-adjusted P = 5.66 × 10−18) and other conserved inflammatory response gene sets in tumors in the SBRT arm (Fig. 2a and Supplementary Table 9). Each of these gene sets was more upregulated from baseline to on therapy in the SBRT arm than in the control arm (Fig. 2a and Supplementary Tables 9 and 10). Notable on-therapy expression differences between the SBRT and control arms were also observed in the downregulation of gene sets related to cell-cycle progression (NES = −1.50, FDR-adjusted P = 0.0188), double-stranded DNA break repair (NES = −1.52, FDR-adjusted P = 0.011), Myc targets (NES = −2.16, FDR-adjusted P = 6.63 × 10−7), glycolysis (NES = −1.82, FDR-adjusted P = 1.19 × 10−5), ribosome biogenesis (NES = −1.54, FDR-adjusted P = 0.021) and Sonic hedgehog signaling (NES = −1.68, FDR-adjusted P = 0.0072), all of which were more downregulated in the SBRT arm compared with the control arm (Fig. 2b and Supplementary Tables 9 and 10). Overall, systemic radiation-induced effects spanned a number of immune gene expression programs (Methods, Fig. 2c–e and Supplementary Table 11) that were found to be upregulated with radioimmunotherapy in abscopal, nonirradiated tumor sites.

Fig. 2: TME reshaping with radioimmunotherapy.
figure 2

a, Bar plot of the most upregulated gene sets in on-therapy tumors by treatment arm as ranked by adjusted P values from GSEA. A number of inflammatory gene sets were differentially upregulated in on-therapy tumors in the SBRT arm (n = 14 samples) compared with the control arm (n = 12 samples), including interactions between lymphoid and nonlymphoid cells (FDR-adjusted P = 7.35 × 1035 in the SBRT arm and P = 3.16 × 1015 in the control arm), neutrophil degranulation (FDR-adjusted P = 1.46 × 1034 in the SBRT and P = 2.67 × 1011 in the control arm), IFNγ response (FDR-adjusted P = 1.03 × 1025 in the SBRT and P = 8.72 × 1017 in the control arm) and overall inflammatory response (FDR-adjusted P = 5.31 × 1017 in the SBRT arm and P = 4.58 × 109 in the control arm). A detailed description of all upregulated gene sets can be found in Supplementary Tables 9 and 10. b, Differentially downregulated gene sets in on-therapy tumors by treatment arm included cell-cycle targets of E2f transcription factors (FDR-adjusted P = 9.13 × 109 in the SBRT arm and P = 5.65 × 10−1 in the control arm) and genes regulated by Myc (FDR-adjusted P = 6.63 × 107 in the SBRT arm and P = 9.78 × 101 in the control arm). A detailed description of all downregulated gene sets can be found in Supplementary Tables 9 and 10. c, Heat map of GSEA results showing greater enrichment of immune programs from baseline to on therapy in the SBRT arm (n = 14 samples) compared with the control arm (n = 12 samples) across a broad range of immune gene sets. Each row represents a gene set; gene sets are grouped into ten categories shown in the legend. Enrichment scores were normalized by row to a maximum value of 1. d, Enrichment plot showing the leading edge of the IFNγ gene set, clearly upregulated on therapy in the SBRT arm (FDR-adjusted P = 1.03 × 1025). e, Enrichment plot showing the leading edge of the inflammatory response gene set, clearly upregulated on therapy in the SBRT arm (FDR-adjusted P = 5.31 × 1017). f, Investigation of differences in B cell density in SBRT responders, who showed a numerically greater BCR CDR3 count on therapy than at baseline (mean: 3.24 × 106 versus 2.45 × 105; Mann–Whitney U-test, P = 0.19). g, Bar plot of the gene sets most enriched on therapy in SBRT long-term survivors (OS ≥ 3 years) versus SBRT short-term survivors (OS < 3 years), including interactions between lymphoid and nonlymphoid cells (FDR-adjusted P = 1.09 × 1029), BCR signaling (FDR-adjusted P = 9.42 × 1016) and IFNγ response (FDR-adjusted P = 5.28 × 106). Extensive results can be found in Supplementary Table 15. h, Bar plot of the gene sets most downregulated on therapy in SBRT long-term survivors (OS ≥ 3 years) versus SBRT short-term survivors (OS < 3 years), including cell-cycle targets of E2f transcription factors (FDR-adjusted P = 3.64 × 1014), G2/M checkpoint progression (FDR-adjusted P = 1.61 × 1010), glycolysis (FDR-adjusted P = 1.20 × 108) and double-stranded DNA break repair (FDR-adjusted P = 1.95 × 105). Extensive results can be found in Supplementary Table 15. All statistical results are FDR-adjusted and two-sided P values. Box plots depict the median value and hinges correspond to the first and third quartiles. The whiskers extend from the corresponding hinge to the furthest value within 1.5× the interquartile range from the hinge. Dotted black horizontal lines indicate the FDR-adjusted P = 0.05. EMT, epithelial–mesenchymal transition; ID, immunodeficiency; Ag, antigen; iC, intracellular; RT, Reactome; HM, Hallmark; KG, Kegg.

Source data

We next assessed dynamic changes in the abundance of immune cell subsets by deconvolution of serial transcriptomic data in the SBRT and control arms (Methods). In line with the GSEAs, several immune cell populations increased in absolute abundance on therapy within the SBRT cohort. We observed significant upregulation of CD8 T cells (Mann–Whitney U-test, P = 0.027) and M1 macrophages (Mann–Whitney U-test, P = 0.013) and a trend toward increased density of activated CD4 memory T cells (Mann–Whitney U-test, P = 0.074) and activated NK cells (Mann–Whitney U-test, P = 0.09) (Extended Data Fig. 3a–d and Supplementary Table 12). All deconvolution results are shown in Extended Data Fig. 3a–v. In contrast, these dynamic shifts were not apparent in on-therapy tumors in the control arm (Extended Data Fig. 3 and Supplementary Table 12). Taken together, these findings support that radioimmunotherapy may induce reshaping of the TME of abscopal tumor sites toward a more inflamed phenotype. Given the increased abundance in CD8 T cell population in on-therapy tumors with radioimmunotherapy, we next examined whether this coincided with an upregulation of coinhibitory receptors. We indeed found a significant upregulation of the inhibitory receptors PD1, LAG3 and TIM3 (FDR-adjusted P = 0.017, P = 0.026 and P = 0.009, respectively) with a similar trend observed for cytotoxic T lymphocyte antigen 4 (CTLA4) and TIGIT (FDR-adjusted P = 0.15 and P = 0.11, respectively) in on-therapy tumors in the SBRT arm (Supplementary Table 13). These findings are suggestive of the upregulation of coinhibitory receptors by tumor antigen stimulation in the context of radiation-induced immunogenic cell death and potentially open a therapeutic window of opportunity for further modulation of inhibitory pathways that can reverse T cell exhaustion and reinvigorate immune responses.

Interestingly, induction of BCR signaling was unique to the SBRT arm (Fig. 2a,c) (NES = 2.48, FDR-adjusted P = 5.66 × 1018 in the SBRT arm and NES = 1.28, FDR-adjusted P = 0.15 in the control arm). To further investigate the effect of radioimmunotherapy on B cell responses, we performed bulk RNA-seq deconvolution and parsed BCR complementary-determining region 3 (CDR3) assemblies from baseline and on-therapy samples (Methods). BCR CDR3 count (reflecting BCR clonotypic density) was similar between responding and nonresponding tumors at baseline (Mann–Whitney U-test, P = 0.395) (Extended Data Fig. 4a and Supplementary Table 14) but significantly greater in responding tumors on therapy (Mann–Whitney U-test, P = 0.015) (Extended Data Fig. 4a and Supplementary Table 14). We next compared B cell density between on-therapy and baseline samples within each therapy arm, stratified by treatment response. Nonresponding tumors showed no difference in BCR CDR3 count between baseline and on-therapy samples in the control (Mann–Whitney U-test, P = 0.97) or SBRT arms (Mann–Whitney U-test, P = 0.74) (Supplementary Table 14). While our statistical power was limited by sample size, responding tumors in the SBRT arm showed a numerically greater BCR CDR3 count on therapy (Mann–Whitney U-test, P = 0.19) (Fig. 2f and Supplementary Table 14). As an example, we observed that three participants in the SBRT arm showed markedly greater BCR CDR3 counts on therapy than any participant in the control arm, which was reflective of durable clinical benefit on radioimmunotherapy for all three participants (Extended Data Fig. 4b and Supplementary Table 14). These findings provide anecdotal evidence for the potential of radioimmunotherapy to induce B cell expansions in the context of durable clinical benefit.

Transcriptomic analyses in long-term survivors undergoing radioimmunotherapy

Next, we sought to interpret the systemic effects of radioimmunotherapy on the basis of long-term outcomes, which would indirectly provide insights into the durability of the radiotherapy-induced systemic immune response. To this end, we performed differential expression gene set analyses of baseline and on-therapy tumors for participants with short-term versus long-term survival in the SBRT arm (Methods). At baseline, long-term survivors in the SBRT arm showed heightened expression of IFNγ (NES = 2.41, FDR-adjusted P = 6.69 × 10−16) and IFNα (NES = 2.32, FDR-adjusted P = 3.47 × 108) gene sets compared with SBRT short-term survivors (Supplementary Table 8). Notably, in the on-therapy samples from long-term survivors in the SBRT arm, we found an upregulation of a wider variety of immune-related pathways, including BCR signaling (NES = 2.54, FDR-adjusted P = 9.42 × 1016), neutrophil degranulation (NES = 1.69, FDR-adjusted P = 5.18 × 106) and IFNγ (NES = 1.94, FDR-adjusted P = 5.28 × 106) gene sets (Fig. 2g and Supplementary Table 15). In tandem, baseline tumors of long-term survivors in the SBRT arm showed significantly greater expression of cellular proliferation and cell-cycle progression gene sets (E2f targets: FDR-adjusted P = 3.42 × 1021, NES = 2.59; G2/M checkpoint: FDR-adjusted P = 1.68 × 1013, NES = 2.35) (Supplementary Table 8), which was reversed on therapy after radioimmunotherapy (E2f targets: FDR-adjusted P = 3.64 × 1014, NES = −2.48; G2/M checkpoint: FDR-adjusted P = 1.61 × 1010, NES = −2.31) (Fig. 2h and Supplementary Table 15). Furthermore, we found a downregulation in double-stranded DNA repair gene sets in on-therapy tumors of long-term responders (NES = −2.05, FDR-adjusted P = 1.95 × 105), which may reflect synergies between genomic instability and radiation-induced tumor immunogenicity. Taken together, these findings suggest that tumor immune infiltration and an inflamed TME at baseline is a favorable prognostic feature for long-term survival, which is further enhanced on therapy after radioimmunotherapy. Interestingly, while tumors of long-term survivors harbor transcriptional programs that point toward a high proliferation and DNA replication phenotype before therapy, these seem to be reversed after radioimmunotherapy; collectively, these findings suggest rewiring of cancer cells and their TME with radiotherapy.

Synergistic upregulation of adaptive immunity programs in TMB-low, PDL1-null or Wnt-mutated tumors with radioimmunotherapy

We subsequently evaluated whether the upregulation of inflammatory responses observed in the SBRT arm was also evident in immunologically cold tumors. Consistent with our findings in the whole cohort, in the TMB-low group, we observed significant post-SBRT and on-therapy upregulation of IFNγ (NES = 2.34, FDR-adjusted P = 1.69 × 1012), IFNα (NES = 2.37, FDR-adjusted P = 8.51 × 109), chemokine signaling (NES = 2.24, FDR-adjusted P = 7.94 × 1011), antigen processing and presentation (NES = 2.35, FDR-adjusted P = 5.40 × 108), NK cell cytotoxicity (NES = 2.16, FDR-adjusted P = 1.43 × 106) and conserved inflammatory response gene sets in tumors in the SBRT arm (Fig. 3a and Supplementary Table 16). The magnitude of on-therapy upregulation of each gene set was greater in the SBRT arm compared with the control arm (Supplementary Table 17). Similarly, in the PDL1-null group of the SBRT arm, we observed significant post-SBRT and on-therapy upregulation of IFNγ (NES = 2.89, FDR-adjusted P = 3.94 × 1034), IFNα (NES = 2.71, FDR-adjusted P = 1.29 × 1017), chemokine signaling (NES = 2.47, FDR-adjusted P = 5.14 × 1015), antigen processing and presentation (NES = 2.60, FDR-adjusted P = 1.15 × 1014), NK cell cytotoxicity (NES = 2.60, FDR-adjusted P = 3.28 × 1016), BCR signaling (NES = 2.49, FDR-adjusted P = 6.47 × 1015) and conserved inflammatory response gene sets (Fig. 3b and Supplementary Table 18). Again here, the magnitude of on-therapy upregulation of each gene set was greater in the SBRT arm than the control arm (Supplementary Table 19). In the Wnt-mutated group, we found significant post-SBRT and on-therapy upregulation of similar inflammatory gene sets as noted in the PDL1-null and TMB-low groups. These included IFNγ (NES = 2.32, FDR-adjusted P = 4.78 × 1013), IFNα (NES = 2.26, FDR-adjusted P = 7.29 × 109), antigen processing and presentation (NES = 2.35, FDR-adjusted P = 6.49 × 109) and BCR signaling (NES = 2.72, FDR-adjusted P = 5.74 × 1026) (Supplementary Table 20). The greater enrichment of immune programs induced by SBRT extended to a broad range of inflammatory gene sets (Supplementary Table 11) in the TMB-low (Fig. 3c and Supplementary Table 16), PDL1-null (Fig. 3d and Supplementary Table 18) and Wnt-mutated (Fig. 3e and Supplementary Table 20) groups. As a representative example, IFNγ signaling was significantly upregulated on therapy in abscopal nonirradiated sites in each of the immunologically cold tumor subsets, indicating the mounting of systemic antitumor immune responses (Fig. 3f–h). Given the induction of tertiary lymphoid structures (TLSs) by immunogenic cell death, we postulated that TLSs may be upregulated at the nonirradiated tumor sites in the SBRT arm compared with the control arm. A TLS gene expression signature was indeed significantly upregulated on therapy in PDL1-null tumors in the SBRT arm (NES = 1.71, FDR-adjusted P = 0.02) (Extended Data Fig. 5a and Supplementary Table 8) but not in the control arm (NES = 1.22, FDR-adjusted P = 0.45) (Extended Data Fig. 5b and Supplementary Table 8). Furthermore, the TLS signature was significantly upregulated in Wnt-mutated tumors in the SBRT arm after SBRT and on therapy (NES = 1.90, FDR-adjusted P = 0.002) (Extended Data Fig. 5c and Supplementary Table 8). Collectively, these findings support the induction of systemic immune responses involving both T and B cell immunity and are suggestive of a potential ‘cold-to-hot’ conversion of the TME with radioimmunotherapy.

Fig. 3: TME reshaping with radioimmunotherapy in immune cold tumors.
figure 3

a, Bar plot of the 10 most upregulated gene sets from baseline to 3–6 weeks on therapy in TMB-low tumors in the SBRT (n = 12 samples) and control (n = 11 samples) arms, including neutrophil degranulation (FDR-adjusted P = 4.93 × 1030 in the SBRT arm and P = 1.58 × 109 in the control arm), IFNγ response (FDR-adjusted P = 1.69 × 1012 in the SBRT arm and P = 5.97 × 1012 in the control arm), chemokine signaling (FDR-adjusted P = 7.94 × 1011 in the SBRT arm and P = 9.79 × 106 in the control arm), overall inflammatory response (FDR-adjusted P = 1.12 × 109 in the SBRT arm and P = 2.68 × 107 in the control arm), and antigen processing and presentation (FDR-adjusted P = 5.40 × 108 in the SBRT arm and P = 1.92 × 105 in the control arm). Extensive findings can be found in Supplementary Table 16. b, Bar plot of the 10 most upregulated gene sets from baseline to 3–6 weeks on therapy in PDL1-null tumors in the SBRT (n = 9 samples) and control (n = 9 samples) arms, including IFNγ response (FDR-adjusted P = 3.94 × 1034 in the SBRT arm and P = 5.52 × 106 in the control arm), IFNα response (FDR-adjusted P = 1.29 × 1017 in the SBRT arm and P = 1.08 × 102 in the control arm), NK cell cytotoxicity (FDR-adjusted P = 3.28 × 1016 in the SBRT arm and P = 6.43 × 106 in the control arm), BCR signaling (FDR-adjusted P = 6.47 × 1015 in the SBRT arm and P = 0.84 in the control arm), and antigen processing and presentation (FDR-adjusted P = 1.15 × 1014 in the SBRT arm and P = 3.73 × 105 in the control arm). Extensive findings can be found in Supplementary Table 16. ce, Heat map of GSEAs showing the most upregulated immune gene sets from baseline to 3–6 weeks on therapy in TMB-high tumors in the SBRT arm (n = 4 samples), TMB-low tumors in the SBRT arm (n = 11 samples) and TMB-low tumors in the control arm (n = 10 samples) (c), in PDL1-positive tumors in the SBRT arm (n = 6 samples), PDL1-null tumors in the SBRT arm (n = 8 samples), PDL1-positive tumors in the control arm (n = 4 samples) and PDL1-null tumors in the control arm (n = 8 samples) (d), and in Wnt wild-type tumors in the SBRT arm (n = 12 samples), Wnt-mutated tumors in the SBRT arm (n = 2 samples) and Wnt wild-type tumors in the control arm (n = 10 samples) (e). Each row represents a gene set; gene sets are grouped into nine categories shown in the legend. Enrichment scores were normalized by row to a maximum value of 1. For every cohort stratification (TMB-high versus TMB-low, PDL1-positive versus PDL1-null and Wnt-mutated versus Wnt wild-type tumors), we observed greater enrichment of immune gene sets in the SBRT arm than the control arm across a broad range of immune gene sets. f, Enrichment plot of the IFNγ gene set in on-therapy tumors with low TMB in the SBRT arm (n = 12 samples; FDR-adjusted P = 1.69 × 1012). g, Enrichment plot of the IFNγ gene set in on-therapy PDL1-null tumors in the SBRT arm (n = 9 samples; FDR-adjusted P = 3.94 × 1034). h, Enrichment plot of the IFNγ gene set in on-therapy Wnt-mutated tumors in the SBRT arm (n = 5 samples) from baseline to 3–6 weeks on therapy (FDR-adjusted P = 4.78 × 1013). All statistical results are two-sided P values. Dotted black horizontal lines indicate FDR-adjusted P = 0.05.

Source data

Radioimmunotherapy confers longer clinical outcomes in participants with immunologically cold tumors

Building on the differential association of TMB and PDL1 with therapy response in the SBRT arm compared with pembrolizumab monotherapy and prompted by the transcriptomic analyses supporting the abscopal effect of radioimmunotherapy, we sought to determine whether tumors that are less likely to respond to ICI monotherapy demonstrated radiographic responses with radioimmunotherapy (Fig. 4a). We first evaluated RECIST (response evaluation criteria in solid tumors) measurements of the biopsied nonirradiated lesions at baseline and at 12 weeks for each participant (Methods). Baseline tumor measurements were similar between participants in the SBRT and control groups and across all subsets analyzed (TMB-low, TMB-high, PDL1-null, PDL1-positive, Wnt-mutated and Wnt wild-type tumors). Overall, the abscopal (biopsied) tumor sites in the SBRT arm had a notable decrease in RECIST measurements between 0 and 12 weeks (mean: 45.52 mm to 30.44 mm; Mann–Whitney U-test, P = 0.05) (Fig. 4b and Supplementary Table 21), whereas rebiopsied tumor sites in the control arm did not show a significant change in RECIST measurements (mean: 45.48 mm to 44.85 mm; Mann–Whitney U-test, P = 0.64) (Supplementary Table 21). Similarly, in TMB-low and PDL1-null participants, we observed a notable regression in RECIST measurements of the biopsied abscopal site at 12 weeks in the SBRT arm (TMB-low mean: 47.93 mm to 30.44 mm; PDL1-null mean: 50.78 mm to 32 mm) (Fig. 4c, Supplementary Table 21 and Extended Data Fig. 6a), with similar trends in the Wnt-mutant subgroup (Extended Data Fig. 6b). Interestingly, the SBRT-associated abscopal site tumor regression was not as pronounced in TMB-high and PDL1-positive tumors (Fig. 4c, Supplementary Table 21 and Extended Data Fig. 6a). These findings highlight the factual tumor regression of abscopal sites in the SBRT arm in comparison to the control arm associated with improved response.

Fig. 4: Differential outcomes and abscopal site tumor regression with radioimmunotherapy for participants with TMB-low, PDL1-null and Wnt-mutated tumors.
figure 4

a, Swimmer’s plot showing OS and clinical and pathological features for each participant in the control (left) and SBRT (right) arms. Participants with TMB-low, PDL1-null or Wnt-mutated tumors were observed to attain the longest clinical outcomes in the SBRT arm. NA, not applicable. b, The abscopal (biopsied) tumor sites in the SBRT arm had a notable decrease in RECIST measurements between 0 and 12 weeks (mean: 45.52 mm to 30.44 mm; Mann–Whitney U-test, P = 0.054), whereas rebiopsied tumor sites showed no change in RECIST measurements in the control arm (Mann–Whitney U-test, P = 0.64). Extensive findings can be found in Supplementary Table 21. c, In TMB-low and PDL1-null participants, we observed a notable regression in RECIST measurements of the biopsied abscopal site in the SBRT arm (TMB-low mean: 47.93 mm to 30.44 mm, Mann–Whitney U-test, P = 0.13; PDL1-null mean: 50.78 mm to 32 mm; Mann–Whitney U-test, P = 0.29), which was not evident in the control arm. Interestingly, the SBRT-associated abscopal site tumor regression was not as pronounced in TMB-high and PDL1-positive tumors; these had similar regressions in the SBRT and control arms. Extensive findings can be found in Supplementary Table 21. d, Kaplan–Meier curve of probability of PFS in TMB-low participants (n = 43 participants) treated in the control arm (n = 21 participants) and SBRT arm (n = 22 participants). TMB-low participants had longer PFS in the SBRT arm than in the control arm (median PFS: 5.21 versus 1.81 months; log-rank test, P = 0.029). e, Kaplan–Meier curve of probability of PFS in PDL1-null participants (n = 41 participants) treated in the control arm (n = 23 participants) and SBRT arm (n = 18 participants). PDL1-null participants had longer PFS in the SBRT arm than in the control arm (median PFS: 4.22 versus 1.71 months; log-rank test, P = 0.022). f, Kaplan–Meier curve of probability of PFS in the SBRT arm (n = 28 participants) in the Wnt-mutated (n = 5 participants) and Wnt wild-type (n = 23 participants) groups. In the SBRT arm, participants with Wnt-mutated tumors had longer PFS compared with the wild-type subgroup (median PFS: not reached versus 5.45 months; log-rank test, P = 0.02). Box plots depict the median value and hinges correspond to the first and third quartiles. The whiskers extend from the corresponding hinge to the furthest value within 1.5× the interquartile range from the hinge.

Source data

In assessing survival outcomes, participants with tumors harboring TMB-low or PDL1-null expression attained significantly longer PFS in the SBRT arm (median PFS: 5.21 versus 1.81 months; log-rank test, P = 0.029) (Fig. 4d) compared with the control arm (median PFS: 4.22 versus 1.71 months; log-rank test, P = 0.022) (Fig. 4e). Similarly, participants in the SBRT arm with Wnt-mutated tumors (n = 5) had significantly longer PFS compared with those with wild-type tumors (n = 30) (median PFS: not reached versus 5.95 months; log-rank test, P = 0.037) (Fig. 4f). Notably, participants with TMB-high tumors (n = 15), PDL1-positive expression (≥1% staining on immunohistochemistry (IHC); n = 29), PDL1-high tumors (≥50% staining on IHC; n = 15) and PDL1-low tumors (1–49% staining on IHC; n = 14) did not attain longer PFS in the SBRT arm compared with the control arm (Extended Data Fig. 7a–d). Furthermore, there was no difference in PFS between participants with Wnt-mutated compared with wild-type tumors in the control arm (Extended Data Fig. 7e). Taken together, the survival analyses indicate that radioimmunotherapy may be particularly effective for tumors harboring features of ICI primary resistance. We then examined the difference in OS for these participants between treatment arms. Participants with tumors harboring Wnt mutations had a longer OS in the SBRT arm (median OS: not reached versus 9.92 months; log-rank test, P = 0.013) (Extended Data Fig. 8a). Similarly, participants with PDL1-null tumors showed a trend toward longer OS in the SBRT arm compared with the control arm (median OS: 7.21 versus 6.05 months; log-rank test, P = 0.084) (Extended Data Fig. 8b). Participants with TMB-low tumors attained a numerically longer OS in the SBRT arm compared with the control arm (median OS: 9.89 versus 6.70 months; log-rank test, P = 0.16) (Extended Data Fig. 8c).

To control for potential participant-related and technical factors that may confound the association between immunologically cold tumors and clinical response with radioimmunotherapy, we performed multivariate Cox regression analyses adjusting for age, gender, smoking status, tumor histology and tumor purity (Methods). Participants with low TMB tumors had a favorable hazard ratio (HR) for PFS in the SBRT arm (HR: 0.38, P = 0.01); similarly, participants with PDL1-null tumors had a favorable HR when receiving radioimmunotherapy (HR: 0.43, P = 0.066) (Supplementary Table 22). With respect to Wnt-mutated tumors, Wnt mutations were favorably associated with PFS (HR: 0.23, P = 0.065) and OS (HR: 0.19, P = 0.046) in the SBRT arm (Supplementary Table 22). Overall, these findings support the independent association between radioimmunotherapy and survival for participants with immunologically cold tumors.

T cell repertoire reshaping and expansion of new T cell clones with radioimmunotherapy

Tying in the transcriptomic analyses, pointing toward mounting of systemic antitumor immune responses and a more permissive TME after radioimmunotherapy, with T cell repertoire dynamics and composition, we evaluated new and existing TCR clonotypic expansions in the nonirradiated tumor and peripheral blood compartments by TCR Vb CDR3 next-generation sequencing (Methods). To this end, we hypothesized that the noted upregulation of IFN signaling would result in an influx of T cells; as such, the abscopal effect of radiotherapy may be captured by detection of new and expanded TCR clones in the TME and blood compartments. Consistent with our hypothesis, we found a significantly higher density of new (Fig. 5a and Supplementary Table 23) and pre-existing (Fig. 5b and Supplementary Table 23) intratumoral TCR clones in abscopal tumor sites in the SBRT arm compared with the control arm (mean count of all clones: 34.83 versus 18.69, respectively; generalized linear model (GLM) Wald test, P = 0.025; mean count of new clones: 16.17 versus 7.54, respectively; Wald test, P = 0.022). Similar dynamics were observed in peripheral blood, supporting pre-existing and new clonotypic expansions in the SBRT arm compared with the control arm (mean count of all clones: 21.61 versus 8.15, respectively; Wald test, P = 0.030; mean count of new clones: 4.56 versus 1.00; Wald test, P = 0.039) (Fig. 5a,b and Supplementary Table 23). Focusing on TCR clones expanding in both nonirradiated tumor and blood compartments, we again found a higher density of TCR clonotypic expansions in the SBRT arm (Wald test, P = 0.04 for all clones and P = 0.098 for new clones) (Fig. 5a,b and Supplementary Table 16). TCR repertoire reshaping occurred independently of TMB and PDL1 expression, as both variables were incorporated into the GLM as covariates (Methods and Supplementary Table 23). Within the SBRT arm specifically, we did not observe significant differences in expanded clone counts between TMB-low and TMB-high tumors (intratumoral count of all clones: Mann–Whitney U-test, P = 0.86; intratumoral count of new clones: Mann–Whitney U-test, P = 0.95; blood count of all clones: Mann–Whitney U-test, P = 0.43; blood count of new clones: Mann–Whitney U-test, P = 0.95) (Fig. 5c and Supplementary Table 24) or between PDL1-null and PDL1-positive tumors (intratumoral count of all clones: Mann–Whitney U-test, P = 0.33; intratumoral count of new clones: Mann–Whitney U-test, P = 0.17; blood count of all clones: Mann–Whitney U-test, P = 0.63; blood count of new clones: Mann–Whitney U-test, P = 0.075) (Fig. 5d and Supplementary Table 25). These findings suggest that radioimmunotherapy induces expansion of both pre-existing and new T cell clones, an effect that was independent of TMB status and PDL1 expression. As a representative example of the immunomodulatory effects of radioimmunotherapy, participant CGLU727 who attained a partial response (PR) with radioimmunotherapy despite a TMB of 0.9 mutations per Mb and null PDL1 expression, showed a pattern of TCR clonotypic expansion and reshaping in both TME (Fig. 5e, Supplementary Table 26 and Extended Data Fig. 9) and blood (Fig. 5f, Supplementary Table 26 and Extended Data Fig. 9) compartments. We subsequently evaluated the duration of TCR clonotypic expansions by serial sampling of blood and tumor tissue at the time of acquired resistance for a small number of participants for which biospecimens were collected. While limited by sample size, we noted that only a small fraction of the intratumoral TCR clones that expanded at the time of response showed sustained expansion at the time of acquired resistance in two participants in the SBRT arm (7% sustained TCR expansions) and one participant in the control arm (3% sustained TCR expansions), with higher fractions detected in the blood compartment (14% in the SBRT and 63% in the control arm) (Fig. 5g,h, Supplementary Table 27 and Extended Data Fig. 10). Taken together, our findings suggest that combined radioimmunotherapy may induce reshaping of the peripheral T cell repertoire together with abscopal responses in nonirradiated tumor sites that may drive tumor elimination and clinical responses.

Fig. 5: TCR clonotypic reshaping with radioimmunotherapy and longitudinal monitoring of the peripheral blood and intratumoral repertoire during therapy and at the time of acquired resistance.
figure 5

a, Counts of clonotypes expanded on therapy that were not observed at baseline in the blood and/or tumor compartments. There were significantly more newly expanded clones in blood alone and in tumor alone, with a trend of more newly expanded clones in both compartments (SBRT arm, n = 18 samples versus control arm, n = 13 samples; blood mean: 4.56 versus 1.00; Wald test (two-sided), P = 0.039; tumor mean: 16.17 versus 7.54; Wald test (two-sided), P = 0.022; both compartments mean: 1.00 versus 0; Wald test (two-sided), P = 0.098) in the SBRT cohort than control cohort. b, Counts of all clonotypes expanded on therapy relative to baseline in the blood and/or tumor compartments. There were significantly more expanded clones in all compartments in the SBRT cohort than control cohort (SBRT, n = 18 samples versus control arm, n = 13 samples; blood mean: 21.61 versus 8.15; Wald test (two-sided), P = 0.030; tumor mean: 34.83 versus 18.69; Wald test (two-sided), P = 0.025; both compartments mean: 2.61 versus 0.54; Wald test (two-sided), P = 0.045). c, Counts of new clonotypes expanded on therapy within the SBRT arm, stratified by TMB status. Counts of newly expanded clones were not significantly different between TMB-low (n = 13 samples) and TMB-high (n = 4 samples) tumors in the SBRT arm in either compartment (blood: Mann–Whitney U-test (two-sided), P = 0.95; tumor: Mann–Whitney U-test (two-sided), P = 0.95; both compartments: Mann–Whitney U-test (two-sided), P = 0.44). d, Counts of new clonotypes expanded on therapy within the SBRT arm, stratified by PDL1 status. Counts of newly expanded clones were not significantly different between PDL1-null (n = 9 samples) and PDL1-positive (n = 9 samples) tumors in the SBRT arm in either compartment (blood: Mann–Whitney U-test (two-sided), P = 0.075; tumor: Mann–Whitney U-test (two-sided), P = 0.17; both compartments: Mann–Whitney U-test (two-sided), P = 0.46). e,f, TCR dynamics of newly expanded TCR clones in blood (e) and tumor (f) compartments for a participant with low TMB (0.9 mutations per Mb) and PDL1-null expression (0% on IHC) who exhibited PR in the SBRT cohort. Participant 727 had 14 total and 6 new clones expanded in the tumor and 34 total and 29 new clones expanded in the blood following SBRT and 2 cycles of pembrolizumab (Mann–Whitney U-test, two-sided). g, Clonotype dynamics at the time of resistance for clones that expanded on therapy in three PR tumors: two in the SBRT (participants 680 and 690) and one in the control arm (participant 743). Approximately 39% (12/31) of the clonotypes that expanded in blood and 4% (2/51) that expanded in the tumor on treatment across the 3 participants showed sustained increase at the time of resistance. At the participant level, most clones that expanded on treatment showed no sustained increase at the time of resistance (93% (13/14), 86% (12/14) and 77% (43/56) for the 3 participants). h, Expansion dynamics of all T cell clones that expanded in the tumor on treatment for participant 680 (PR in SBRT cohort; Mann–Whitney U-test, two-sided). Most clones did not show sustained increase at the resistance time point. Box plots depict the median value and hinges correspond to the first and third quartiles. The whiskers extend from the corresponding hinge to the furthest value within 1.5× the interquartile range from the hinge.

Source data

Radioimmunotherapy induces systemic neoantigen-reactive T cell responses

We next hypothesized that the systemic effect of radiotherapy would be reflected in the detection of neoantigen-reactive T cell responses, targeting clonal expressed mutations that would be shared across the primary, irradiated and abscopal (biopsied) tumor sites. To test this hypothesis, we used the MANA functional expansion of specific T cells (MANAFEST) assay21 to identify MANA-specific T cell clones (Methods) in 3 long-term survivors who attained PR in the SBRT arm despite their tumors harboring features of immunotherapy resistance (participant 680 with an OS of 104 months, participant 690 with an OS of 102 months and participant 727 with an OS of 46 months). We synthesized neopeptides using each tumor’s WES data, followed by peptide stimulation of autologous T cells from each participant and identification of neoantigen-specific clonotypic amplifications by TCR sequencing (Methods and Supplementary Table 28). For participant CGLU680, we identified ten neoantigen-reactive TCR clones against clonal expressed mutations in DAXX, FN3K, GCN1L1, ZBTB44, EYA2 and KIAA1211 and a frameshift mutation in BAI2. Of note, the DAXX neopeptide-reactive clone was detected expanding in T cell cultures from baseline and cycle 3 on radioimmunotherapy, whereas the remainder of neoantigen-reactive clonotypic expansions were detected upon testing autologous T cell cultures procured after initiation of radioimmunotherapy (at cycles 2, 3 and 4 of pembrolizumab) (Fig. 6a and Supplementary Table 29). Furthermore, a BDH1 mutation-associated neopeptide stimulated expansion of TCR clone CASSLWAGGGSREQFF in participant CGLU690 and, similarly, a RHNO1 mutation-associated neopeptide stimulated expansion of TCR clone CASSIPGEGYTF in participant GGLU727, both after sequential radioimmunotherapy (Fig. 6b,c and Supplementary Tables 30 and 31). These findings support MANA-reactive T cell responses in long-term survivors receiving sequential radiotherapy and anti-PD1 therapy and provide further evidence to support the induction of systemic antitumor immune responses with radioimmunotherapy.

Fig. 6: Neoantigen-reactive T cell responses in participants with long-term survival after radioimmunotherapy.
figure 6

We assessed neoantigen-specific T cell responses in three participants who attained radiographic response and long-term OS benefit in the SBRT arm. a, For participant CGLU680 who attained PR and an OS of 104 months, 10 neoantigen-reactive TCRs were detected specific to 8 MANAs across 4 time points (colored bars). Neopeptide sequences are listed on the horizontal axis with mutated genes in parentheses. TCR CDR3 amino acid sequences are listed along the depth axis with the time point of significant expansion prepended to each TCR sequence. Opaque dark-gray columns represent a significant expansion of T cell clones in response to the positive control peptide, while colored columns represent a significant expansion of T cell clones in response to cancer neopeptides. Opaque light-gray columns represent a nonsignificant expansion of T cell clones in response to mutation-associated neopeptides. Translucent gray columns represent nonspecific clonotypic T cell expansions. b, For participant CGLU727 who attained PR and an OS of 46 months in the SBRT arm, the RHNO1 MANA-reactive TCR clone CASSIPGEGYTF was detected expanding at cycle 2. The TCR sequence is highlighted in red in the volcano plot, with the mutated gene in parentheses. c, For participant CGLU690 who attained PR with an OS of 102 months in the SBRT arm, the BDH1 MANA-reactive TCR clone CASSLWAGGGSREQFF was detected expanding at cycle 2. The TCR sequence is highlighted in green in the volcano plot, with the mutated gene in parentheses. C2, cycle 2 day 1; C3, cycle 3 day 1; C4, cycle 4 day 1; C3/C4, pooled PBMCs from cycle 3 day 1 and cycle 4 day 1.

Source data

Discussion

Although preclinical studies support the synergistic role of radiotherapy and immunotherapy at both irradiated and nonirradiated sites8,22,23,24,25, clinical trials for patients with advanced NSCLC have yielded mixed results. These shortcomings are amplified by the scarcity of studies supporting the systemic effects of radioimmunotherapy and induction of antitumor immune responses at nonirradiated tumor sites in the context of clinical trials26. To broaden our understanding of the systemic effects of radioimmunotherapy, we performed serial multiomic analyses of abscopal tumor sites together with their evolving TME and tied these findings to radiotherapy-induced peripheral and intratumoral T cell repertoire reshaping and MANA-reactive T cell responses in the PEMBRO-RT clinical trial of radioimmunotherapy (NCT02492568). These analyses revealed an induction of systemic immune responses with radioimmunotherapy including tumors harboring molecular features of immunotherapy resistance, likely driven by MANA cross-presentation in abscopal tumor sites. Our findings support the immunomodulatory effects of radiation together with a path forward for radioimmunotherapy as a potential strategy to overcome immunotherapy resistance.

Radiotherapy has been shown to drive immunogenic cell death through means such as induction of the cyclic GMP–AMP synthase (cGAS)–stimulator of IFN genes (STING) and type I IFN pathways10,27,28, induced upregulation of major histocompatibility complex (MHC) molecules29 and presentation of neoantigens at the irradiated site. While radiotherapy has also been shown to have immunosuppressive effects at the irradiated tumor site18 through induction of PDL1 expression on tumor cells30, addition of pembrolizumab, an anti-PD1 antibody, may be synergistic by counteracting this negative effect on the antitumor immune response and help explain why abscopal effects by radiotherapy alone are rare in the clinical setting. The premise of radiotherapy outside of local effects and disease control lies in the ability to elicit adaptive cytotoxic T cell responses recruited to nonirradiated sites2,31,32. Following radiotherapy, dendritic cells at the irradiated sites may capture tumor antigens and then cross-present these antigens in draining lymph nodes to prime new T cell responses33. In line with this notion11,34, the pronounced reshaping of the TCR repertoire, including expansions of new TCR clones, at nonirradiated sites observed in the SBRT cohort suggested a radiation-driven abscopal effect in our cohort. The presence of a radioimmunotherapy-induced systemic antitumor immune response was further supported by the identification of neoantigen-reactive T cell clones in autologous T cells from participants attaining radiographic response and long-term survival in the SBRT arm. While the yield of functional T cell assays may be limited after radiotherapy given the known radiosensitivity of circulating lymphocytes and T cells10, which may in turn be reflected in the smaller fraction of neoantigen-reactive T cell expansions detected at nonbaseline time points, our findings support MANA-reactive T cell responses in long-term survivors after radioimmunotherapy. In tandem, a broad array of inflammatory response gene expression programs were upregulated in nonirradiated tumor sites in our study, supporting the role of radioimmunotherapy in enriching systemic immune responses at nonirradiated sites beyond direct T cell cytotoxicity. Notably, induction of B cell responses on therapy was unique to the SBRT arm, with marked B cell expansion observed in a few SBRT participants who sustained durable clinical responses, highlighting the potential role of humoral adaptive responses in driving tumor regression after radioimmunotherapy. These findings align with our previous studies, supporting the role of SBRT in inducing systemic neoantigen-specific T cell responses in persons with early-stage NSCLC receiving definitive radiotherapy35.

Immunologically cold NSCLC tumors, characterized by low TMB or low PDL1 expression, are typically less likely to regress with ICI when administered as monotherapy36. We found that the systemic immune-stimulatory benefits of radioimmunotherapy coincided with significantly improved therapeutic response and longer survival in this subset of persons when compared with pembrolizumab alone. In contrast, such associations were not noted for participants with immunologically hot tumors characterized by high TMB or high PDL1 expression, suggesting that the addition of SBRT to pembrolizumab may have limited benefit for this subpopulation. Similarly, activation of the Wnt–β-catenin pathway has also been linked with tumor immune evasion20 and immunotherapy resistance37. Immunotherapy resistance may be driven by a causal relationship between tumor cell-intrinsic oncogenic activation of β-catenin and T cell exclusion, resulting in impaired host priming of antigen-specific T cells and impaired trafficking of effector T cells into the TME38. Interestingly, we detected an improved PFS for participants with Wnt-mutated tumors compared with wild-type tumors in the radioimmunotherapy arm. As a representative example, a tumor harboring an oncogenic CTNNB1 gain-of-function hotspot mutation and a hotspot missense likely Wnt-activating mutation in SFRP4 attained a complete response (CR) with radioimmunotherapy. The improved clinical response to radioimmunotherapy in participants with Wnt-mutated tumors may stem from the induction of cytotoxic T cell responses with radiation, partially overcoming the impaired priming and trafficking of T cell responses characteristic of these tumors.

While the statistical power of certain subset analyses was limited by the small sample size, potentially leading to type II error, our findings suggest that radioimmunotherapy may be effective in immunologically cold tumors that harbor features of primary resistance and may explain why clinical trials of unselected participants with solid tumors including advanced NSCLC have often yielded mixed results22,23,39,40,41,42. To this end, our findings may inform participant selection for radioimmunotherapy approaches including the subset of persons with primary resistance to immunotherapy that currently lacks effective treatment options. Certainly, given the multifaceted nature of immunologically cold tumors that are driven by the interplay between tumor genotypes and TME phenotypes, future studies are needed to validate the role of radioimmunotherapy in tumors with overlapping characteristics of immunotherapy resistance.

Tumor aneuploidy has previously been shown to correlate with immunotherapy resistance in persons with metastatic NSCLC treated with sequential radioimmunotherapy18. In our study, we found a numerically shorter yet not statistically significant PFS and OS for participants with highly aneuploid tumors in the SBRT arm. We believe that this difference may be in part attributed to the types of tumor samples analyzed, as our study focuses on nonirradiated and abscopal lesions rather than irradiated lesions. Furthermore, our study design did not allow for evaluation of the immunomodulatory effect of concurrent radioimmunotherapy driven by cGAS–STING signaling in the context of radiation-induced DNA damage and concurrent anti-CTLA4 therapy43, which may be heightened in tumors harboring a high degree of genomic instability18. As such, the role of tumor aneuploidy in therapy response may be context dependent and our findings highlight the complexity of identifying the subset of tumors and hosts that maximally benefit from radioimmunotherapy approaches.

Despite the findings of our study, the clinical translation of the immunomodulatory effects of radiotherapy has not been straightforward. Participant selection for radioimmunotherapy approaches remains key together with SBRT dose and timing. The underlying premise of radiotherapy lies first in its potential to sensitize immunologically cold tumors to ICI and overcome primary resistance in these populations and second in reinvigorating antitumor immune responses after progression on anti-PD(L)1 therapy. The latter is currently being tested in the RAD-IO clinical trial (NCT05401786), which incorporates rechallenging with anti-PD1 therapy after immune priming by ipilimumab and immune boosting by radiotherapy for participants with advanced NSCLC; in this trial, multiple metastatic sites are irradiated to counteract tumor heterogeneity and divergent antitumor immune responses. Additional clinical trials are needed to specifically investigate the benefit of radioimmunotherapy over immunochemotherapy or in overcoming immunotherapy resistance in persons with immunologically cold tumors. In conclusion, our study supports the abscopal effect of radioimmunotherapy in persons with metastatic NSCLC, including persons with immunologically cold tumors. Our findings suggest that radioimmunotherapy may be a promising avenue of further exploration for persons with therapeutic resistance to ICI; future efforts will specifically investigate the benefit of radioimmunotherapy in selected populations.

Methods

Cohort description and clinical response assessments

The study protocol was approved by the Institutional Review Board of Johns Hopkins University (Johns Hopkins Medicine Institutional Review Board) and the Netherlands Cancer Institute Antoni van Leeuwenhoek Institutional Review Board (registered under number CFMPB573). Written informed consent was provided by all study participants; participants were not compensated. We examined serial tumor and peripheral blood samples collected from participants treated with pembrolizumab (200 mg kg−1 every 3 weeks) either alone (control arm) or in combination with SBRT (three doses of 8 Gy to a single tumor lesion; SBRT arm) during the multicenter randomized phase 2 PEMBRO-RT clinical trial (NCT02492568)13. This trial enrolled 92 participants with advanced NSCLC between July 1, 2015 and March 31, 2018, regardless of PDL1 status. Of those 92 participants, 76 were randomized to the control arm (n = 40) or the SBRT arm (n = 36) and biospecimens were procured for all participants who received at least one dose of pembrolizumab (n = 72 total: n = 37 in the control arm and n = 35 in the SBRT arm). Treatment continued until confirmed radiographic progression, unacceptable toxicity, investigator decision, participant withdrawal of consent or a maximum of 12 months, extended to 24 months in September 2017 for alignment with the therapeutic landscape at the time. Therapy response was assessed by overall response rate at 12 weeks using RECIST 1.1. Tumors with radiographic CR or partial response (PR) were classified as responding, while tumors with radiographic stable disease (SD) or progressive disease (PD) were classified as nonresponding. PFS and OS, estimated from time of trial enrollment to progression or status at last follow-up, respectively, according to a data cutoff date of April 16, 2024, were used to capture long-term clinical outcomes. Two participants in the control arm experienced progression at week 6 followed by PR at week 12, which was considered pseudoprogression (as described in the original trial) and this was reflected in the therapy response assignments13. Participants attaining an OS of at least 3 years from time of trial enrollment were classified in the long-term response group (12 in the control arm and 8 in the SBRT arm). Clinical data collection and clinical trial endpoint determination were performed independent of the conditions of the experiments.

We leveraged 293 serial tumor and peripheral blood samples collected at baseline and after two cycles of pembrolizumab from 72 participants in the control (n = 37) and SBRT (n = 35) arms, for which biospecimens were procured. Tumor biopsies were serially obtained from a nonirradiated tumor lesion and matched baseline and on-therapy tumor and blood samples were used for genomic, transcriptomic and immunologic analyses. TMB-low (<300 mutations per exome; n = 43), PDL1-null (0% expression on IHC; n = 41) or Wnt-mutated (presence of mutations affecting genes in the Wnt pathway; n = 10) tumors were defined as immunologically cold. A cutoff of 300 mutations per exome was selected to optimize the difference in both PFS and OS between the TMB-low and TMB-high groups across the cohort.

WES

We performed WES on 116 samples (58 tumor–normal pairs) from 30 participants treated with pembrolizumab monotherapy (control) and 28 participants treated with SBRT followed by pembrolizumab (SBRT) (Supplementary Table 1). DNA was extracted from baseline tumor and matched peripheral blood samples, fragmented and used to prepare WES libraries with the Illumina TruSeq library kit. Exonic regions were captured in solution using the Agilent SureSelect v4 kit, followed by paired-end sequencing using Illumina HiSeq 2000/2500 instruments17,44,45. The mean depth of total and distinct coverage was 207× and 160× for tumor and 91× and 78× for normal samples, respectively (Supplementary Table 1). WES data were processed to identify somatic variants using VariantDx44,45 (Supplementary Table 2). Recurring mutations with a frequency of 10 or higher in the COSMIC46 database (version 100; https://cancer.sanger.ac.uk/cosmic/login) were considered cancer hotspots.

Mutation signature analysis

Mutation signatures were based on the fraction of coding point mutations in each of 96 trinucleotide contexts and the contribution of each signature to each tumor sample was estimated using the deconstructSigs R package (version 1.8.0) with the default ‘signatures.nature2013’ settings (https://cran.r-project.org/package=deconstructSigs) (Supplementary Table 5).

Aneuploidy assessment

We used FACETS 0.6.1 for assessing the purity of individual tumor samples, generating integer allele-specific copy-number profiles across the entire genome and determining the cellular fraction associated with each aberrant somatic copy-number alteration47. The extent of genome aneuploidy was estimated by the proportion of the genome with allelic imbalance17 and several additional metrics (ploidy, entropy, modal ploidy, nonmodal genome fraction, fraction of the genome with loss of heterozygosity and nondiploid genome fraction) (Supplementary Tables 5 and 6). To directly compare aneuploidy analyses in the PEMBRO-RT cohort with previously published studies18, we computed ASCETS aneuploidy scores per tumor sample and compared ASCETS scores between responding and nonresponding tumors.

RNA-seq, differential expression and enrichment analysis

We analyzed 48 tumor samples from 12 participants treated with pembrolizumab (12 baseline and 10 on therapy) and 16 participants treated with SBRT followed by pembrolizumab (14 baseline and 12 on therapy) (Supplementary Table 3). Total RNA was extracted from 10-µm formalin-fixed paraffin-embedded (FFPE) sections using the RNeasy FFPE kit (Qiagen). The quality of the obtained total RNA was evaluated by calculating the DV200 index, measured with the RNA 6000 Pico kit (Agilent Technologies). RNA-seq libraries were constructed through poly(A) selection (NEBNext poly(A) isolation kit) followed by reverse transcription to generate strand-specific complementary DNA libraries (NEBNext Ultra Directional RNA library prep kit for Illumina). Subsequently, paired-end sequencing (150 bp) was performed using Illumina NovaSeq 6000 S4, resulting in an average of 200 million total reads per library. RNA-seq data were aligned to the human transcriptome using STAR-2.7.3a (ref. 48), followed by RSEM-1.2.30 for isoform and gene-level quantification49. Transcripts associated with RNA genes, mitochondrial genes and ribosomal proteins were masked during the analysis. Sample-level quality control was performed using the total count, number of counts aligned to the genome, number of counts aligned to the transcriptome and percentage of reads aligning to the transcriptome for each sample. The batch effect was evaluated using both principal component analysis and correlation across the entire transcriptome in conjunction with universal human reference samples sequenced in each batch. Normalization of raw transcript counts and differential expression analysis were performed with DESeq2 1.42.0 (ref. 50) and the resulting P values were corrected for multiple testing using the Benjamini–Hochberg procedure. Normalized gene expression counts were analyzed with the fgsea48 (version 1.20.0)51 R package for GSEA with a preselected suite of gene sets (Supplementary Table 32), with resulting P values corrected for multiple testing. Genes that passed the count threshold were ranked by −log(P value) × sign(FC), where FC is the fold change. For TLS GSEA analyses, the TLS gene set included the following genes: MS4A1, CD4, MKI67, AICDA, MADCAM1, IL33, CDR2, CD40, CXCR5, LTB, CXCL13, CCL19, SELL, CCR7, CXCR4 and CCL5.

RNA-seq deconvolution

We used CIBERSORT (version 1.06)52 to perform deconvolution of transcriptome data, producing relative and absolute proportion estimates for 22 immune cell types. CIBERSORT uses a deconvolution-based approach with a reference gene signature matrix to assess the proportion of each immune cell type within the total immune cells present in the mixture. In our differential abundance analyses, we used the absolute abundance measurements of immune cell subsets. We used TRUST4 to reconstruct the BCR repertoires from transcriptomic data and better estimate B cell diversity and BCR repertoire at the specific BCR clonotype level53. We compared B cell densities between baseline and on-therapy tumors and by treatment arms and radiographic response.

TCR sequencing and enrichment analyses

TCR CDR3Vb sequencing was performed using serial peripheral blood and tumor tissue samples; we analyzed 129 samples from 31 participants (18 in the SBRT arm and 13 in the control arm), consisting of 64 blood samples (31 baseline, 31 on therapy and 2 resistance) and 65 tumor samples (31 baseline, 31 on therapy and 3 resistance) (Supplementary Table 4). DNA from baseline tumor and blood samples was isolated with Qiagen DNA FFPE and Qiagen DNA blood mini kits. TCR CDR3β regions were amplified using the ImmunoSeq survey assay with multiplex PCR using 45 forward primers to specific Vβ segments and 13 reverse primers to specific Jβ segments (Adaptive Biotechnologies)54. TCR sequences representing in-frame productive clones were further analyzed according to their CDR3 amino acid sequences. For each participant, clones achieving at least 0.05% relative abundance were included for differential abundance analysis between baseline and on-therapy time points in tumor and blood specimens. To conduct a differential abundance analysis between baseline and on-therapy tumors, we identified the most expanded and most regressed TCR clonotypes. These were determined on the basis of FC in the productive frequency of TCR clones, only counting clones with an FDR-adjusted P value < 0.05 (Mann–Whitney U-test). Aggregated clone counts with significant expansion dynamics (increasing or decreasing) between baseline and on-therapy samples (in tumor and/or blood) were collated and compared across participants by response and treatment categories using GLMs. Counts of expanded TCR clones were obtained in three compartments: tumor but not blood, blood but not tumor and both settings. For each compartment, association between clone count and treatment group was assessed using GLMs. Two sets of analyses were performed for each compartment, first including all clones that expanded on treatment (any level of pretreatment abundance) and then including only those clones that expanded on treatment but were not found in baseline samples (pretreatment abundance of 0, ‘newly expanded clones’). To adjust for TMB and PDL1 status, these variables were included in the GLM.

Functional T cell assays

We selected clonal expressed mutations detected at abscopal (biopsied) sites from three participants with radiographic responses and long-term OS in the SBRT arm and tested for neoantigen-reactive T cell responses in autologous T cells using the MANAFEST assay, which detects neoantigen-reactive T cell expansions with high sensitivity and specificity21,44. This approach combines ex vivo T cell culture and peptide stimulation with TCR sequencing to identify neoantigen-reactive T cell clonotypic expansions induced by neoantigens. From each participant, 7–13 somatic mutations were selected on the basis of predicted MHC class I binding rank of associated neoantigens, expression in participant-matched RNA-seq data, mutation clonality and mutation type (insertions and deletions that satisfied previous criteria were prioritized when present). Peptides containing the mutation were synthesized and are listed in Supplementary Table 28 (GenScript). T cells were isolated from peripheral blood mononuclear cells (PBMCs) by negative selection (EasySep; StemCell Technologies) from baseline, cycle 2 day 1, cycle 3 day 1 and cycle 4 day 1, cultured and stimulated in separate wells with each of the synthesized neopeptides21,44. TCR sequencing of extracted DNA from cultured CD8+ cells was performed by the Johns Hopkins Fest and TCR Immunogenomics Core Facility using the AmpliSeq for Illumina TCR beta-SR assay. Nonproductive TCR sequences were eliminated and aligned to obtain only the CDR3 region. Sequences not beginning with C or ending with F or W and having fewer than seven amino acids were eliminated. For singlet analyses performed for participant CGLU680 (Supplementary Table 29), processed data files were analyzed using the publicly available MANAFEST analysis web application (http://www.stat-apps.onc.jhmi.edu/FEST) to define neoantigen-specific T cell clonotypes. For participants CGLU690 and CGLU727, for whom adequate numbers of PBMCs were available for triplicate analyses, we analyzed TCR clone abundances per subject time points across peptide stimulation configurations. Multiple replicates per configuration were evaluated by (1) determining the set of TCR clones that were positive in the majority of replicates available for a given configuration and (2) determining clones at least fivefold greater in percentage abundance in the target condition (as measured by the lowest positive value) than the next highest single replicate from a different configuration. This procedure was performed for each time point within each participant separately (Supplementary Tables 30 and 31).

Statistical analysis and reproducibility

The progression-free interval was defined as the time from diagnosis to disease progression or death. Median point estimates and 95% confidence intervals for PFS and OS were calculated using the Kaplan–Meier method and survival curves were compared using the nonparametric log-rank test. Differences in genomic and transcriptomic features between responding and nonresponding tumors were assessed using the Fisher’s exact test for categorical variables and the Mann–Whitney test for continuous variables. P values were corrected using the Benjamini–Hochberg procedure and the FDR values were calculated. We further conducted multivariate Cox proportional hazards regression analyses to assess the independent association between SBRT therapy and PFS and OS in immunologically cold tumors while controlling for potential confounding variables (age, sex, tumor histology, smoking status and tumor purity). Statistical analyses were conducted using R version 3.6 and higher (http://cran.r-project.org). Nonparametric tests were used and, thus, data distribution was not tested for normality. No statistical methods were used to predetermine sample sizes but our sample sizes are similar to those reported in previous publications17. Randomization was not applicable for the exploratory multiomic analyses reported in this study. In the PEMBRO-RT study, participants were randomly assigned using a 1:1 ratio to receive treatment with pembrolizumab either after SBRT to a single tumor site (experimental arm) or without SBRT (control arm). While blinding is not entirely relevant outside the context of a clinical trial, clinical data collection and clinical trial endpoint determination were performed independent of the conditions of the experiments.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.