Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Oct 2.
Published in final edited form as: Essays Biochem. 2023 Sep 28;67(6):979–989. doi: 10.1042/EBC20220248

Immunologically relevant effects of radiation therapy on the tumor microenvironment

Claudia Galassi 1, Vanessa Klapp 2,3, Silvia C Formenti 1,4,5, Sandra Demaria 1,*, Lorenzo Galluzzi 1,5,6,*
PMCID: PMC10543618  NIHMSID: NIHMS1914452  PMID: 37199227

Abstract

Focal radiation therapy (RT) has been successfully employed to clinically manage multiple types of cancer for more than a century. Besides being preferentially cytotoxic for malignant cells over their non-transformed counterparts, RT elicits numerous microenvironmental alterations that appear to factor into its therapeutic efficacy. Here, we briefly discuss immunostimulatory and immunosuppressive microenvironmental changes elicited by RT and their impact on tumor recognition by the host immune system.

Keywords: CGAS, ICD, mtDNA, type IFN, TGFβ, TREG cells

Introduction

Focal radiation therapy (RT) has been a pillar in modern clinical cancer management for more than a century, both as a curative intervention (e.g., in patients with squamous cell carcinoma or anal carcinoma) and as a palliative measure (e.g., for the control of painful metastatic lesions to the bones from a variety of cancers) (1, 2). While recent technological advances including (but not limited to) stereotactic body RT (SBRT) and intensity-modulated RT (IMRT) have enabled an ever more precise delivery of increasing RT doses to specific anatomical location with improved sparing of adjacent healthy tissues (3, 4), the therapeutic effects of RT mainly build on the differential ability of transformed versus normal cells to repair RT-elicited macromolecular damage (5, 6). Indeed, at odds with their healthy counterparts, malignant cells generally exist in an altered biological state that is often associated with the constitutive activation of stress-response pathways, which ultimately limits their capacity to adapt to additional alterations of homeostasis (7, 8). More specifically, RT mediates cytostatic/cytotoxic effects by eliciting oxidative damage to macromolecules, especially (but not solely) DNA, either directly or – more so – upon the ionization of water molecules that in turn behave as oxidants (5, 6, 912). This is generally associated with the activation of the so-called DNA damage response (DDR) (13), a complex series of molecular events that can have diametrically opposed consequences on cellular homeostasis: (1) if DNA damage is sufficiently contained, the DDR promotes repair in the context of a temporary cell cycle arrest, followed by cell cycle re-entry and resumed proliferation; (2) if DNA is damaged beyond repair, the DDR either elicits a permanent proliferative arrest commonly known as cellular senescence (14) or triggers regulated variants of cell death (15), measures that are aimed at protecting global organismal homeostasis in the context of compromised cellular fitness in irradiated tissues (16).

Notably, most solid tumors exhibit a considerable degree of intratumoral heterogeneity (17), implying that not all malignant cells are equally sensitive to standard clinical RT schedules. Moreover, many non-transformed cells are also exposed to RT during irradiation as part of the integral dose exposure. These cells encompass all components of the tumor microenvironment (TME) and adjacent tissues, as well as peripheral blood mononuclear cells (PBMCs) and granulocytes circulating within the target volume while the beam is on (18). Some of these cells, especially circulating lymphocytes, are exquisitely radiosensitive and hence can be exposed to lethal RT doses (19), which explains the lymphopenic side effects associated with some RT procedures (e.g., spleen exposure during RT for the management of gastric carcinoma) (20). This is particularly relevant in the clinical setting as post-RT lymphopenia and/or the consequent increase in circulating neutrophil-to-leukocyte ratio (NLR) have been linked with poor prognosis in multiple cohorts of patients with cancer treated with RT (21, 22). Conversely, most terminally differentiated cells (e.g., skeletal muscle cells, neurons) are relatively radioresistant and thus survive RT employed at dose and fractionation schedules commonly employed in cancer patients (23).

Importantly, both failing and successful adaptation to the effects of RT (and exogenous stress in general) (16) in the TME and adjacent tissues is associated with the activation of numerous signaling pathways involved in tissue remodeling, inflammation and immunity, many of which ultimately factor in the therapeutic effects of RT (2426). Here, we provide an overview of immunostimulatory and immunosuppressive effects of focal RT and their impact on the recognition and elimination of cancer cells by the host immune system.

Immunostimulatory effects of RT

When used according to specific dose and fractionation schedules, focal RT can exert potent immunostimulatory effects, including (but not limited to): (1) the upregulation of genes encoding potentially immunogenic tumor-associated antigens (TAAs), (2) the overexpression of MHC class I molecules on the surface of irradiated cells, (3) the exposure of NK cell-activating ligands (NKALs), (4) the activation of immunogenic cell death (ICD), and (5) the upregulation of death receptors (DRs).

TAAs and MHC Class I molecules.

Three daily RT fractions of 8 Gy each promoted the expression of 4 proteins containing potentially immunogenic mutations in mouse triple negative breast cancer (TNBC) 4T1 cells, and the antigenic peptides of three of them – namely, cullin associated and neddylation disassociated 1 (CAND1), DEXH (Asp-Glu-X-His) box polypeptide 58 (DHX58) and adhesion G protein-coupled receptor F5 (ADGRF5) – were able to elicit CD8+ (CAND1 and DHX58) and CD4+ (ADGRF5) T cell responses that could be further boosted by RT. In line with this notion, neo-epitopes from all these proteins induced interferon gamma (IFNG, best known as IFNγ) responses in 4T1-draining lymph nodes, especially when combined with RT. Moreover, vaccination of mice with CAND1-derived, DHX58-derived and ADGRF5-derived epitopes improved the control of both irradiated 4T1 primary lesions as well as non-irradiated lung metastases, an effect that pointed to at least some degree of antigen spread (27). Along similar lines, focal RT delivered as a sub-cytotoxic single dose of 10 or 20 Gy has been shown to induce the expression of various TAAs of the carcinoembryonic antigen protein family as well as of mucin 1, cell surface associated (MUC1) in a panel of 17 human cancer cell lines (28).

Mouse glioma GL261 cells treated with a single dose of 4 Gy exhibited accrued levels of MHC Class I molecules on their surface, an effect that could be attributed to increased MHC Class I heavy chains and beta-2-microglobulin (B2M) expression (29). In line with this notion, whole-brain RT delivered to glioblastoma-bearing mice in 2 fractions of 4 Gy each resulted in recovered MHC Class I expression at the invading edge of the tumor and restored disease control by peripheral, vaccine-elicited CD8+ T cell responses (29). Along similar lines, human melanoma MelJuSo cells exposed to single RT dose of 1 Gy, 7 Gy and 25 Gy manifested a dose-dependent increase in surface-exposed MHC class I molecules, as did mouse colorectal carcinoma (CRC) MC38 cells exposed to a single RT of 2 Gy, 8 Gy and 10 Gy (30). Interestingly, such an effect (1) was accompanied by alterations in the MHC Class I immunopeptidome, and (2) could also be documented in vivo, in healthy mouse kidneys exposed to a single RT fraction of 25 Gy (30). Similar findings have been obtained in a panel of human cancer cell lines irradiated with a single dose of 8 Gy, an effect that could be enhanced by the pre-administration of the epigenetic modifier decitabine (31), as well as in MC38 cells responding to 10 Gy in a single fraction (32). Recent findings point to the transactivator NLR family CARD domain containing 5 (NLRC5) as to a transcriptional regulator involved in the ability of RT to promote MHC Class I expression (33). Moreover, it has recently been suggested that RT may induce the presentation of antigens generated during the pioneer round of translation from mRNAs containing nonsense mutations (34). However, whether the latter mechanism has a role in RT-driven anticancer immunity remains to be investigated.

NKALs.

Human lung adenocarcinoma NCI-H23 and A549 cells treated with a single RT dose of 8 Gy exhibited accrued levels of multiple NKALs specific for NK cell-activating receptor killer cell lectin like receptor K1 (KLRK1; best known as NKG2D), including MHC class I polypeptide-related sequence A (MICA), MICB, UL16 binding protein 1 (ULBP1), ULBP2 and ULBP3, an effect that could be exacerbated by histone deacetylase (HDAC) inhibitors (35). Moreover, a panel of mouse and human glioblastoma cells exhibited a dose-dependent increased in multiple NKALs upon irradiation with a single RT fraction of 2 Gy, 4Gy or 8 Gy, culminating with increased susceptibility to natural killer (NK)-mediated cytotoxicity (36). Similar results have been obtained by irradiation a panel of human cancer cell lines of multiple histological derivation with 20 Gy in a single fraction (37). These observations suggest that at least in some cancer cell lines, the upregulation of MHC Class I molecules elicited by RT (which potently inhibit NK cell activity) (38) may be effectively counteracted by an increased exposure of NKALs. That said, a fraction of CD8+ T cells express NKG2D (39), and the upregulation of NKALs by RT has been shown to stabilize the immune synapse between effector CD8+ T cells and cancer cells in mice treated with a CTLA4 blocker (40), suggesting that T cells may also contribute to tumor control by RT downstream of NKAL upregulation.

ICD and cytokines.

Mouse CRC CT26 cells, as well as mouse hormone receptor (HR)+ breast carcinoma TS/A cells exposed to γ irradiation in vitro can be effectively used as vaccines upon inoculation in immunocompetent syngeneic animals in the absence of any immunological adjuvant (41, 42), demonstrating that RT can elicit bona fide ICD (43). This reflects RT cytotoxicity as well as its ability to promote the emission of various adjuvant-like signals commonly known as damage-associated molecular patterns (DAMPs). Such signals encompass (1) the exposure of endoplasmic reticulum (ER) chaperones such as calreticulin on the plasma membrane, where it serves as an “eat-me” signal for dendritic cell (DC) precursors upon binding to LDL receptor-related protein 1 (LRP1, best known as CD91) (4446); (2) the autophagy-dependent secretion of ATP into the extracellular microenvironment, where it operates both as a chemoattractant, upon binding to purinergic receptor P2Y2 (P2RY2), and as an immunostimulatory signal, upon binding to purinergic receptor P2X 7 (P2RX7) (44, 4749); (3) the release of the non-histone chromatin-binding protein high mobility group box 1 (HMGB1), which mediates immunostimulatory effects upon binding to advanced glycosylation end-product-specific receptor (AGER, best known as RAGE) and Toll-like receptor 4 (TLR4) (44, 50, 51). Interestingly, abundant DAMP emission in response to irradiation has also been documented with carbon ions (52). Whether charged particles are superior to photon-based RT at eliciting ICD, however, remains to be formally demonstrated.

RT is also a potent inducer of type I interferon (IFN) secretion, which is not only a critical component of ICD signaling, but also a key factor for RT to elicit potent tumor-targeting immune responses with systemic outreach (i.e., so-called abscopal responses) (42, 5355). Interestingly, type I IFN secretion by irradiated malignant cells (and the consequent elicitation of abscopal responses) appears to be elicited by the accumulation of mitochondrial DNA (mtDNA) in the cytosol of irradiated cells (42, 56), culminating with the activation of cyclic GMP–AMP synthase (CGAS) signaling (57). This process is therefore particularly sensitive to mechanisms that limit either mtDNA availability, such as the dose-dependent upregulation of the mtDNA-degrading enzyme three prime repair exonuclease 1 (TREX1) (54) and the activation of autophagy (which degrades permeabilized, mtDNA-releasing mitochondria (42), or CGAS signaling, such as the activation of apoptotic caspases (which cleave CGAS) (58, 59). Interestingly, irradiated cancer cells can also elicit CGAS activation in dendritic cells (57), at least in part upon the delivery of extracellular vesicles containing CGAS-activating double-stranded DNA molecules (60). Importantly, RT has been shown to effectively restore proficient type I IFN signaling in preclinical lung cancer models, thus restoring sensitivity to ICIs (61). Supporting a role for type I IFN signaling in the clinical efficacy of RT, elevations in circulating interferon beta 1 (IFNB1) about two weeks after completion of palliative RT plus an immune checkpoint inhibitor (ICI) specific for cytotoxic T lymphocyte-associated protein 4 (CTLA4) were as strong predictor of response in a cohort of patients with non-small cell lung carcinoma (62).

Mouse 4T1, 67NR and 4T07A mammary carcinoma cell lines with different metastatic potential as well as human breast cancer HTB20 cells (but not non-transformed breast epithelial MCF10A cells) secreted chemokine (C-X-C motif) ligand 16 (CXCL16) in a dose-dependent fashion upon exposure to a single RT dose of 2 Gy, 6 Gy, and 12 Gy (63), exemplifying the notion that RT can promote the release of other cytokines beyond type I IFN (see below). Pointing to a role for CXCL16 signaling in the therapeutic effects of RT, 4T1-bearing mice lacking the CXCL16 receptor chemokine (C-X-C motif) receptor 6 (CXCR6) exhibited not only limited tumor infiltration by CXCR6+ T cells upon focal RT with a single dose of 12 Gy, but also reduced tumor control (63).In summary, RT stands out as a potent inducer of ICD, a particularly immunogenic form of cell death coupled to DAMP emission and cytokine secretion with profound clinical relevance.

Death receptors.

4T1 cells exposed to 3 RT fractions of 8 Gy expressed increased levels of the DRs Fas cell surface death receptor (FAS) and tumor necrosis factor receptor superfamily, member 10b (TNFRSF10B, best known as DR5 or TRAIL-R2) on their membrane, which along with an increase in surface-exposed MHC Class II molecules rendered them more susceptible to lysis by cytotoxic CD4+ T cells (27). Similar results have been obtained with human pancreatic adenocarcinoma Capan 2 cells engineered to express a model antigen (sLeA) upon exposure to a single dose of 2 Gy in vitro (64), as well as with human CD19+ acute lymphoblastic leukemia NALM cells irradiated with 1 Gy in vitro or in vivo (upon inoculation in immunodeficient hosts) (65). Importantly, in both these settings, low-dose RT delivered either focally (64) or as total body irradiation (TBI) (65) extended the therapeutic potential of TAA-targeting CAR T cells. However, while in the former setting such an increased response derived from TAA-independent, DR-mediated CAR T cells cytotoxicity (64), in the latter it reflected an improved expansion and persistence of CAR T cells (65). These observations delineate DR upregulation on malignant cells as yet another therapeutically relevant immunostimulatory effect of RT.

Immunosuppressive effects of RT

Depending on a number of factors, including (but not limited to) specifical experimental or clinical setting as well as dose and fractionation regimen, focal RT can also elicit immunosuppressive effects such as: (1) the secretion and activation of transforming growth factor beta 1 (TGFB1), inhibin subunit beta A (INHBA) and other cytokines/chemokines; (2) the accrued exposure on the plasma membrane of immunosuppressive factors such as CD274 (best known as PD-L1), CD47 and 5’-nucleotidase ecto (NT5E, best known as CD73); (3) vascular disruption coupled to decreased oxygen tension; and (4) chronic, indolent type I IFN signaling.

TGFB1, INHBA and other cytokines.

TGFB1 and INHBA are cytokines from the TGFβ family with potent immunosuppressive activity, at least in part reflecting their capacity to recruit CD4+CD25+FOXP3+ regulatory T (TREG) cells to the irradiated TME and to generate a potent fibrotic reaction (24, 66). In immunocompetent mice bearing bilateral MC38 tumors, TGFβ neutralization with a monoclonal antibody synergized with focal RT delivered to a single tumor in 3 fractions of 8 Gy coupled with systemic TNF receptor superfamily member 9 (TNFRSF9, best known as CD137 or 4–1BB agonism and programmed cell death 1 (PDCD1, best known as PD-1) blockage, ultimately enabling systemic disease eradication in a majority of mice (67). Importantly, this was paralleled by increased recruitment of cytotoxic CD8+ T cells to non-irradiated tumors and elevations in circulating IFNγ in the absence of remarkable toxicity for the hosts (67). Similar results have been obtained in immunocompetent mice bearing bilateral TS/A lesions or unilateral 4T1 lesions (which metastasize rapidly to the lungs) that were treated with 6 RT fraction of 5 Gy each at one disease site, plus a TGFβ-blocking antibody (68), as well as in immunocompetent mice bearing syngeneic SCC VII head and neck cancers, TC-1 lung carcinomas or LL2 lung carcinomas receiving a single RT dose of 8 Gy in combination with transforming growth factor beta receptor 1 (TGFBR1) or TGFBR2 inhibitors (69). Moreover, administration of the TGFβ blocker fresolimumab along with hypo-fractionated RT was well tolerated in a cohort of patients with metastatic breast carcinoma, and subjects receiving fresolimumab at the 10 mg/Kg dose had a favorable systemic immune response and experienced longer median overall survival than patients treated with fresolimumab 1 mg/Kg (70). Interestingly, TGFβ neutralization has been shown to exacerbate the ability of RT to elicit the secretion of INHBA by a panel of mouse and human breast cancer cell lines (71, 72). In line with this notion, Inhba deletion from 4T1 cells improved the ability of RT combined with TGFβ neutralization to control 4T1 in vivo, correlating with a strong reduction in tumor-infiltrating TREG cells (71, 72). Of note, TREG cells as well as immunosuppressive myeloid cells can also be recruited to the irradiated TME upon the secretion of C-C motif chemokine ligand 2 (CCL2) (73). Accordingly, the therapeutic effects of a single RT fraction of 7.5 Gy against mouse TC-1 lung carcinomas growing in immunocompetent hosts were improved in Ccr2−/− mice (which lack the main CCL2 receptor) as compared to wild-type mice (73). In summary, multiple cytokines secreted by irradiated cancer cells support the establishment of an immunosuppressive TME that limit therapeutic responses to RT.

PD-L1, CD47 and CD73.

One single RT dose of 12 Gy delivered to BALB/c mice bearing TUBO mammary carcinomas promoted an accrued exposure of PD-L1 on the surface of malignant cells, an immunosuppressive effect that could be effectively offset by the co-administration of ICIs targeting PD-1 (74). Along similar lines, mouse Res177, Res499, and B16F10 melanomas established in immunocompetent, syngeneic hosts and treated with RT in a single fraction of 20 Gy plus a CTLA4-targeting ICI evaded immune control via PD-L1 upregulation, and this could be effectively limited by PD-1 blockage (75). Accordingly, cancer cells from patients with melanoma failing to respond to SBRT plus a CTLA4 blocker in the context of a clinical trial exhibited high levels of PD-L1 and signs of persistence CD8+ T cell exhaustion (75). Of note, PD-L1 upregulation on the surface of malignant cells from irradiated tumors have also been shown to reflect CD8+ T cell infiltration and IFNγ secretion (76).

Exposure of human HCT116 and HT29 CRC cells to a single RT dose of 8 Gy elicited not only increased levels of membrane-exposed PD-L1, but elevations of the “don’t eat-me signal” CD47, an effect that was ascribed to ATR serine/threonine kinase (ATR) signaling as elicited by DNA damage (77). In line with these observations, immunocompetent mice bearing bilateral MC38 lesions experienced superior tumor control at both disease site upon irradiation of one lesion with a single RT fraction of 8 Gy combined with systemic PD-1 and CD47 blockage (77). Similar results have been obtained in wild-type mice bearing bilateral small cell lung carcinoma (SCLC) lesions receiving RT in one fraction of 5 Gy or 5 fractions of 4 Gy each to a single lesion combined with systemic CD47 blockage (78, 79). Finally, RT has been shown to promote the dose-dependent upregulation of CD73 – a plasma membrane enzyme that facilitates the conversion of extracellular ATP into the immunosuppressive metabolite adenosine (80) – in a panel of human and mouse breast cancer cell lines (81). Supporting the therapeutic relevance of this pathway, CD73 blockage with a monoclonal antibody improved the capacity of RT delivered as a single dose of 20 Gy along with systemic CTLA4 inhibition to elicit abscopal responses in immunocompetent mice bearing bilateral TS/A lesions (81). These observations delineate PD-L1, CD47 and CD73 as potential targets to limit RT-driven immunosuppression in support of superior therapeutic responses.

Vascular disruption and hypoxia.

Focal RT is relatively cytotoxic to endothelial cells, which – combined with an increase in interstitial pressure due to RT-driven inflammatory reactions – can further limit oxygen availability in (at least some regions of) the TME (82). This is relevant not only because pre-existing as well newly formed hypoxic TME regions are particularly resistant to the cytotoxic effects of RT (83), but also because vascular disruption and hypoxia promote local immunosuppression by a number of mechanisms. These include (but are not limited to): (1) the downregulation of MHC class I molecules on the surface of malignant cells (84); (2) the upregulation of immunosuppressive factors such as PD-L1, vascular endothelial growth factor A (VEGFA) and colony stimulating factor 1 (CSF1) (8587); (3) the activation of bona fide CD4+CD25+FOXP3+ TREG cells and immunosuppressive DC populations (88); (4) the inhibition of CD8+ T cell effector functions coupled to the conversion of exhausted T (TEX) cells into TREG-like cells (89, 90); and (5) the repolarization of TAMs towards an immunosuppressive (M2-like) phenotype (91, 92). In line with these observations, various strategies to limit hypoxia have been shown to increase the efficacy of RT in preclinical tumor models, although the actual contribution of anticancer immunity to this effect remains to be formally elucidated. Despite this unknown, the ability of RT to kill cancer cells in well-perfused tumor regions promotes the selection of hypoxic areas of the TME, where immunosuppression further limits therapeutic responses.

Chronic type I IFN.

While acute, robust and resolving type I IFN responses have been shown to support clinically relevant anticancer immunity as driven by RT (54, 62), intratumoral type I IFN levels as well as genetic signatures of type I IFN signaling have also been associated with treatment resistance in various cohorts of cancer patients (58, 93). The reasons for such an apparent discrepancy remain to be completely explained. Suboptimal and chronic type I IFN signaling has been shown to support the accumulation of cancer stem cell (CSC)-like cells that enable accelerated tumor progression in immunocompetent mouse models of fibrosarcoma exposed to ICD-chemotherapeutics (94), pointing to the intensity and duration of type I signaling as to key determinant of ultimate therapeutic outcome. In line with this possibility, chronic type I IFN signaling has been consistently associated with CD8+ T cell exhaustion and immunotherapy resistance (9597). Taken together, these observations suggest efficient immunostimulation by RT should induce robust and resolving (as opposed to indolent and chronic) type I IFN responses. Whether dose and fractionation can affect the magnitude and kinetic of type IFN signaling as driven by RT remains to be defined.

Conclusions

It is now clear that RT imposes immunologically relevant alterations to the TME that (at least in some settings) influence local and/or systemic disease control (Figure 1). In line with this notion, RT has been shown to synergize with numerous immunotherapeutic or immunomodulatory agents in a variety of immunocompetent mouse tumor models. These agents include (but are not limited to): (1) ICIs targeting CTLA4, PD-1, PD-L1, TGFβ or INHBA (54, 68, 71, 74, 98); (2) immunostimulatory monoclonal antibodies specific for TNF receptor superfamily member 9 (TNFRSF9; best known as CD137 or 4–1BB), TNFRSF4 (best known as OX40) or CD40 (67, 99); (3) recombinant cytokines that stimulate lymphoid cells, such as IL15 (100); and (4) other immunomodulatory molecules with diverse mechanisms of action, such as Toll-like receptor 7 (TLR7) agonists, CDK4/6 inhibitors and 5’-nucleotidase ecto (NT5E; best known as CD73) blockers (81, 101, 102). However, combining RT with IT in the clinic has been less straightforward than expected. Indeed, while some prospective randomized clinical trials have demonstrated a survival advantage for patients receiving IT in addition to RT-based standard of care (SOC) over SOC alone (103), others failed to document such a benefit (104106). While many reasons can be invoked to explain the lack of synergy between RT and IT in some of these studies, it is tempting to speculate that the use of RT according to conventional doses, fractionation, schedules and target volumes may not always be optimal to preserve the immune fitness of the host in support of IT responsiveness (107). Additional work is required to delineate improved RT approaches that optimally cooperate with IT in patients with cancer.

Figure 1. Main immunomodulatory mechanisms elicited by RT in the TME.

Figure 1.

Focal radiation therapy (RT) has been shown to impose various immunologically relevant changes to the tumor microenvironment (TME) that may influence local and systemic cancer control. A. On the one hand, focal RT has been shown to (1) render malignant cells more immunogenic upon the derepression of genes encoding tumor-associated antigens (TAAs); (2) cause a potently immunostimulatory form of cell deaths coupled with the acute release of danger signals, cytokines and chemokines; (3) favor the recognition of malignant cells by the immune system via the upregulation of MHC Class I molecules or natural killer (NK) cell-activating ligands (NKALs) on the cell surface; and (4) increase the sensitivity of cancer cells to immune effectors as consequence of death receptor (DR) upregulation. B. On the other hand, focal RT has been associated with the establishment of local immunosuppression as a consequence of: (1) three prime repair exonuclease 1 (TREX1) upregulation and hence degradation of potentially interferogenic double-stranded DNA (dsDNA) species in the cytosol of irradiated cells; (2) transforming growth factor beta 1 (TGFB1, best known as TGFβ) and inhibin subunit beta A (INHBA) signaling, resulting in regulatory T (TREG) recruitment and fibrosis; (3) CD274 (best known as PD-L1) upregulation on the surface of malignant cells; (4) repolarization of tumor-associated macrophages (TAMs) towards an M2-like, immunosuppressive phenotype; and (5) endothelial cell cytotoxicity coupled with accrued interstitial pressure, culminating with dynamic changes in oxygenation and potential selection of hypoxic tumor areas with accrued immunosuppressive features. A variety of therapeutic strategies impinging on these and other immunologically relevant microenvironmental alterations elicited by RT has been shown to ameliorate the therapeutic profile of RT in immunocompetent tumor models.

Summary points.

  • Focal radiation therapy (RT) is an established treatment modality for multiple tumor types

  • RT mediates cytostatic/cytotoxic effects preferentially on malignant cells, largely reflecting their reduced capacity to repair RT-induced macromolecular damage compared to normal cells

  • RT also causes microenvironmental alterations that may factor into its therapeutic activity

  • Immunostimulatory mechanisms engaged by RT, such as the activation of immunogenic cell death (ICD) coupled to the release of type I IFN in the microenvironment of irradiated tumors, may support anticancer immunity

  • Conversely, RT-driven immunosuppressive pathways including TGFβ signaling and the recruitment of TREG cells to the tumor microenvironment (TME) may favor cancer immune escape

  • Immunostimulatory and immunosuppressive alterations elicited by radiation therapy in the TME may be harnessed to improve the therapeutic effects of RT

Acknowledgements.

CG is supported by a fellowship from the American Italian Cancer Foundation (AICF; #223565–01). VK is supported by a grant from the Luxembourg National Research Fund (FNR) (PRIDE19/14254520/i2TRON). SCF is supported by a U54 grant from NIH/NCI (#CA274291), by a Transformative Breast Cancer Consortium Grant from the US DoD BCRP (#W81XWH2120034) by a Breakthrough Level 3 grant from the US DoD BCRP (#W81XWH1910141), by a Breakthrough Level 2 grant from the US DoD BCRP (#W81XWH-1910101) and by a grant from the Breast Cancer Research Foundation (BCRF-22-053). SD is supported by an R01 grant from NIH/NCI (#R01CA201246), a grant from the STARR Cancer Consortium (#I14–0043), by a Breakthrough Level 3 grant from the US DoD BCRP (#W81XWH1910142), by a grant from the BCRF, and by funding from Boehringer Ingelheim (#BIP578201). LG is/has been supported (as a PI unless otherwise indicated) by two Breakthrough Level 2 grants from the US DoD BCRP (#BC180476P1; #BC210945), by a grant from the STARR Cancer Consortium (#I16–0064), by a Transformative Breast Cancer Consortium Grant from the US DoD BCRP (#W81XWH2120034, PI: Formenti), by a U54 grant from NIH/NCI (#CA274291, PI: Deasy, Formenti, Weichselbaum), by the 2019 Laura Ziskin Prize in Translational Research (#ZP-6177, PI: Formenti) from the Stand Up to Cancer (SU2C), by a Mantle Cell Lymphoma Research Initiative (MCL-RI, PI: Chen-Kiang) grant from the Leukemia and Lymphoma Society (LLS), by a Rapid Response Grant from the Functional Genomics Initiative (New York, US), by startup funds from the Dept. of Radiation Oncology at Weill Cornell Medicine (New York, US), by industrial collaborations with Lytix Biopharma (Oslo, Norway), Promontory (New York, US) and Onxeo (Paris, France), as well as by donations from Promontory (New York, US), the Luke Heller TECPR2 Foundation (Boston, US), Sotio a.s. (Prague, Czech Republic), Lytix Biopharma (Oslo, Norway), Onxeo (Paris, France), Ricerchiamo (Brescia, Italy), and Noxopharm (Chatswood, Australia).

Footnotes

Competing Interests.

SCF is/has been holding research contracts with Merck, Varian, Bristol Myers Squibb, Celldex, Regeneron, Eisai, and Eli-Lilly, and has received consulting/advisory honoraria from Bayer, Bristol Myers Squibb, Varian, Elekta, Regeneron, Eisai, AstraZeneca, MedImmune, Merck US, EMD Serono, Accuray, Boehringer Ingelheim, Roche, Genentech, AstraZeneca, View Ray and Nanobiotix. SD is/has been holding research contracts with Lytix Biopharma, Nanobiotix, and Boehringer Ingelheim, has received consulting/advisory honoraria from Lytix Biophrama and Johnson & Johnson Enterprise Innovation Inc. LG is/has been holding research contracts with Lytix Biopharma, Promontory and Onxeo, has received consulting/advisory honoraria from Boehringer Ingelheim, AstraZeneca, OmniSEQ, Onxeo, The Longevity Labs, Inzen, Imvax, Sotio, Promontory, Noxopharm, EduCom, and the Luke Heller TECPR2 Foundation, and holds Promontory stock options.

References

  • 1.Castaneda SA, Romak LB. Radiotherapy for Anal Cancer: Intensity-Modulated Radiotherapy and Future Directions. Surg Oncol Clin N Am. 2017;26(3):467–75. 10.1016/j.soc.2017.01.004 [DOI] [PubMed] [Google Scholar]
  • 2.Mercadante S Breakthrough cancer pain in the radiotherapy setting: a systematic and critical review. Expert Rev Anticancer Ther. 2023;23(3):229–34. 10.1080/14737140.2023.2182773 [DOI] [PubMed] [Google Scholar]
  • 3.Lo SS, Fakiris AJ, Chang EL, Mayr NA, Wang JZ, Papiez L, et al. Stereotactic body radiation therapy: a novel treatment modality. Nat Rev Clin Oncol. 2010;7(1):44–54. 10.1038/nrclinonc.2009.188 [DOI] [PubMed] [Google Scholar]
  • 4.Martin JM, Frantzis J, Eade T, Chung P. Clinician’s guide to prostate IMRT plan assessment and optimisation. J Med Imaging Radiat Oncol. 2010;54(6):569–75. 10.1111/j.1754-9485.2010.02217.x [DOI] [PubMed] [Google Scholar]
  • 5.Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Vanden Berghe T, et al. Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis. 2020;11(11):1003. 10.1038/s41419-020-03209-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bednarski JJ, Sleckman BP. At the intersection of DNA damage and immune responses. Nat Rev Immunol. 2019;19(4):231–42. 10.1038/s41577-019-0135-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Petroni G, Buqué A, Coussens LM, Galluzzi L. Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat Rev Drug Discov. 2022;21(6):440–62. 10.1038/s41573-022-00415-5 [DOI] [PubMed] [Google Scholar]
  • 8.Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 2009;136(5):823–37. 10.1016/j.cell.2009.02.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30(2):146–62. 10.1038/s41422-019-0263-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Matthews HK, Bertoli C, de Bruin RAM. Cell cycle control in cancer. Nat Rev Mol Cell Biol. 2022;23(1):74–88. 10.1038/s41580-021-00404-3 [DOI] [PubMed] [Google Scholar]
  • 11.Pilié PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019;16(2):81–104. 10.1038/s41571-018-0114-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ye LF, Chaudhary KR, Zandkarimi F, Harken AD, Kinslow CJ, Upadhyayula PS, et al. Radiation-Induced Lipid Peroxidation Triggers Ferroptosis and Synergizes with Ferroptosis Inducers. ACS Chem Biol. 2020;15(2):469–84. 10.1021/acschembio.9b00939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Groelly FJ, Fawkes M, Dagg RA, Blackford AN, Tarsounas M. Targeting DNA damage response pathways in cancer. Nat Rev Cancer. 2023;23(2):78–94. 10.1038/s41568-022-00535-5 [DOI] [PubMed] [Google Scholar]
  • 14.Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular Senescence: Defining a Path Forward. Cell. 2019;179(4):813–27. 10.1016/j.cell.2019.10.005 [DOI] [PubMed] [Google Scholar]
  • 15.Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25(3):486–541. 10.1038/s41418-017-0012-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Galluzzi L, Yamazaki T, Kroemer G. Linking cellular stress responses to systemic homeostasis. Nat Rev Mol Cell Biol. 2018;19(11):731–45. 10.1038/s41580-018-0068-0 [DOI] [PubMed] [Google Scholar]
  • 17.Vitale I, Shema E, Loi S, Galluzzi L. Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med. 2021;27(2):212–24. 10.1038/s41591-021-01233-9 [DOI] [PubMed] [Google Scholar]
  • 18.Heylmann D, Ponath V, Kindler T, Kaina B. Comparison of DNA repair and radiosensitivity of different blood cell populations. Sci Rep. 2021;11(1):2478. 10.1038/s41598-021-81058-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nakamura N, Kusunoki Y, Akiyama M. Radiosensitivity of CD4 or CD8 positive human T-lymphocytes by an in vitro colony formation assay. Radiat Res. 1990;123(2):224–7 [PubMed] [Google Scholar]
  • 20.Lee SF, Yip PL, Wong A, Ng F, Koh V, Wong LC, et al. Splenic irradiation contributes to grade ≥ 3 lymphopenia after adjuvant chemoradiation for stomach cancer. Clin Transl Radiat Oncol. 2022;36:83–90. 10.1016/j.ctro.2022.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen D, Verma V, Patel RR, Barsoumian HB, Cortez MA, Welsh JW. Absolute Lymphocyte Count Predicts Abscopal Responses and Outcomes in Patients Receiving Combined Immunotherapy and Radiation Therapy: Analysis of 3 Phase 1/2 Trials. Int J Radiat Oncol Biol Phys. 2020;108(1):196–203. 10.1016/j.ijrobp.2020.01.032 [DOI] [PubMed] [Google Scholar]
  • 22.Punjabi A, Barrett E, Cheng A, Mulla A, Walls G, Johnston D, et al. Neutrophil-Lymphocyte Ratio and Absolute Lymphocyte Count as Prognostic Markers in Patients Treated with Curative-intent Radiotherapy for Non-small Cell Lung Cancer. Clin Oncol (R Coll Radiol). 2021;33(8):e331–e8. 10.1016/j.clon.2021.03.019 [DOI] [PubMed] [Google Scholar]
  • 23.Shi L, Molina DP, Robbins ME, Wheeler KT, Brunso-Bechtold JK. Hippocampal neuron number is unchanged 1 year after fractionated whole-brain irradiation at middle age. Int J Radiat Oncol Biol Phys. 2008;71(2):526–32. 10.1016/j.ijrobp.2008.02.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cytlak UM, Dyer DP, Honeychurch J, Williams KJ, Travis MA, Illidge TM. Immunomodulation by radiotherapy in tumour control and normal tissue toxicity. Nat Rev Immunol. 2022;22(2):124–38. 10.1038/s41577-021-00568-1 [DOI] [PubMed] [Google Scholar]
  • 25.Rodriguez-Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol. 2020;21(2):120–34. 10.1038/s41590-019-0561-4 [DOI] [PubMed] [Google Scholar]
  • 26.McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17. 10.1038/s41568-020-0246-1 [DOI] [PubMed] [Google Scholar]
  • 27.Lhuillier C, Rudqvist NP, Yamazaki T, Zhang T, Charpentier M, Galluzzi L, et al. Radiotherapy-exposed CD8+ and CD4+ neoantigens enhance tumor control. J Clin Invest. 2021;131(5). 10.1172/JCI138740 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004;64(21):7985–94. 10.1158/0008-5472.CAN-04-1525 [DOI] [PubMed] [Google Scholar]
  • 29.Newcomb EW, Demaria S, Lukyanov Y, Shao Y, Schnee T, Kawashima N, et al. The combination of ionizing radiation and peripheral vaccination produces long-term survival of mice bearing established invasive GL261 gliomas. Clin Cancer Res. 2006;12(15):4730–7. 10.1158/1078-0432.CCR-06-0593 [DOI] [PubMed] [Google Scholar]
  • 30.Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203(5):1259–71. 10.1084/jem.20052494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Son CH, Lee HR, Koh EK, Shin DY, Bae JH, Yang K, et al. Combination treatment with decitabine and ionizing radiation enhances tumor cells susceptibility of T cells. Sci Rep. 2016;6:32470. 10.1038/srep32470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tailor A, Estephan H, Parker R, Woodhouse I, Abdulghani M, Nicastri A, et al. Ionizing Radiation Drives Key Regulators of Antigen Presentation and a Global Expansion of the Immunopeptidome. Mol Cell Proteomics. 2022;21(11):100410. 10.1016/j.mcpro.2022.100410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zebertavage LK, Alice A, Crittenden MR, Gough MJ. Transcriptional Upregulation of NLRC5 by Radiation Drives STING- and Interferon-Independent MHC-I Expression on Cancer Cells and T Cell Cytotoxicity. Sci Rep. 2020;10(1):7376. 10.1038/s41598-020-64408-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Uchihara Y, Permata TBM, Sato H, Kawabata-Iwakawa R, Katada S, Gu W, et al. DNA damage promotes HLA class I presentation by stimulating a pioneer round of translation-associated antigen production. Mol Cell. 2022;82(14):2557–70 e7. 10.1016/j.molcel.2022.04.030 [DOI] [PubMed] [Google Scholar]
  • 35.Son CH, Keum JH, Yang K, Nam J, Kim MJ, Kim SH, et al. Synergistic enhancement of NK cell-mediated cytotoxicity by combination of histone deacetylase inhibitor and ionizing radiation. Radiat Oncol. 2014;9:49. 10.1186/1748-717x-9-49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Weiss T, Schneider H, Silginer M, Steinle A, Pruschy M, Polić B, et al. NKG2D-Dependent Antitumor Effects of Chemotherapy and Radiotherapy against Glioblastoma. Clin Cancer Res. 2018;24(4):882–95. 10.1158/1078-0432.Ccr-17-1766 [DOI] [PubMed] [Google Scholar]
  • 37.Kim JY, Son YO, Park SW, Bae JH, Chung JS, Kim HH, et al. Increase of NKG2D ligands and sensitivity to NK cell-mediated cytotoxicity of tumor cells by heat shock and ionizing radiation. Exp Mol Med. 2006;38(5):474–84. 10.1038/emm.2006.56 [DOI] [PubMed] [Google Scholar]
  • 38.Wolf NK, Kissiov DU, Raulet DH. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol. 2023;23(2):90–105. 10.1038/s41577-022-00732-1 [DOI] [PubMed] [Google Scholar]
  • 39.Harris R, Mammadli M, Hiner S, Suo L, Yang Q, Sen JM, et al. TCF-1 regulates NKG2D expression on CD8 T cells during anti-tumor responses. Cancer Immunol Immunother. 2022. 10.1007/s00262-022-03323-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ruocco MG, Pilones KA, Kawashima N, Cammer M, Huang J, Babb JS, et al. Suppressing T cell motility induced by anti-CTLA-4 monotherapy improves antitumor effects. J Clin Invest. 2012;122(10):3718–30. 10.1172/JCI61931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, et al. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007;14(10):1848–50. 10.1038/sj.cdd.4402201 [DOI] [PubMed] [Google Scholar]
  • 42.Yamazaki T, Kirchmair A, Sato A, Buque A, Rybstein M, Petroni G, et al. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol. 2020;21(10):1160–71. 10.1038/s41590-020-0751-0 [DOI] [PubMed] [Google Scholar]
  • 43.Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol. 2022;23(4):487–500. 10.1038/s41590-022-01132-2 [DOI] [PubMed] [Google Scholar]
  • 44.Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. 2014;3:e28518. 10.4161/onci.28518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fucikova J, Spisek R, Kroemer G, Galluzzi L. Calreticulin and cancer. Cell Res. 2021;31(1):5–16. 10.1038/s41422-020-0383-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, et al. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123(2):321–34. 10.1016/j.cell.2005.08.032 [DOI] [PubMed] [Google Scholar]
  • 47.Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461(7261):282–6. 10.1038/nature08296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15(10):1170–8. 10.1038/nm.2028 [DOI] [PubMed] [Google Scholar]
  • 49.Kepp O, Bezu L, Yamazaki T, Di Virgilio F, Smyth MJ, Kroemer G, et al. ATP and cancer immunosurveillance. Embo j. 2021;40(13):e108130. 10.15252/embj.2021108130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191–5. 10.1038/nature00858 [DOI] [PubMed] [Google Scholar]
  • 51.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13(9):1050–9. 10.1038/nm1622 [DOI] [PubMed] [Google Scholar]
  • 52.Zhou H, Tu C, Yang P, Li J, Kepp O, Li H, et al. Carbon ion radiotherapy triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy in mice. Oncoimmunology. 2022;11(1):2057892. 10.1080/2162402x.2022.2057892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.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(7):2488–96. 10.1158/0008-5472.CAN-10-2820 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.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. 10.1038/ncomms15618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15(17):5379–88. 10.1158/1078-0432.CCR-09-0265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Marchi S, Guilbaud E, Tait SWG, Yamazaki T, Galluzzi L. Mitochondrial control of inflammation. Nat Rev Immunol. 2023;23(3):159–73. 10.1038/s41577-022-00760-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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(5):843–52. 10.1016/j.immuni.2014.10.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rodriguez-Ruiz ME, Buqué A, Hensler M, Chen J, Bloy N, Petroni G, et al. Apoptotic caspases inhibit abscopal responses to radiation and identify a new prognostic biomarker for breast cancer patients. Oncoimmunology. 2019;8(11):e1655964. 10.1080/2162402x.2019.1655964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ning X, Wang Y, Jing M, Sha M, Lv M, Gao P, et al. Apoptotic Caspases Suppress Type I Interferon Production via the Cleavage of cGAS, MAVS, and IRF3. Mol Cell. 2019;74(1):19–31.e7. 10.1016/j.molcel.2019.02.013 [DOI] [PubMed] [Google Scholar]
  • 60.Diamond JM, Vanpouille-Box C, Spada S, Rudqvist NP, Chapman JR, Ueberheide BM, et al. Exosomes Shuttle TREX1-Sensitive IFN-Stimulatory dsDNA from Irradiated Cancer Cells to DCs. Cancer Immunol Res. 2018;6(8):910–20. 10.1158/2326-6066.CIR-17-0581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang X, Schoenhals JE, Li A, Valdecanas DR, Ye H, Zang F, et al. Suppression of Type I IFN Signaling in Tumors Mediates Resistance to Anti-PD-1 Treatment That Can Be Overcome by Radiotherapy. Cancer Res. 2017;77(4):839–50. 10.1158/0008-5472.CAN-15-3142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier C, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 2018;24(12):1845–51. 10.1038/s41591-018-0232-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008;181(5):3099–107. 10.4049/jimmunol.181.5.3099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.DeSelm C, Palomba ML, Yahalom J, Hamieh M, Eyquem J, Rajasekhar VK, et al. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape. Mol Ther. 2018;26(11):2542–52. 10.1016/j.ymthe.2018.09.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yamazaki T, Sugita M, Martinet J, Boyer O, Galluzzi L, Guzman ML, et al. Boosting CAR T Cell Expansion and Therapeutic Activity with Low-dose Radiation Therapy. Int J Radiat Oncol. 2020;108(3):S158–S9 [Google Scholar]
  • 66.Batlle E, Massagué J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity. 2019;50(4):924–40. 10.1016/j.immuni.2019.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rodríguez-Ruiz ME, Rodríguez I, Mayorga L, Labiano T, Barbes B, Etxeberria I, et al. TGFβ Blockade Enhances Radiotherapy Abscopal Efficacy Effects in Combination with Anti-PD1 and Anti-CD137 Immunostimulatory Monoclonal Antibodies. Mol Cancer Ther. 2019;18(3):621–31. 10.1158/1535-7163.Mct-18-0558 [DOI] [PubMed] [Google Scholar]
  • 68.Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, et al. TGFβ Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res. 2015;75(11):2232–42. 10.1158/0008-5472.Can-14-3511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hamon P, Gerbe De Thore M, Classe M, Signolle N, Liu W, Bawa O, et al. TGFbeta receptor inhibition unleashes interferon-beta production by tumor-associated macrophages and enhances radiotherapy efficacy. J Immunother Cancer. 2022;10(3). 10.1136/jitc-2021-003519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, et al. Focal Irradiation and Systemic TGFbeta Blockade in Metastatic Breast Cancer. Clin Cancer Res. 2018;24(11):2493–504. 10.1158/1078-0432.CCR-17-3322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.De Martino M, Daviaud C, Diamond JM, Kraynak J, Alard A, Formenti SC, et al. Activin A Promotes Regulatory T-cell-Mediated Immunosuppression in Irradiated Breast Cancer. Cancer Immunol Res. 2021;9(1):89–102. 10.1158/2326-6066.CIR-19-0305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.De Martino M, Daviaud C, Vanpouille-Box C. Activin A backs-up TGF-ß to promote regulatory T cells. Oncoimmunology. 2021;10(1):1883288. 10.1080/2162402x.2021.1883288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mondini M, Loyher PL, Hamon P, Gerbe de Thore M, Laviron M, Berthelot K, et al. CCR2-Dependent Recruitment of Tregs and Monocytes Following Radiotherapy Is Associated with TNFalpha-Mediated Resistance. Cancer Immunol Res. 2019;7(3):376–87. 10.1158/2326-6066.CIR-18-0633 [DOI] [PubMed] [Google Scholar]
  • 74.Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687–95. 10.1172/JCI67313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–7. 10.1038/nature14292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Mandai M, Hamanishi J, Abiko K, Matsumura N, Baba T, Konishi I. Dual Faces of IFNγ in Cancer Progression: A Role of PD-L1 Induction in the Determination of Pro- and Antitumor Immunity. Clin Cancer Res. 2016;22(10):2329–34. 10.1158/1078-0432.Ccr-16-0224 [DOI] [PubMed] [Google Scholar]
  • 77.Hsieh RC, Krishnan S, Wu RC, Boda AR, Liu A, Winkler M, et al. ATR-mediated CD47 and PD-L1 up-regulation restricts radiotherapy-induced immune priming and abscopal responses in colorectal cancer. Sci Immunol. 2022;7(72):eabl9330. 10.1126/sciimmunol.abl9330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Nishiga Y, Drainas AP, Baron M, Bhattacharya D, Barkal AA, Ahrari Y, et al. Radiotherapy in combination with CD47 blockade elicits a macrophage-mediated abscopal effect. Nat Cancer. 2022;3(11):1351–66. 10.1038/s43018-022-00456-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Guilbaud E, Yamazaki T, Galluzzi L. T cell-independent abscopal responses to radiotherapy. Trends Cancer. 2023;9(2):93–5. 10.1016/j.trecan.2022.12.005 [DOI] [PubMed] [Google Scholar]
  • 80.Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17(10):611–29. 10.1038/s41571-020-0382-2 [DOI] [PubMed] [Google Scholar]
  • 81.Wennerberg E, Spada S, Rudqvist NP, Lhuillier C, Gruber S, Chen Q, et al. CD73 Blockade Promotes Dendritic Cell Infiltration of Irradiated Tumors and Tumor Rejection. Cancer Immunol Res. 2020;8(4):465–78. 10.1158/2326-6066.CIR-19-0449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5(5):429–41 [DOI] [PubMed] [Google Scholar]
  • 83.Price JM, Prabhakaran A, West CML. Predicting tumour radiosensitivity to deliver precision radiotherapy. Nat Rev Clin Oncol. 2023;20(2):83–98. 10.1038/s41571-022-00709-y [DOI] [PubMed] [Google Scholar]
  • 84.Sethumadhavan S, Silva M, Philbrook P, Nguyen T, Hatfield SM, Ohta A, et al. Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells. PLoS One. 2017;12(11):e0187314. 10.1371/journal.pone.0187314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Barsoum IB, Smallwood CA, Siemens DR, Graham CH. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74. 10.1158/0008-5472.CAN-13-0992 [DOI] [PubMed] [Google Scholar]
  • 86.Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212(2):139–48. 10.1084/jem.20140559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Xu J, Escamilla J, Mok S, David J, Priceman S, West B, et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013;73(9):2782–94. 10.1158/0008-5472.CAN-12-3981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Suthen S, Lim CJ, Nguyen PHD, Dutertre CA, Lai HLH, Wasser M, et al. Hypoxia-driven immunosuppression by Treg and type-2 conventional dendritic cells in HCC. Hepatology. 2022;76(5):1329–44. 10.1002/hep.32419 [DOI] [PubMed] [Google Scholar]
  • 89.Scharping NE, Rivadeneira DB, Menk AV, Vignali PDA, Ford BR, Rittenhouse NL, et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat Immunol. 2021;22(2):205–15. 10.1038/s41590-020-00834-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Vignali PDA, DePeaux K, Watson MJ, Ye C, Ford BR, Lontos K, et al. Hypoxia drives CD39-dependent suppressor function in exhausted T cells to limit antitumor immunity. Nat Immunol. 2023;24(2):267–79. 10.1038/s41590-022-01379-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Movahedi K, Laoui D, Gysemans C, Baeten M, Stangé G, Van den Bossche J, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70(14):5728–39. 10.1158/0008-5472.Can-09-4672 [DOI] [PubMed] [Google Scholar]
  • 92.Wenes M, Shang M, Di Matteo M, Goveia J, Martín-Pérez R, Serneels J, et al. Macrophage Metabolism Controls Tumor Blood Vessel Morphogenesis and Metastasis. Cell Metab. 2016;24(5):701–15. 10.1016/j.cmet.2016.09.008 [DOI] [PubMed] [Google Scholar]
  • 93.Weichselbaum RR, Ishwaran H, Yoon T, Nuyten DS, Baker SW, Khodarev N, et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc Natl Acad Sci U S A. 2008;105(47):18490–5. 10.1073/pnas.0809242105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Musella M, Guarracino A, Manduca N, Galassi C, Ruggiero E, Potenza A, et al. Type I IFNs promote cancer cell stemness by triggering the epigenetic regulator KDM1B. Nat Immunol. 2022;23(9):1379–92. 10.1038/s41590-022-01290-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chen W, Teo JMN, Yau SW, Wong MY, Lok CN, Che CM, et al. Chronic type I interferon signaling promotes lipid-peroxidation-driven terminal CD8(+) T cell exhaustion and curtails anti-PD-1 efficacy. Cell Rep. 2022;41(7):111647. 10.1016/j.celrep.2022.111647 [DOI] [PubMed] [Google Scholar]
  • 96.Jacquelot N, Yamazaki T, Roberti MP, Duong CPM, Andrews MC, Verlingue L, et al. Sustained Type I interferon signaling as a mechanism of resistance to PD-1 blockade. Cell Res. 2019;29(10):846–61. 10.1038/s41422-019-0224-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Benci JL, Xu B, Qiu Y, Wu TJ, Dada H, Twyman-Saint Victor C, et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell. 2016;167(6):1540–54 e12. 10.1016/j.cell.2016.11.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ban Y, Markowitz GJ, Zou Y, Ramchandani D, Kraynak J, Sheng J, et al. Radiation-activated secretory proteins of Scgb1a1(+) club cells increase the efficacy of immune checkpoint blockade in lung cancer. Nat Cancer. 2021;2(9):919–31. 10.1038/s43018-021-00245-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Niknam S, Barsoumian HB, Schoenhals JE, Jackson HL, Yanamandra N, Caetano MS, et al. Radiation Followed by OX40 Stimulation Drives Local and Abscopal Antitumor Effects in an Anti-PD1-Resistant Lung Tumor Model. Clin Cancer Res. 2018;24(22):5735–43. 10.1158/1078-0432.Ccr-17-3279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pilones KA, Charpentier M, Garcia-Martinez E, Daviaud C, Kraynak J, Aryankalayil J, et al. Radiotherapy Cooperates with IL15 to Induce Antitumor Immune Responses. Cancer Immunol Res. 2020;8(8):1054–63. 10.1158/2326-6066.CIR-19-0338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Dovedi SJ, Melis MH, Wilkinson RW, Adlard AL, Stratford IJ, Honeychurch J, et al. Systemic delivery of a TLR7 agonist in combination with radiation primes durable antitumor immune responses in mouse models of lymphoma. Blood. 2013;121(2):251–9. 10.1182/blood-2012-05-432393 [DOI] [PubMed] [Google Scholar]
  • 102.Petroni G, Buqué A, Yamazaki T, Bloy N, Liberto MD, Chen-Kiang S, et al. Radiotherapy Delivered before CDK4/6 Inhibitors Mediates Superior Therapeutic Effects in ER(+) Breast Cancer. Clin Cancer Res. 2021;27(7):1855–63. 10.1158/1078-0432.Ccr-20-3871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC. N Engl J Med. 2018;379(24):2342–50. 10.1056/NEJMoa1809697 [DOI] [PubMed] [Google Scholar]
  • 104.Lee NY, Ferris RL, Psyrri A, Haddad RI, Tahara M, Bourhis J, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 2021;22(4):450–62. 10.1016/S1470-2045(20)30737-3 [DOI] [PubMed] [Google Scholar]
  • 105.Lim M, Weller M, Idbaih A, Steinbach J, Finocchiaro G, Raval RR, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 2022;24(11):1935–49. 10.1093/neuonc/noac116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Omuro A, Brandes AA, Carpentier AF, Idbaih A, Reardon DA, Cloughesy T, et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro Oncol. 2023;25(1):123–34. 10.1093/neuonc/noac099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Demaria S, Guha C, Schoenfeld J, Morris Z, Monjazeb A, Sikora A, et al. Radiation dose and fraction in immunotherapy: one-size regimen does not fit all settings, so how does one choose? J Immunother Cancer. 2021;9(4). 10.1136/jitc-2020-002038 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES