Abstract
In addition to inducing lethal DNA damage in tumor and stromal cells, radiation can alter the interactions of tumor cells with their microenvironment. Recent technological advances in planning and delivery of external beam radiotherapy have allowed delivery of larger doses per fraction (hypofractionation) while minimizing dose to normal tissues with higher precision. The effects of radiation on the tumor microenvironment vary with dose and fractionation schedule. In this review, we summarize the effects of conventional and hypofractionated radiation regimens on the immune system and tumor stroma. We discuss how these interactions may provide therapeutic benefit in combination with targeted therapies. Understanding the differential effects of radiation dose and fractionation can have implications for choice of combination therapies.
Keywords: Radiation, tumor microenvironment, stroma, tumor immunity
Introduction
To date, radiobiological models have been primarily focused on improving the therapeutic ratio of radiation treatment by enabling a toxic dose of radiation to tumor cells while sparing normal tissue. The linear quadratic (LQ) model of radiobiology is based on differential DNA repair in normal and tumor tissues. The LQ model predicts single-strand lethal damage (α parameter in LQ equation) and double-strand lethal damage (β parameter in the LQ equation) induced by ionizing radiation. Subsequent sublethal damage repair can also be modeled. The LQ model predicts that delivery of radiation dose in small fractions induces maximum tumor cell death compared with single-dose delivery, with minimal effects on normal tissue.1
Traditionally, delivery of doses of 50 to 70 Gy using multiple fractions of 1.8 to 2 Gy per fraction in the curative setting of solid tumors is preferred to deliver the best therapeutic ratio. Recent technological advances in planning and delivery of external beam radiotherapy such as 3-dimensional image guidance, modulation of beam intensity and multileaf collimators, and 4-dimensional tumor tracking have allowed conformal delivery of larger doses per fraction while minimizing dose to normal tissues with higher precision. Thus, hypofractionated and extreme hypofractionated delivery schedules have been employed in treatment of many tumor types to increase tumor cell death while maintaining the therapeutic ratio. Stereotactic body radiation therapy (SBRT) and stereotactic radiosurgery (SRS) deliver focused, high-dose radiation in 1 to 5 fractions. Both SBRT and SRS have been used effectively for the treatment of lung, liver, brain, prostate, and recurrent head and neck cancers, among others.2–6 Damage to tumor cell DNA is thought to account for only part of the efficacy of hypofractionated regimens.7 Many studies indicate that, in addition to direct impact on DNA, the effects of high-dose radiation on the tumor microenvironment (TME) may play a role in tumor control by SBRT and SRS.7–10
Profound evidence of the effect of radiation on the TME can be demonstrated by the abscopal effect, in which focal radiation can result in out-of-field responses. First reported in 1953, the abscopal effect is thought to occur through radiation-induced activation of immune responses within the TME.11 Although clinical reports of abscopal immune responses induced by radiation alone are relatively rare,12 recent advances in immunotherapy have renewed interest in radiation-induced immune responses. Ionizing radiation can alter interactions between tumor cells and their microenvironment and lead to immune cell priming. Cellular and DNA damage induced by radiation can result in the generation and release of tumor-associated neoantigens,13,14 as well as release of cytokines from both tumor and stromal cells.15–17 These events can shift the balance toward an immunoreactive, as opposed to immunosuppressive microenvironment, leading to the recruitment and activation of cytotoxic T cells. As such, radiation may expand the use of immunotherapeutics into nonimmunogenic tumors. Likewise, clinical reports suggest that radiation may rescue response in patients who have acquired resistance to checkpoint inhibitors.18 Several clinical trials are underway to determine the effects of combination therapy with radiation and immune checkpoint inhibitors.19 However, the optimal dose and fractionation schedules and predictive biomarkers required for radiation-induced immune priming are unclear.
In addition to the effects on the immune system, hypofractionated radiation therapy can also have profound effects on stromal tissue that may either promote or decrease tumor responses. Hypofractionated radiation can have both direct and indirect effects on the tumor vasculature20,21 and may synergize with antiangiogenic therapy.22–24 In addition to its effects on the tumor vasculature, radiation may also induce release of protumor cytokines and growth factors from cancer-associated fibroblasts (CAFs)25 and lead to remodeling of the extracellular matrix (ECM)26,27 and decreased radiation response. This review summarizes radiation-induced immune and stromal effects and highlights differences that may arise from conventional versus hypofractionated radiation schedules.
Radiation Effects on the Immune System
Escape from immune surveillance is increasingly being recognized as a hallmark of cancer. Two aspects of immune escape are the ability to suppress immune reactivity and the ability to evade immune recognition.28 Radiotherapy in combination with immunotherapy may aid in activating an immune response and switching the TME from immunosuppressive to immunoreactive. It was previously thought that radiation had a suppressive effect on the immune system, particularly due to the radiosensitivity of lymphocytes.29–31 Studies now suggest that radiation can stimulate the immune system, particularly through high-dose, hypofractionated administration. Preclinical studies demonstrating radiation effects on the immune TME are listed in Table 1.
Table 1.
Mechanisms | Cancer type | Model(s) | Strain(s) | Dose(s) | References |
---|---|---|---|---|---|
Activation of CD8+ T-cell responses | |||||
Melanoma | OVA-expressing B16/F0 | C57BL/6J | Single dose: 15 Gy | Lugade et al16 | |
Melanoma and breast cancer | 4T1 and B16 | C57BL/6, nude, B6/Rag, OTI, and BALB/c mice | Single dose: 20 Gy | Lee et al17 | |
Breast cancer | TSA | BALC/c | Fractionated dose: 3 × 8 Gy | Devan et al15 | |
Colon cancer | CT26 and MC38 | WT BALB/c (H2d) and C57BI (H2d), BALB/cRAG2−/−, BALB/c Batf3−/− | Single dose: 30 Gy | Filatenkov et al32 | |
Colon cancer | CT26 | BALB/c and C57L/6 | Fractionated dose: 5 × 2 Gy | Dovedi et al33 | |
Melanoma | MC38 | C57BL/6 | Single dose: 10 Gy | Reits et al14 | |
Breast cancer | 4T1 | BALB/c | Single dose: 12 Gy or fractionated dose: 2 × 12 Gy | Demaria et al34 | |
DC maturation/antigen presentation | |||||
Melanoma | OVA-expressing B16/F0 | C57aBL/6J | Single dose: 15 Gy | Lugade et al16 | |
Melanoma and breast cancer | 4T1 and B16 | C57BL/6, nude, B6/Rag, OTI, and BALB/c mice | Single dose: 20 Gy | Lee et al17 | |
Colon cancer, lung cancer, and melanoma | MC38, LLC, and B16gp tumors | C57BL/6 | Single dose: 10 Gy | Gupta et al35 | |
Colon cancer and breast cancer | EL4 CT26 and TS/A tumors | C57BL/6 | Single dose: 10 Gy | Apetoh et al36 | |
Increased IFN-γ expression | |||||
Colon cancer | Colon38 | C57BL/6J and B612957-Ifngtm1Ts (IFN-γ−/−) | Single dose: 15 Gy | Gerber et al37 | |
Melanoma | OVA-expressing B16-F0 tumors | C57BL/6J and B6.12957-Ifntm/Ts (IFN-γ−/−) | Single dose: 20 Gy | Lugade et al38 | |
Increased type 1 IFN responses | |||||
Melanoma | B16-F10 or TSA | WT or B6/IFNAR1−/− | Single dose: 20 Gy | Burnette et al39 | |
Colon cancer | MC38 tumors | C57BL/6J, Myd88−/−, Trif−/−, Camp−/−, 2C CD8+ TCR-Tg, Cd11cCre+-Tg, Ifnar1flox/flox Tmem173−/−, Irf3−/− | Single dose: 20 Gy or fractionated dose: 2 × 15 Gy | Deng et al40 | |
Breast cancer and colon cancer | MCA38 or TSAshN MDA-MB-231 tumors | WT or Ifnar−/− or Batf3−/− BALB/c mice; NOG | Single dose: 20 Gy | Vanpouille-Box et al41 | |
Pancreatic, lung, breast, and squamous cell carcinoma | PancO2, 3LL, MMTV-PYMT, and SCCVII | C57BL/6; C3H; FVB | Single dose: 10 Gy | Baird et al42 | |
MHC-1 expression | |||||
Colon, lung, and prostate cancer | Panel of human colon (12), lung (7), and prostate (4) cancer cell lines | In vitro | Single dose: 10 or 20 Gy | Garnett et al43 | |
Breast cancer | ZR-75-1, T47D, MDA-MB-231, MCF-7 | In vitro | Fractionated dose: 3, 4, or 5 × 2 Gy | Wan et al44 | |
Regulatory T cells | |||||
Prostate cancer | TRAMPC1 | C57BL/6 and Foxp3EGFP | Single dose: 10 Gy | Kachikwu et al45 | |
Melanoma | D5 and D5-G6 tumors | C57BL/6 (B6) and Thy1a/CyJ (CD90.1) m | Fractionated dose: 2 × 7.5 Gy | Wei et al46 | |
Melanoma | B16-OVA tumors | C57BL/6 | Fractionated dose: 5 × 8 Gy | Schaue et al47 | |
MDSC | |||||
Prostate cancer | TRAMP-C1 and TRAMP-HR | C57BL/6J | Single dose: 15 Gy | Wu et al48 | |
Prostate cancer | g RM-1, RM-9, or MycCaP tumors | C57BL6 | Fractionated dose: 5 × 20 Gy | Xu et al49 | |
Breast cancer and pancreatic cancer | Panc02 tumors | BALB/cs or C57BL/6 | Fractionated dose: 5 × 20 Gy | Crittenden et al50 | |
TAM polarity | |||||
Pancreatic cancer | RipTag-5 | Fractionated dose: 2 × 4 Gy | Prakash et al51 | ||
Prostate cancer, glioma, and astrocytoma | TRAMP-C1, ALTS1C1, or GL261 cells | C57BL/6J m | Single dose: 25 Gy, 8 Gy | Chiang et al52 | |
Pancreatic cancer | RT5, TCRtg | RT5, TCRtg, NOD/SCID γ, or C3HeB/Fe | Single dose: 0.5, 1, 2, or 6 Gy | Klug et al53 | |
Squamous cell carcinoma | g OSC-19, HSC-3, or YCU-OR891 | BALB/c or nude | Single dose: 12 Gy | Okubu et al54 | |
Pancreatic cancer | LSL-KrasG12D | C57BL/6 mice, p48CRE; LSL-KrasG12D | Single dose: 2 to 12 Gy or fractionated dose: 3 × 6 Gy | Seifert et al55 | |
Fas expression | |||||
Colon cancer | MC38-CEA+ | C57BL/6 or CDE-Tg | Single dose: 8 Gy | Chakraborty et al56,57 | |
Combination with PD-1/PD-L1 inhibition | |||||
Colon cancer | CT26 | BALB/c or C57L/6 | Fractionated dose: 5 × 2 Gy | Gupta et al35 | |
Colon cancer and breast cancer | TUBO or CD38 | BALB/c or C57L/6 | Single dose: 12 Gy or 20 Gy | Deng et al58 | |
Melanoma, renal cell, and breast cancer | RENCA, 4T1, or B16-OVA | BALB/c or C57L/6 | Single dose: 15 Gy | Park et al59 | |
Melanoma and breast cancer | B16-F10 or TSA | BALB/c or C57L/6 | Single dose: 20 Gy or fractionated dose: 3 × 8 Gy | Twyman-Saint Victor et al60 | |
Colon and breast cancer | CT26 or 4T1 | BALB/c or C57L/6 | Fractionated dose: 5 × 2 Gy | Dovedi et al61 | |
Colon cancer | CT26 | BALB/c or C57L/6 | Fractionated dose: 5 × 2 Gy | Gupta et al35 |
Abbreviations: DC, dendritic cell; OVA, ovalbumin; WT, wild type.
Cytotoxic T cell–driven radiation-induced tumor regression
The optimal radiation dose and fractionation regimens to activate cytotoxic immune responses are unclear. Many studies have suggested that ablative or hypofractionated radiation is superior to conventional fractionation schemes for the activation of antitumor CD8+ T-cell response.15–17 High-dose radiation induces enhanced double-strand breakage leading to increased cell death of tumor cells, as well as release of tumor antigens and cytokines, such as IFN-γ, that promote activation of cytotoxic CD8+ T cells.16,17 In a murine model of ovalbumin (OVA)-expressing B16-F10 melanoma, Lugade et al16 compared the effects of a single dose of 15 Gy radiation to fractionated radiotherapy (5 × 3 Gy). Although T-cell responses were observed in both regimens, the single 15 Gy dose resulted in increased numbers of antigen-presenting cells and CD8+ T cells within tumor-draining lymph nodes, increased frequency of IFN-γ–secreting cells, and increased cell lysis by CD8+ T cells compared with fractionated regimens. In addition, ablative radiotherapy (20-Gy single dose) was reported to induce tumor rejection in an immunocompetent murine model of B16 melanoma but failed to induce regression in immunodeficient nude mice or mice that had been depleted of CD8+ T cells.17 Although an initial response was observed in mice treated with moderate doses of fractionated radiation (4 × 5 Gy), tumors relapsed in a manner similar to T-cell–depleted mice. These findings suggest that fractionated regimens may result in death of infiltrating lymphocytes, resulting in relapse or recurrence. However, differential effects of radiation on tumor cell death between ablative and fractionated radiation could not be ruled out in this study. In contrast, a study in TSA breast carcinoma xenografts showed that hypofractionated (3 × 8 Gy) but not ablative (20-Gy single dose) radiotherapy resulted in an abscopal effect when combined with an anti-cytotoxic T cell–associated protein-4 (CTLA-4) antibody.15 Likewise, evidence of T-cell activation following SBRT has been documented in patients with early-stage non–small-cell lung cancer (NSCLC). Increases in activated T cells were detected in the peripheral blood of patients with NSCLC following a total dose of 48 Gy in 6 to 8 fractions of SBRT.62 In addition, changes in both CD8+ and CD4+ T cells have been documented in NSCLC following SBRT.63 The proportion of overall CD8+ T cells and CD4+ T cells as well as activated CD4+ T cells expressing GATA-3, T-bet, and ROR-γt increased following SBRT in patients with NSCLC, whereas the levels of CD4+ FoxP3+ regulatory T cells (Tregs) decreased.
Most of the studies have suggested that high-dose radiation regimens are required for optimal induction of T-cell immune responses following radiotherapy, but less is known regarding conventional fractionation schedules. Filatenkov et al32 demonstrated that while an ablative single dose (30 Gy) induced a robust CD8+ T-cell–dependent response that lead to tumor regression, addition of fractionated therapy (10 × 3 Gy) following this dose diminished the response. These findings indicate that conventional fractionation schedules may have a detrimental effect on the immune microenvironment that may result from death of infiltrating lymphocytes, resulting in relapse or recurrence. However, a recent study reported local and systemic immune responses that resulted in tumor clearance and abscopal effects in mice bearing dual CT26 colon carcinoma xenografts in which a single tumor site was treated with a conventional fractionation regimen (5 × 2 Gy) in combination with an anti-programmed cell death protein-1 (PD-1) monoclonal antibody.33 Despite a reduction in T-cell number 24 hours after each fraction, there was expansion of local polyclonal T-cell responses and increases in infiltrating T cells in both irradiated and nonirradiated tumor sites 7 days after the conclusion of treatment, suggesting that conventional fractionation may also stimulate cytotoxic T cell responses. Interestingly, there was a high level of concordance between T-cell receptors from irradiated and nonirradiated tumors, whereas only 0.5% of T cells were unique to the irradiated tumor. This indicates that most of the reactive T cells in this model were responding to preexisting antigens rather than neoantigens created by radiation-induced injury. These findings are consistent with a previous study that showed increased intracellular peptide pools and expression of unique class I major histocompatibility molecules (MHC-1) in a dose-dependent manner.14 Therefore, the formation of neoantigens may be optimal at doses higher than those delivered during conventional radiation regimens.
Dendritic cells aid in the activation of an antitumor immune response following radiotherapy
As the major antigen-presenting cell in antitumor immunity, dendritic cells process antigenic material and present tumor-associated antigens (TAAs) to CD8+ T cells, thereby aiding in their activation. Many studies have reported enhanced priming of T-cell responses after treatment with hypofractionated radiation due to increased presentation of TAA to CD8+ T cells in draining lymph nodes.16,17,35,36 In mouse models of thymoma and breast cancer, dendritic cells have been shown to be important in the antitumor T-cell–mediated immune response through radiation-induced release of high-mobility group box 1 (HMGB1) by dying tumor cells, which act on toll-like receptor 4 (TLR-4) expressed by dendritic cells, aiding in processing and cross-presentation of TAA.36 In a B16 melanoma model, increases in antitumor immunity after radiation of tumors with a single dose of 10 Gy was dependent on infiltration of both CD8+ T cells and CD11c+ dendritic cells.35 In this model, therapeutic efficiency was found to depend on the radiation-induced activation status of dendritic cells and not dependent on the presence of antigens. These findings are consistent with another study which also found that single high-dose radiation (20 Gy) resulted in activation and maturation of dendritic cells, improving T-cell priming in a mouse model of B16 melanoma.17
Radiation-induced IFN-γ expression aids in T-cell activation
Radiation can influence cytokine production in the TME and alter the ways in which immune cells traffic to the tumor and function. Radiation promotes necrotic cell death, causing the release of DNA and damage-associated molecular patterns (DAMPs) including HMGB1 and ATP. The DAMPS can activate inflammatory signaling pathways which result in the release of a multitude of pro-inflammatory cytokines such as IFN-γ, TNF-α (tumor necrosis factor α), TGF-β (transforming growth factor β), interleukin (IL)-6, IL-10, IL-1α, IL-1β, and IFN-β that can be either immunosuppressive or immunostimulatory depending on their target cell.36,64,65,38,39 One of the most essential cytokines for radiation-induced T-cell priming is IFN-γ. IFN-γ is produced by cytotoxic T cells on antigen-specific activation, indicating presence of tumor antigen–specific cytotoxic T cells following radiation.65 In a murine model of colon cancer, Gerber et al37 demonstrated the necessity of IFN-γ release in radiation-induced immunity. In this study, increased secretion of IFN-γ was observed by CD8+ T cells after treatment with a single dose of 15 Gy and depletion of CD8+ T cells greatly reduced intratumoral levels of IFN-γ. Increased secretion of IFN-γ by CD8+ T cells was associated to increased lytic capability. Although IFN-γ was shown to have no effect on tumor cell growth, radiation-induced tumor regression was not evident in IFN-γ knockout mice, indicating its essential role in radiation-induced immune responses. Likewise, IFN-γ production within the TME in response to radiation has been shown to influence the antitumor immune response in a murine model of B16 melanoma.38 In this study, a single dose of 15 Gy localized radiation led to increased T-cell trafficking through upregulation of vascular cell adhesion protein 1 (VCAM-1) on tumor vasculature as well as the chemoattractants MIG and IP-10 in wild-type mice but not in IFN-γ–negative mice, demonstrating the importance of IFN-γ in the radiation-induced T-cell–mediated immune response.
Radiation-induced type 1 interferon–dependent responses
Type 1 interferons (IFN-α and IFN-β) are important for activation of both innate and adaptive immune responses and are well known for their role in viral immunity.66 In addition to natural killer and T-cell activation, type 1 interferons can also play a role in cross-priming of dendritic cells during adaptive immune responses.67 Induction of type 1 interferon responses has been shown to be important in tumor reduction in response to radiotherapy.39,40 Local, single dose of 20-Gy radiation to B16 melanoma tumors in mice resulted in increased gene expression of IFN-β.39 IFN-β production led to expansion of antigen-specific T cells and tumor reduction in a T-cell–dependent manner. In addition, the IFN-α/β receptor was necessary for the therapeutic benefit of radiotherapy to these tumors. Radiation-induced activation of type 1 interferon pathways correlated with abscopal responses in combination with anti-CTLA immunotherapy in mice treated with hypofractionated (3 × 8 Gy) regimens in a murine model of immune refractory mammary carcinoma.41 Similar to previous studies, abscopal responses were abrogated in mice in which the interferon receptor had been knocked down in the irradiated tumor. These effects were not observed with single high-dose treatment in this study, as treatment with a single dose of 20 Gy in combination with anti-CTLA immunotherapy failed to induce IFN-β secretion or abscopal responses.
Radiation-induced expression of type 1 interferons can be initiated through recognition of DAMPs by toll-like receptors (TLRs).36,68 However, studies conducted in mice deficient for the TLR adaptor proteins MYD88 and TRIF failed to show any difference in radiation-induced immune responses compared with wild-type animals.40 Alternatively, induction of type 1 interferons can be mediated through the TLR-independent Gas-STING pathway.69,70 Recognition of cytosolic DNA by the cGAMP synthase cGAS leads to the generation of cGAMP which then binds to and activates the endoplasmic reticulum protein STING (stimulator of interferon genes). STING activation results in the nuclear translocation of transcription factors which promote expression of type 1 interferons.69–71 CD11c+ dendritic cells, which are thought to have taken up exogenous tumor cell DNA, have been shown to be the major producers of IFN-β after radiation treatment, although STING-dependent activation of type 1 interferons has also been observed in tumor cells.40,41,72,73 Deng et al40 showed that radiation-induced IFN-β was abrogated in dendritic cells from STING-deficient mice and these mice lacked the ability to cross-prime reactive T cells in response to radiation compared with wild-type controls. Treatment with exogenous IFN-β was able to restore this effect. Knockdown of cGAS in TSA breast cancer xenografts was shown to impair radiation recruitment and activation of CD8α+ tumor-infiltrating dendritic cells and led to a reduction in priming of tumor-specific CD8+ T cells in tumor-draining lymph nodes and spleen.41 These studies indicate that STING-mediated induction of IFN-β may play an important role in radiation-induced immune priming. Furthermore, co-treatment with cGAMP or STING agonists has been shown to amplify the radiation-induced antitumor immune response and result in tumor regression.40,42
The optimal dose and fractionation schedule are unclear for STING-mediated activation of type 1 interferon responses. IFN-β–driven immune responses have been observed at both high and low radiation doses.39–42,72,73 However, Vanpouille-Box et al41 reported that accumulation of cytosolic DNA was attenuated by the expression of the DNA exonuclease Trex1. Trex1 upregulation was observed in a dose-dependent manner after treatment with radiation and correlated with a reduction in cytosolic DNA at single doses above 12 Gy, but not on fractionation with lower doses. In addition, accumulation of cytosolic DNA in tumor cells was associated to time in S-phase.73,74 Higher radiation doses may induce blocks in cell cycle which decrease S-phase–dependent accumulation. However, more work is needed to understand the effects of conventional radiation on type 1 interferon response.
MHC-I upregulation by tumor cells following radiotherapy
Tumor cells present TAAs on MHC-I, which is recognized by cytotoxic T cells specific to that antigen, leading to lysis of tumor cells by cytotoxic T cells. One mechanism through which tumor cells escape immune recognition is through downregulation of MHC-I expression, which decreases their antigen presentation and recognition by lymphocytes. Studies more than 20 years old have shown that radiation can increase MHC-I expression by tumor cells, which may lead to enhanced immune recognition of tumor cells.75–77 In 1994, Klein et al75 showed that irradiation of glioblastoma cells isolated from patients increased MHC-I expression in 4 out of 8 cases and was dose dependent, with the highest increase in MHC-I following 12 Gy. In 1996, Santin et al76 demonstrated that high doses of radiation of 50 to 100 Gy upregulated MHC-I in human ovarian cancer cell lines. In 1997, Santin et al77 showed similar effects in cervical cancer cell lines, with 25, 50, or 100 Gy leading to increased expression of MHC-I. More recently, a panel of 23 human colon, lung, and prostate cancer cell lines were analyzed for their expression of MHC-I following a single dose of 10 or 20 Gy radiation.43 About 35% of the cell lines had upregulation of MHC-I expression following irradiation with 10 and 20 Gy, all of which were colon and lung cancer cell lines, and none of which were prostate cancer cell lines. The 10 to 25 Gy of radiation has also been shown to upregulate MHC-1 expression in a human melanoma cell line, MelJuSo.14 In addition, a radiation dose of 25 Gy increased MHC-I binding to antigenic peptides. Therefore, MHC-I upregulation appears to be a mechanism by which high-dose, hypofractionated radiation induces antitumor immunity and this response differs among types of cancer and between patients of the same cancer type. Although limited studies have been performed with conventional radiation regimens, lower doses of radiation have also been shown to upregulate MHC.44
Radiation effects on Tregs
Although radiation is known to induce antitumor immunity, some dose and fractionation schedules have been shown to increase the proportion of Tregs, which can impair the therapeutic effects of radiation. The Tregs have been shown to increase in mice lacking tumors following whole-body radiation as low as 0.25 to 0.5 Gy, which is representative of that given during conventional radiotherapy.45 In addition, the same study found that a local, single dose of radiation (10 Gy) to a murine model bearing melanoma resulted in increased Tregs in both the spleen and in the tumors. Elimination of Tregs along with radiotherapy improved tumor growth delay and regression. Local irradiation with 10, 20, or 30 Gy in human patients with cervical cancer has been shown to decrease CD8+ and CD4+ T cells but have no effect on FoxP3+ Tregs and therefore shifts the balance of lymphocytes by enriching for an immunosuppressive, regulatory subtype.78 These increases in Tregs following radiation is thought to result from their increased radioresistance compared with other T-cell subtypes.
In contrast, hypofractionated radiation treatment (5 × 8 Gy) in a murine model of melanoma found that radiation reduced the number and impaired the functionality of FoxP3+CD4+ Tregs, both in the blood and within the TME.46 The reduction was equal to that of conventional T cells 0 to 2 days following radiation but greater than conventional T cells 4 to 7 days after radiation, indicating that Tregs are slower to recover. This study also found that local tumor radiation increased both the proliferation and the functionality of both host T cells and adoptively transferred T cells, and tumor radiation plus adoptive T-cell transfer significantly inhibited tumor cell growth.
The differences in Treg enrichment or depletion are most likely dependent on the radiation dose and fractionation schedule given. In a murine model of B16 murine melanoma expressing the antigen OVA, local, single dose of 5 Gy increased the Treg population but did not increase the effector T-cell response.47 Alternatively, a single dose of 7.5 or 10 Gy resulted in a decrease in Tregs in the spleen and an increase in anti-OVA T cells, whereas a single dose of 15 Gy of radiation increased both the effector T-cell response and the Treg population. In this study, the best tumor control was seen following irradiation with 2 fractions of 7.5 Gy and the Treg population was lowest with this dose and fractionation schedule.
Radiation effects on myeloid derived dendritic cells
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells that are defined by their functional ability to induce immunosuppression.79,80 In mice, 2 distinct subsets of MDSCs have been described: monocytic MDSCs (m-MDSC), which bear the phenotype CD11b+, Ly6ChighLy6G−, and granulocytic MDSCs (g-MDSC), which bear the phenotype CD11b+, Ly6ChighLy6G+.81,82 In humans, a single subset of MDSC has been identified, characterized by the phenotype CD11b+CD14−CD33+.65 The MDSCs accumulate within the TME in response to the pro-inflammatory microenvironment and suppress the activation of both CD4+ and CD8+ T-cell responses.80,83,84 Influx of MDSC into the TME has been reported after both single high-dose and conventional fractionation regimens and are often associated with an influx of tumor-associated macrophages (TAMs).16,48,52,85 In a murine model of prostate cancer, Wu et al48 showed that a single high dose of radiation (15 Gy) resulted in recruitment of MDSCs mediated by radiation-induced release of IL-6 from tumor cells. Likewise, Xu et al49 reported increases in both m-MDSC and g-MDSC in tumors after radiation (5 × 3 Gy) treatment in 2 prostate xenograft models, RM-1 and Myc-Cap. Tumor influx was shown to be dependent on expression of colony-stimulating factor 1 (CSF-1) by the tumor cells on radiation treatment and blockade of CSF-1 was found to retard tumor regrowth after radiation. Although increases in peripheral blood MDSCs were observed after radiation treatment in the model, there were no increases in MDSCs in secondary lymphoid organs. Similarly, Crittenden et al demonstrated in a murine model of breast cancer that hypofractionated radiation treatment (3 × 20 Gy) leads to reduction in peripheral MDSC in the blood and spleen. This decline in MDSCs was linked to radiation response of the tumor, with declining numbers of MDSCs reported on decreased tumor burden.50
Radiation-induced Fas expression by tumor cells
Although CD8+ cytotoxic T cells appear to be involved in high-dose, hypofractionated radiation-induced tumor regression, the mechanism by which this occurs is complex and not fully understood. CD8+ cytotoxic T cells mediate their lytic ability through release of granzyme and perforin and through engagement of Fas ligand (FasL) on their surface with Fas on target cells. Engagement of Fas with FasL leads to apoptosis through a caspase 1–mediated mechanism.86 High-, single dose radiation has been shown to upregulate Fas expression by tumor cells, leading to increased cytotoxic T cell lysis through a Fas/FasL-dependent mechanism.87 Murine MC38 colon adenocarcinoma cells expressing carcinoembryonic antigen (MC38-CEA+) showed increased expression of Fas following different doses of irradiation in vitro, which was highest following a single dose of 20 Gy, and this corresponded with increased lytic ability of CEA-specific cytotoxic T cells. The increased lytic ability of CEA-specific cytotoxic T cells was blocked with addition of anti-FasL, demonstrating the importance of the Fas/FasL interaction. In the same study, B6 mice injected with MC38-CEA+ tumor cells and irradiated with 8 Gy showed increased Fas expression. Adoptive transfer of CEA-specific cytotoxic T cells decreased tumor growth rate and volume. The same group subsequently demonstrated that, using the MC38-CEA+ murine model, vaccination with recombinant viruses expressing CEA and co-stimulation of T cells followed by a single dose of 8 Gy led to upregulation of Fas and significant reduction of tumors through a Fas/FasL-mediated pathway.57 These studies indicate that Fas upregulation following high-dose, hypofractionated radiation is a mechanism through which cytotoxic T cells facilitate their antitumor activity.
Radiation effects on macrophage recruitment and polarization
Many studies in a variety of tumor models have shown that radiotherapy induces recruitment of macrophages into the tumor site, irrespective of dose and fractionation regimen.87 Radiation-induced recruitment of TAMs was shown to be dependent on increased expression of the chemokine CSF-1.49 Gene transcription of CSF-1 was demonstrated after radiotherapy, and blocking of the CSF-1 receptor in a xenograft model of prostate cancer treated with 5 fractions of 3 Gy reduced radiation-induced macrophage recruitment.49 Alternatively, hypofractionated or high-dose single-fraction radiotherapy may promote TAM recruitment through induction of hypoxia by disruption of the tumor vasculature.85,88 Ceradini et al88 showed that HIF-1–induced expression of stromal-derived factor 1 (SDF-1), which promoted recruitment of TAMs in a xenograft model of glioma following a single dose of 15 Gy.
The impact of increased presence of TAMs after radiation is unclear and is dependent on their polarization into either M1 or M2 subtypes. Although M1 macrophages promote inflammation and antitumor immune responses, those of the M2 phenotype are thought to support tumor growth, angiogenesis, and metastasis.89 The impact of radiation on macrophage polarization may be related to dose and fractionation. Conventional doses of radiation have been shown to promote M1 polarization. Treatment of pancreatic tumor xenografts with radiation given as 4 Gy in 2 fractions resulted in a switch in tumor-infiltrating macrophages from a protumorigenic M2 phenotype to an antitumorigenic M1 phenotype.51 Likewise, increased infiltration of T cells into tumors and tumor killing mediated by iNOS+ M1 macrophages through expression of TH1 cytokines have been reported in murine models of pancreatic cancer and melanoma after low-dose radiation treatment.51,53 Interestingly, this effect was not observed on high-dose radiation in these studies. Moreover, many studies have demonstrated M2 polarization after treatment with single high-dose and hypofractionated radiation regimens.52,54,55 M2 polarization often occurs in hypoxic areas90 and thus may be promoted by the vascular damage and hypoxia associated with high-dose radiation.
Effects of Radiation on the Tumor Stroma
In addition to its effect on the immune microenvironment, many studies have shown that radiation can alter the tumor stroma with varying effects observed with different dose regimens. Preclinical studies demonstrating changes in the tumor stroma induced by radiation are listed in Table 2.
Table 2.
Mechansims | Cancer type | Model | Strain | Dose | References |
---|---|---|---|---|---|
Vasculature | |||||
Decreased tumor vascularity, blood flow, and/or vessel function | Breast carcinoma | Walker 256 | Sprague-Dawley rats | Single dose: 20 Gy | Song et al91 |
Breast carcinoma | Walker 257 | Sprague-Dawley rats | Single dose: 5-60 Gy | Song and Levitt92 | |
Breast carcinoma | Walker 258 | Sprague-Dawley | Single dose: 2.5-20 Gy | Wong et al93 | |
Brain metastases | Computer simulation | Single dose: 20 Gy | Kocher et al94 | ||
Pancreas | BxPC3 | NRG mice | Single dose: 4-24 Gy | Maeda et al95 | |
Melanoma | Patient-derived xenograft | Athymic nude mice | Single dose: 10-20 Gy | Solesvik et al96 | |
Ovarian carcinoma | MA148 | Athymic nude mice | Fractionated dose: 4 × 5 Gy | Dings et al97 | |
Rhabdomyosarcoma | BA-1112 | WAG/Rij | Single dose: 60.5 Gy | Emami et al98 | |
Prostate | TRAMP-C1 | C57BL/6J | Single dose: 25 Gy; fractionated dose: 15 × 4 Gy | Chen et al99 | |
Lung carcinoma | Lewis | C57BL/6 | Single dose: 20 Gy; fractionated dose: 2 × 20 Gy | Kim et al100 | |
Squamous cell carcinoma | SCCVII | C3H | Single dose: 15 Gy | Kobayashi et al101 | |
Increased tumor vascularity, blood flow, and/or improved vessel function | Breast carcinoma | Walker 256 | Sprague-Dawley rats | Single dose: <5 Gy | Wong et al93 |
Oral squamous cell carcinoma | SAS-R | BALB/c nude | Fractionated dose: 30 × 2 Gy | Kuwahara et al102 | |
Mammary adenocarcinoma | R3230 AC | Fischer-344 rats | Single dose: 5 Gy | Dewhirst et al103 | |
Hepatocellular carcinoma | TLT | RJ NMRI | Single dose: 6 Gy | Sonveaux et al104 | |
Fibrosarcoma and hepatocellular carcinoma | TLT and FSaII | RJ NMRI and C3H | Fractionated dose: 2 Gy; 2 × 9 Gy | Crokart et al105 | |
Induction of vasculogenesis, angiogenesis, and pro-angiogenic factors | Normal | C57BL/6 | Single dose: <5 Gy | Heissig et al106 | |
Acute lymphoblastic leukemia | MOLT-4 | NOD-SCID | Single dose: 0.3 Gy | Sofia Vala et al107 | |
Normal | Human umbilical vein endothelial cells (HUVECs) | Single dose: 5 Gy | Lerman et al108 | ||
Normal | FVB | Single dose: 5 Gy | Thanik et al109 | ||
Prostate | TRAMP-CI | C57BL/6J | Fractionated dose: 15 × 4 Gy | Chen et al110 | |
Endothelial cell survival | Normal | Primary human dermal microvascular endothelial cells | Primary human dermal microvascular endothelial cells | Single dose: 2 Gy | Wagner-Ecker et al111 |
Normal | Human lung microvascular endothelial cells | Human lung microvascular endothelial cells | Single dose: <1 Gy | Sofia Vala et al107 | |
Induction of endothelial cell apoptosis | Normal | Human lung microvascular endothelial cells | Human lung microvascular endothelial cells | Single dose: 1 Gy -20 Gy | Sofia Vala et al107 |
Melanoma and fibrosarcoma | B16F1 and MCA129 | asmase−/− or Bax−/− mice | Single dose: 10-25 Gy | Gracia-Barros et al112 | |
Cancer-associated fibroblasts | |||||
Radioresistance and induction of senescence | Breast | Primary human normal breast fibroblasts, tumor counterpart fibroblasts, and breast cancer-associated fibroblasts | Single dose: 30 Gy | Hawsawi et al113 | |
Normal | Human HCA2 foreskin fibroblasts | Single dose: >10 Gy | Rodier et al114 | ||
Lung | Primary lung cancer–associated fibroblasts from non–small-cell lung carcinoma tumors | Single dose: 18 Gy | Hellevik et al115 | ||
Protumorigenic | Lung | IR-induced senescent lung fibroblasts, A549, or H1299 | SCID mice | Fractionated dose: 12 × 4 Gy | Papadopoulou and Kletsas116 |
Pancreas | MRC5 fibroblasts, Suit-2 | Nude mice | Single dose: 10 Gy | Ohuchida et al117 | |
Extracellular matrix | |||||
Decreased IR-induced matrix stiffening | Normal mammary stroma | MRC5 fibroblasts | 3D collagen matrix | Fractionated dose: 3 × 1.8 Gy; 3 × 3.6 Gy | Qayyum and Insana118 |
Increased matrix stiffening | Postlumpectomy mammary stroma | MCF-7 | 3D collagen matrix | Fractionated dose: 5 × 1.8 Gy; 5 × 3.6 Gy | Qayyum et al119 |
Formation of reactive stroma | Normal mammary stroma | MRC fibroblasts | 3D collagen matrix | Fractionated dose: 3 × 0.9 Gy | Qayyum and Insana118 |
Postlumpectomy mammary stroma | MCF-7 | 3D collagen matrix | Fractionated dose: 5 × 0.9 Gy | Qayyum et al119 |
Effects of radiation on the tumor vasculature
The most well-studied effect of radiotherapy on the TME is on the tumor microvasculature. Tumor blood vessels do not resemble normal blood vessels in that they are morphologically immature, irregular in diameter, and are more permeable. Mature blood vessels are more radioresistant than developing human blood vessels. Therefore, immature tumor blood vessels and thus tumor endothelial cells are more sensitive to radiation.120 The radiation-induced effects on tumor blood vessels vary widely, especially within animal models, and are dependent on total radiation dose and fractionation schedule as well as tumor type, site, and stage.9,121 However, vascular damage is primarily observed at doses greater than 5 to 10 Gy. In several studies looking at varying radiation doses on vasculature in Walker 256 tumors grown subcutaneously in the hind legs of rats, irradiation with 5 to 20 Gy decreased vascular volume in a dose- and time-dependent manner; however, a single dose of 30 or 60 Gy caused lasting decreases in vascularity and functional vessels in the tumors.91–93 In general, exposure to a single low dose of radiation resulted in tumor blood flow initially increasing and returning to pre-irradiation levels quickly, whereas high doses (>10 Gy) of radiation in a single exposure reduced tumor blood flow dramatically and resulted in destruction of tumor vasculature, changing tumor oxygen levels, and causing indirect cell death resulting in tumor volume decrease. These findings are supported by bioinformatics studies which indicate that high-dose radiation may have a profound effect on the tumor vasculature.94 Using computer modeling of clinical data, changes in tumor vasculature in response to radiation were found to contribute to 19% to 33% of the overall effect from a single high-dose (20 Gy) radiosurgery in brain metastases.
Low-dose radiation schedules are associated with stimulation of angiogenesis and neovascularization, whereas endothelial cell survival is reported at conventional doses of 2 Gy.102,111 Furthermore, irradiation of mammary adenocarcinoma xenografts with doses of 5 Gy resulted in an increase in vascular density and perfusion.103 In addition, in vitro endothelial cell studies showed that low-dose (<5 Gy) radiation stimulated angiogenesis through increased vascular endothelial growth factor (VEGF) secretion by stromal cells106,107 as well as vasculogenesis.108,109 In a study by Lerman et al,108 low-dose radiation was also found to induce endothelial cell migration and vasculogenesis in an SDF-1–dependent manner. In addition, low-dose radiation has been shown to promote tumor vessel survival and maturation in an SDF-1–dependent manner during fractionated radiotherapy (15 × 4 Gy) in a murine prostate model.110 The response of tumors to radiation is related to the supply of oxygen through adequate blood perfusion of the tumor, which may be improved at low radiation doses. Doses below 10 Gy have been found to promote tumor vessel relaxation and improve oxygenation.104,105 Fractionated regimens were found to have the maximum effect on tumor growth delay, presumably due to reoxygenation of the tumor.105
In contrast, radiation doses higher than 10 Gy per fraction are associated with severe vascular damage leading to deterioration of the TME.8,95 A number of studies have focused on radiation-induced vascular changes of human tumor xenografts in rodents using high-dose irradiation in single-dose or several fractions. Irradiation of human melanoma xenografts with 10 to 15 Gy in a single exposure caused almost half of the tumor vessels to become nonfunctional within a week of treatment.96 In addition, irradiation of human ovarian carcinoma xenografts with 5 Gy /fraction for a total dose of 20 Gy resulted in a decrease in vascular density to about half of that observed in control xenografts.97 Likewise, irradiation of rhabdomyosarcomas grown in the scalp of rats with increasing single doses of radiation decreased blood flow rapidly, by about 50%, but lower doses used in the study had restoration of blood flow within 24 hours.98 Irradiation of mouse prostate tumors grown in the thigh of mice with 25-Gy single dose decreased vascularity by 25%.99 Single-dose radiation treatment (20 Gy) of Lewis lung tumors of mice grown subcutaneously in hind limbs decreased tumor blood flow initially but recovered by 4 days after irradiation. Interestingly, recovery could be suppressed by irradiating again with 20 Gy at 2 days but not 4 days after initial irradiation.100
High-dose radiation has been shown to induce endothelial cell apoptosis through both direct DNA damage and ceramide signaling.9,112,122 Although endothelial cell damage has been shown to be a major factor in the biological mechanism of SBRT and SRS, this phenomenon is sometimes transient and may lead to neovasculogenesis via HIF-1 induction95 and recruitment of bone marrow–derived CD11b+ cells which may serve as a source of endothelial cell replenishment.88,123 Tumors treated with fractionated radiation at 24-hour fractions had less vascular damage than tumors treated with a single high dose of radiation, indicating repair of damaged endothelial cells and thus restoration of tumor blood vessels.9,99,101 Interestingly, treatment with anti-VEGF or anti-VEGFR2 right before high-dose radiotherapy increased ceramide levels and resulted in a higher fraction of apoptotic endothelial cells.124 In addition, restoration of blood flow after fractionated radiation could be suppressed through preventing influx of bone marrow–derived cells through inhibition of SDF-1 signaling.85
Although SBRT and SRS may disrupt tumor vasculature and developing blood vessels, leading to enhanced cell death, they may also generate increased regions of hypoxia.85 Modeling of tumor control probability has indicated that single high-dose regimens may result in impaired local control of hypoxic tumors, compared with hypofractionated regimens.125 It is still under investigation if the loss of reoxygenation with ablative radiation can be overcome by hypofractionated radiation regimens and may require combination therapy with hypoxic cell radiosensitizers to obtain tumor control with lower dose and toxicity.126–128
Effects of radiation on CAFs and the ECM
In the stroma of human carcinomas, CAFs are the most abundant cell types and play a significant role in tumor cell growth, angiogenesis, and invasiveness.129–132 Large numbers of CAFs present in tumors are associated with high-grade malignancy and poor prognosis.132 Within the TME, CAFs control the release of paracrine signals that regulate the behavior of other cells within the tumor and can stimulate the recruitment of cells involved in angiogenesis and inflammatory processes.133,134 Through the secretion of SDF-1 into tumor vasculature, CAFs can recruit endothelial progenitor cells to promote tumor blood vessel formation and contribute to tumor recurrence.134,135 In addition, reactive CAFs are an important source of cytokines and immunomodulators, which influence the inflammatory process during tumor development.131,136 The CAFs are also responsible for deposition of key ECM proteins (eg, collagen, fibronectin, and laminin) as well as secreting ECM-degrading enzymes (eg, matrix metalloproteinases),129,130 which promotes migration of CAFs and degradation of the ECM, allowing the invasion of tumor cells.137 Primary lung tumor CAFs have been found in brain metastases derived from lung carcinomas.138
Both normal tissue fibroblasts and CAFs have been found to be radioresistant cells, being able to survive high doses of radiation totaling above 50 Gy.113,116,139 However, the studies investigating the effects of radiation on CAFs are limited. Most of the studies investigating fibroblast response to radiation were conducted using normal connective tissue fibroblasts. In vitro studies have shown that fibroblasts develop an irreversible senescent phenotype when exposed to a dose >10 Gy of radiation,114 whereas low doses of radiation induce reversible DNA damage without growth arrest.115 Senescent fibroblasts release proteolytic enzymes, cytokines, growth factors, and reactive oxygen species, creating a protumorigenic environment.114,116,117,140,141 In a xenograft rodent model, co-transplantation of senescent fibroblasts with cancer cells increased the stimulatory effects of fibroblasts and promoted tumor growth.116,141 Interestingly, the senescence response is more pronounced after a single high dose of radiation in lung CAFs compared with when the equivalent dose is fractionated into smaller doses, indicating that the size and number of fractions in radiotherapy play an important role in the response of fibroblasts to radiation.115 However, the overall effect of senescent fibroblasts on tumor progression is debated. The cancer-promoting effects of senescent fibroblasts may be organ or tumor specific and better models need to be developed to validate the response of fibroblasts to radiation.
Radiation injury triggers an inflammatory response within the TME, recruiting stromal fibroblasts.142,143 Enhanced release of inflammatory components, especially TGF-β, causes differentiation of the fibroblasts, resulting in the release of inflammatory mediators, ECM proteins, and a decrease in ECM-degrading enzymes secreted from the CAFs.144–147 Over time, excessive accumulation of ECM proteins within the TME thickens and stiffens the tissue, leading to the development of fibrosis.146 This process creates a reactive stroma that promotes tumor growth.148 The extent of radiation-induced fibrosis depends on the dose of radiation and the amount of tissue volume exposed. Qayyum and Insana used a 3-dimensional model of the mammary stroma to investigate fractionated radiotherapy on collagen matrix stiffness and fibroblast activation. Increases in the dose per fraction reduced the formation of reactive stroma necessary for tumor progression and mitigated matrix stiffening induced by radiation alone.118 A lower dose per fraction caused greater activation of fibroblasts when compared with the higher doses. In an in vitro model mimicking postlumpectomy mammary stroma with residual disease, higher doses of radiation per fraction were more effective in reducing the protumorigenic microenvironment through prevention of fibroblast differentiation. However, higher doses per fraction increased ECM stiffening, which could stimulate the growth of surviving cancer cells over time.119
The therapeutic effect of high-dose radiotherapy in certain cancers may be due to a reduction in the tumor promoting properties of CAFs. Hellevik et al149 investigated ablative doses of radiation on CAFs isolated from freshly resected lung tumors from patients with NSCLC. Exposure to a single high dose of radiation (18 Gy) caused a change in the secretory profile of signaling molecules involved in inflammation, angiogenesis, and tumor growth by the lung CAFs. SDF-1, angiopoietin-1, and thrombospondin-2 (TSP-2) were all downregulated, whereas the release of basic fibroblast growth factor (bFGF) was upregulated. Conditioned media from irradiated CAFs did not affect the proliferation or migratory capacity of lung tumor cells in vitro but did partially reduce migratory capacity of endothelial cells, suggesting radiation of CAFs reduces their pro-angiogenic properties. In addition, high single dose of radiation resulted in increased expression of integrins and altered secretion of matrix metalloproteinases resulting in inhibition of the proliferative, migratory, and invasive capacity of lung CAFs.115 However, both single-dose and fractionated radiotherapy elicited a radioprotective response by pancreatic stellate cells on pancreatic cancer cells both in vitro and in vivo.150
Combining the Effects of Radiation on the TME With Targeted Therapies
Immune checkpoint inhibitors
Although radiotherapy has long been known to activate tumor immunity, clinical applications of radiation in combination with immunotherapy have only recently shown promise. The success of immune checkpoint inhibitors directed against the CTLA-4, and the PD-1 pathway has revived clinical interest in using radiation to prime the immune system, particularly in tumors that are not immunogenic or have previously failed immunotherapy. Preclinical evidence of synergy between checkpoint inhibitors and radiation has been demonstrated in numerous animal models, although the optimal timing, dose, and fractionation regimen are not clear. Most of these studies were performed with single ablative doses or hypofractionated regimens.15,34,58–60 CTLA-4 blockade with a monoclonal antibody plus 1 or 2 fractions of 12 Gy leads to improved survival of mice bearing 4TI breast cancer xenografts.34 This improved survival corresponded with inhibition of lung metastases that was dependent on CD8+ but not CD4+ T cells. In addition, the combination of hypofractionated radiotherapy with CTLA-4 blockade also led to an abscopal effect in a TSA murine model of breast cancer.15 In this study, 3 fractions of 8 Gy, but not a single dose of 20 Gy, to the primary tumor site, plus addition of an anti-CTLA-4 monoclonal antibody, led to an improved tumor response at the primary site and a secondary, nonirradiated site. This corresponded to an increase in IFN-γ–producing CD8+ T cells, indicating effector function.
PD-1 and PD-L1 inhibition has also been reported to synergize with radiotherapy. Upregulation of PD-L1 in the TME was observed by Deng et al58 following a high dose of 12-20 Gy radiation. Addition of an anti-PD-L1 monoclonal antibody improved radiation-induced antitumor response in a CD8+ T-cell manner and reduced numbers of MDSCs. In addition, Park et al59 demonstrated that SBRT in combination with anti-PD-1 therapy led to regression of both primary irradiated tumors as well as an abscopal effect.
Although these studies are encouraging, a clinical study of combination therapy with anti-CTLA-4 and radiation treatment in melanoma patients suggested many patients were nonresponders or developed resistance.60 In an animal study designed to recapitulate the clinical trial, the investigators analyzed response to combination treatment with anti-CTLA-4 and radiation in a B16-F10 melanoma model. Although they demonstrated that the combination therapy was more effective than either treatment alone, and responses were observed in both irradiated and nonirradiated tumors, this was not the case in all mice. Mice which failed to respond had evidence of T-cell exhaustion. Transcriptomic profiling revealed upregulation of PD-L1 in the TME of nonresponders and addition of anti-PD-1 to the treatment regimen improved responses in both the naïve B16-F10 tumors and the resistant sublines. In addition, SBRT, anti-CTLA-4, and anti-PD-1 were found to have differential effects on tumor immunity. Although radiation was found to expand the diversity of peripheral T-cell clones, anti-CTLA-4 inhibited Tregs, shifting the balance toward cytotoxic T cells. Addition of anti-PD-1 to the combination therapy prevented T-cell exhaustion and further enhanced T-cell expansion.60
Numerous clinical trials are currently underway to assess the combination of checkpoint inhibition and radiotherapy (reviewed by Crittenden et al151). Although the results are still forthcoming, preliminary data suggest that there may be clinical benefit to combination therapy. A phase 1 trial combining ipilimumab (anti-CTLA-4) in radiation treatment of NSCLC, colorectal carcinoma, sarcoma, and renal cell carcinoma demonstrated a partial response in 10% of patients and clinical benefit in 23% of patients.152 Response correlated with increased peripheral CD8+ T cells, CD8+/CD4+ T-cell ratio, and activation of T cells as defined by expression of 4-1BB and PD-1 on CD8+ T cells. Although results of clinical trials combining PD-1 inhibition and radiation are still forthcoming, case studies demonstrated that the combination may yield therapeutic advantage. A case study of a patient with metastatic melanoma treated with whole brain radiation given as 30 Gy in 10 fractions in combination with PD-1 blockade with pembrolizumab showed complete, durable resolution of disease, including brain metastasis.153
The optimal sequence for delivery of radiation and immunotherapy is still unclear. Murine studies indicate that optimal expression of PD-L1 occurs within 3 days after radiation treatment, and concomitant, but not sequential, treatment is required for T-cell–mediated tumor regression.35,61 In a study of 75 patients with melanoma brain metastases treated with checkpoint therapy in combination with SRS, the administration of either anti-CTLA-4 (ipilimumab) or anti-PD-L1/PD-1 (pembrolizumab or nivolumab) within 4 weeks of SRS resulted in increased early radiographic response, compared with treatment separated by more than 4 weeks.154 However, there was no significant difference observed between concurrent and nonconcurrent therapy, indicating that radiation-induced changes in immune responses are sustained for a short window following treatment. In contrast to this study, secondary analysis of the KEYNOTE-001 phase 1 trial revealed that progression-free survival was significantly longer in patients who had received any form of radiotherapy prior to starting the trial.155 The median time between radiotherapy and the start of pembrolizumab was 9.5 months for patients who received extracranial radiotherapy and 11.5 months for patients who received thoracic radiotherapy. These findings indicate that, for some patients, the immune priming effects of radiation may be long-lasting, and there may still be increased clinical benefit for checkpoint inhibitors in patients who have recurrent disease following radiation therapy. Although studies investigating the effects of checkpoint inhibition prior to radiotherapy are limited, palliative radiation was shown to induce a robust immune response in a patient with refractory disease previously treated with the PD-1 inhibitor nivolumab, indicating that radiation priming may be able to prolong the effects of and/or overcome prior resistance to PD-1 inhibition.18 The optimal sequence, dose, and fractionation schedule will most likely vary depending on type of immunotherapy combinations, as well as tumor features such as immunogenicity and genetic status.
Antiangiogenic agents
The release of angiogenic growth factors such as VEGF and FGF after radiation has been well-documented, and blockade of VEGF signaling has been shown to potentiate radiation effects in animal models.106,107,124 Several angiogenesis inhibitors are under clinical investigation in combination with radiotherapy.156,157 Combination treatment with bevacizumab, a humanized antibody to VEGF-A, has been shown to decrease regions of hypoxia within the TME and improve response to chemoradiation in many types of cancer including rectal, head and neck, colon cancer, and glioma.158–162 In a study comparing the effects of low-dose bevacizumab in meningiomas, which had received prior radiation to those that had not, Furuse et al reported significant radiological responses in 4 of 6 tumors that had been previously irradiated. No response was observed in tumors that had not received radiation, leading the investigators to conclude that the effects of bevacizumab were most likely due to postradiation changes in the TME rather than an effect on tumor cells.22,163 Likewise, low dose bevacizumab has been reported to decrease radiation necrosis164,165; however, this effect was less evident after hypofractionated radiotherapy in tumors with large target volumes.166
Sunitinib is a small molecule tyrosine kinase inhibitor that acts to inhibit downstream signaling pathways of VEGF and has been shown to synergize with both conventional and hypofractionated radiation in animal models of cancer.167 Sunitinib has been shown to increase tumor cell apoptosis and improve oxygenation through vasculature normalization in animal models of cancer.168,169 Several clinical trials are currently in progress that will assess the safety and efficacy of sunitinib as a combination therapy (reviewed in Kleibeuker et al170). Although the optimal dosing and timing of sunitinib combined with radiation have not yet been determined, data from initial trials are encouraging. In a phase 2 trial of concurrent sunitinib and Intensity Modulated Radiotherapy (IMRT) in patients with oligometastases, a local control rate of 75% and a distal control rate of 52% were observed. Mean time to progression was 9.5 months, and 11 patients remained disease free at the 2-year follow-up.171 Promising results were also observed with SRS and SBRT in patients with metastatic renal cell carcinoma who received concurrent sunitinib.172,173 Theoretically, sunitinib may also have an effect on postirradiation changes and has been shown to prevent neovascularization in animal models.157,174 However, to date, use of sunitinib as a maintenance therapy after irradiation has not shown clinical benefit.173,175,176
Conclusions
Understanding the differential effects of conventional fractionation versus SBRT and SRS on the TME may help to inform choice and improve response to combination therapies. Likewise, improved knowledge of the effects of radiation dose and fractionation on the TME may help to optimize treatment planning of radiation delivery. Although hypofractionation may promote enhanced CD8+ T-cell responses, efficacy may be diminished by death of infiltrating lymphocytes (particularly in irradiated lymph nodes) and other microenvironmental changes such as increased tumor hypoxia and polarization of M2 macrophages. Dose modulation with heterogeneous high- and low-dose regions within the tumor may provide a hybrid form of radiation delivery that may promote the antitumor effects of both high-dose and conventional fractionation regimens.
In light of the increasing knowledge of the effects of radiation on the TME, new radiobiological models are needed to allow the radiation oncologist to optimize the treatment plan based on the TME characteristics. Hypofractionated treatments with dose per fraction more than 10 Gy is becoming more popular and accepted in radiation oncology due to advances in treatment delivery techniques; hence, new radiobiological models are being developed. The conventional fractionation models using the LQ equation and the 4Rs of radiobiology (Repair of sublethal damage [DNA], Redistribution in cell cycle, Repopulation, and Reoxygenation) were initially focused only on differential radiation effects between normal and tumor cells, with a major focus on altered rates of DNA repair. In 1989, Steel proposed the addition of a fifth R, Radiosensitivity, to account for the differences in response related to genetic heterogeneity between tumors of the same type.176 Recently, increasing knowledge of radiation effects on the tumor-associated stroma and immune system has led to the proposal of 2 additional Rs to model these interactions, Remodeling of the TME177 and Rejection by the immune system.178 Further incorporation of these additional Rs would bring radiation oncology practice to a whole new level of combinatorial targeted and radiotherapies. Integration of these principles into treatment planning systems could allow for precision treatment planning for true personalized medicine.
Footnotes
Funding:The author(s) received no financial support for the research, authorship, and/or publication of this article.
Declaration of Conflicting Interests:The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Author Contributions: KMA, NJF, and JSM wrote the first draft of the manuscript. KMA, NJF, AR, LR, YY, APD, FM, and JSM contributed to the writing of the manuscript; agree with manuscript results and conclusions; jointly developed the structure and arguments for the paper; and made critical revisions and approved final version. All authors reviewed and approved the final manuscript.
Disclosures and Ethics: As a requirement of publication, authors have provided to the publisher signed confirmation of compliance with legal and ethical obligations including but not limited to the following: authorship and contributorship, conflicts of interest, privacy and confidentiality, and (where applicable) protection of human and animal research subjects. The authors have read and confirmed their agreement with the ICMJE authorship and conflict of interest criteria. The authors have also confirmed that this article is unique and not under consideration or published in any other publication, and that they have permission from rights holders to reproduce any copyrighted material. The external blind peer reviewers report no conflicts of interest.
References
- 1. Cell survival curves. In: Hall EJ, Giaccia AJ, eds. Radiobiology for the Radiologist. 7th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2012:35–23. [Google Scholar]
- 2. Mahajan A, Ahmed S, McAleer MF, et al. Post-operative stereotactic radiosurgery versus observation for completely resected brain metastases: a single-centre, randomised, controlled, phase 3 trial. Lancet Oncol. 2017;18:1040–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Aridgides P, Bogart J. Stereotactic body radiation therapy for stage I non-small cell lung cancer. Thorac Surg Clin. 2016;26:261–269. [DOI] [PubMed] [Google Scholar]
- 4. Qiu H, Moravan MJ, Milano MT, Usuki KY, Katz AW. SBRT for hepatocellular carcinoma: 8-year experience from a regional transplant center [published online ahead of print July 14, 2017]. J Gastrointest Cancer. doi: 10.1007/s12029-017-9990-1. [DOI] [PubMed] [Google Scholar]
- 5. Kishan AU, King CR. Stereotactic body radiotherapy for low- and intermediate-risk prostate cancer. Semin Radiat Oncol. 2017;27:268–278. [DOI] [PubMed] [Google Scholar]
- 6. Baliga S, Kabarriti R, Ohri N, et al. Stereotactic body radiotherapy for recurrent head and neck cancer: a critical review. Head Neck. 2017;39:595–601. [DOI] [PubMed] [Google Scholar]
- 7. Kim MS, Kim W, Park IH, et al. Radiobiological mechanisms of stereotactic body radiation therapy and stereotactic radiation surgery. Radiat Oncol J. 2015;33:265–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Song CW, Lee YJ, Griffin RJ, et al. Indirect tumor cell death after high-dose hypofractionated irradiation: implications for stereotactic body radiation therapy and stereotactic radiation surgery. Int J Radiat Oncol Biol Phys. 2015;93:166–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Park HJ, Griffin RJ, Hui S, Levitt SH, Song CW. Radiation-induced vascular damage in tumors: implications of vascular damage in ablative hypofractionated radiotherapy (SBRT and SRS). Radiat Res. 2012;177:311–327. [DOI] [PubMed] [Google Scholar]
- 10. Brown JM, Carlson DJ, Brenner DJ. The tumor radiobiology of SRS and SBRT: are more than the 5 R’s involved? Int J Radiat Oncol Biol Phys. 2014;88:254–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26:234–241. [DOI] [PubMed] [Google Scholar]
- 12. Reynders K, Illidge T, Siva S, Chang JY, De Ruysscher D. The abscopal effect of local radiotherapy: using immunotherapy to make a rare event clinically relevant. Cancer Treat Rev. 2015;41:503–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Riaz N, Morris L, Havel JJ, Makarov V, Desrichard A, Chan TA. The role of neoantigens in response to immune checkpoint blockade. Int Immunol. 2016;28:411–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Dewan MZ, Galloway AE, Kawashima N, 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;15:5379–5388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. 2005;174:7516–7523. [DOI] [PubMed] [Google Scholar]
- 17. Lee Y, Auh SL, Wang Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114:589–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Yuan Z, Fromm A, Ahmed KA, et al. Radiotherapy rescue of a nivolumab-refractory immune response in a patient with PD-L1 negative metastatic squamous cell carcinoma of the lung. J Thorac Oncol. 2017;12:e135–e136. [DOI] [PubMed] [Google Scholar]
- 19. Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer. 2016;4:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Clement JJ, Tanaka N, Song CW. Tumor reoxygenation and postirradiation vascular changes. Radiology. 1978;127:799–803. [DOI] [PubMed] [Google Scholar]
- 21. Brieger J, Kattwinkel J, Berres M, Gosepath J, Mann WJ. Impact of vascular endothelial growth factor release on radiation resistance. Oncol Rep. 2007;18:1597–1601. [PubMed] [Google Scholar]
- 22. Furuse M, Nonoguchi N, Kawabata S, et al. Intratumoral and peritumoral post-irradiation changes, but not viable tumor tissue, may respond to bevacizumab in previously irradiated meningiomas. Radiat Oncol. 2015;10:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yao M, Galanopoulos N, Lavertu P, et al. Phase II study of bevacizumab in combination with docetaxel and radiation in locally advanced squamous cell carcinoma of the head and neck. Head Neck. 2015;37:1665–1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Hsu HW, Wall NR, Hsueh CT, et al. Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol. 2013;50:19–26. [DOI] [PubMed] [Google Scholar]
- 25. Ji X, Zhu X, Lu X. Effect of cancer-associated fibroblasts on radiosensitivity of cancer cells. Future Oncol. 2017;13:1537–1550. [DOI] [PubMed] [Google Scholar]
- 26. Kargiotis O, Chetty C, Gondi CS, et al. Adenovirus-mediated transfer of siRNA against MMP-2 mRNA results in impaired invasion and tumor-induced angiogenesis, induces apoptosis in vitro and inhibits tumor growth in vivo in glioblastoma. Oncogene. 2008;27:4830–4840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Chetty C, Bhoopathi P, Rao JS, Lakka SS. Inhibition of matrix metalloproteinase-2 enhances radiosensitivity by abrogating radiation-induced FoxM1-mediated G2/M arrest in A549 lung cancer cells. Int J Cancer. 2009;124:2468–2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother. 2011;60:319–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wasserman J, Blomgren H, Rotstein S, Petrini B, Hammarstrom S. Immunosuppression in irradiated breast cancer patients: in vitro effect of cyclooxygenase inhibitors. Bull N Y Acad Med. 1989;65:36–44. [PMC free article] [PubMed] [Google Scholar]
- 30. Rodel F, Frey B, Manda K, et al. Immunomodulatory properties and molecular effects in inflammatory diseases of low-dose X-irradiation. Front Oncol. 2:120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Trowell OA. The sensitivity of lymphocytes to ionising radiation. J Pathol Bacteriol. 1952;64:687–704. [DOI] [PubMed] [Google Scholar]
- 32. Filatenkov A, Baker J, Mueller AM, et al. Ablative tumor radiation can change the tumor immune cell microenvironment to induce durable complete remissions. Clin Cancer Res. 2015;21:3727–3739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Dovedi SJ, Cheadle EJ, Popple A, et al. Fractionated radiation therapy stimulates anti-tumor immunity mediated by both resident and infiltrating polyclonal t-cell populations when combined with PD1 blockade. Clin Cancer Res. 2017;23:5514–5526. [DOI] [PubMed] [Google Scholar]
- 34. Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhibition of metastases after treatment with local 147iation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11:728–734. [PubMed] [Google Scholar]
- 35. Gupta A, Probst HC, Vuong V, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 189:558–566. [DOI] [PubMed] [Google Scholar]
- 36. Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059. [DOI] [PubMed] [Google Scholar]
- 37. Gerber SA, Sedlacek AL, Cron KR, Murphy SP, Frelinger JG, Lord EM. IFN-γ mediates the antitumor effects of radiation therapy in a murine colon tumor. Am J Pathol. 2013;182:2345–2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lugade AA, Sorensen EW, Gerber SA, Moran JP, Frelinger JG, Lord EM. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J Immunol. 2008;180:3132–3139. [DOI] [PubMed] [Google Scholar]
- 39. Burnette BC, Liang H, Lee Y, et al. The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity. Cancer Res. 2011;71:2488–2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Deng L, Liang H, Xu M, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41:843–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Baird JR, Friedman D, Cottam B, et al. Radiotherapy combined with novel STING-targeting oligonucleotides results in regression of established tumors. Cancer Res. 2016;76:50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. 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;64:7985–7994. [DOI] [PubMed] [Google Scholar]
- 44. Wan S, Pestka S, Jubin RG, Lyu YL, Tsai YC, Liu LF. Chemotherapeutics and radiation stimulate MHC class I expression through elevated interferon-beta signaling in breast cancer cells. PLoS ONE. 2012;7:e32542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Kachikwu EL, Iwamoto KS, Liao YP, et al. Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys. 2011;81:1128–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wei S, Egenti MU, Teitz-Tennenbaum S, Zou W, Chang AE. Effects of tumor irradiation on host T-regulatory cells and systemic immunity in the context of adoptive T-cell therapy in mice. J Immunother. 2014;36:124–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. 2012;83:1306–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Wu CT, Chen MF, Chen WC, Hsieh CC. The role of IL-6 in the radiation response of prostate cancer. Radiat Oncol. 2013;8:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Xu J, Escamilla J, Mok S, et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013;73:2782–2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Crittenden MR, Savage T, Cottam B, et al. The peripheral myeloid expansion driven by murine cancer progression is reversed by radiation therapy of the tumor. PLoS ONE. 2013;8:e69527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Prakash H, Klug F, Nadella V, Mazumdar V, Schmitz-Winnenthal H, Umansky L. Low doses of gamma irradiation potentially modifies immunosuppressive tumor microenvironment by retuning tumor-associated macrophages: lesson from insulinoma. Carcinogenesis. 2016;37:301–313. [DOI] [PubMed] [Google Scholar]
- 52. Chiang CS, Fu SY, Wang SC, et al. Irradiation promotes an m2 macrophage phenotype in tumor hypoxia. Front Oncol. 2012;2:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Klug F, Prakash H, Huber PE, et al. Low-dose irradiation programs macrophage differentiation to an INOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24:589–602. [DOI] [PubMed] [Google Scholar]
- 54. Okubo M, Kioi M, Nakashima H, et al. M2-polarized macrophages contribute to neovasculogenesis, leading to relapse of oral cancer following radiation. Sci Rep. 2016;6:27548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Seifert L, Werba G, Tiwari S, et al. Radiation therapy induces macrophages to suppress T-cell responses against pancreatic tumors in mice. Gastroenterology. 2016;150:1659.e5-1672.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Chakraborty M, Abrams SI, Camphausen K, et al. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol. 2003;170:6338–6347. [DOI] [PubMed] [Google Scholar]
- 57. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 2004;64:4328–4337. [DOI] [PubMed] [Google Scholar]
- 58. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124:687–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Park SS, Dong H, Liu X, et al. PD-1 restrains radiotherapy-induced abscopal effect. Cancer Immunol Res. 2015;3:610–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520:373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74:5458–5468. [DOI] [PubMed] [Google Scholar]
- 62. de Goeje PL, Smit EF, Waasdorp C, et al. Stereotactic ablative radiotherapy induces peripheral T-cell activation in early stage lung cancer patients. Am J Respir Crit Care Med. 2017;196:1224–1227. [DOI] [PubMed] [Google Scholar]
- 63. Rutkowski J, Slebioda T, Kmiec Z, Zaucha R. Changes in systemic immune response after stereotactic ablative radiotherapy: preliminary results of a prospective study in patients with early lung cancer. Pol Arch Intern Med. 2017;127:245–253. [DOI] [PubMed] [Google Scholar]
- 64. Hong JH, Chiang CS, Tsao CY, Lin PY, McBride WH, Wu CJ. Rapid induction of cytokine gene expression in the lung after single and fractionated doses of radiation. Int J Radiat Biol. 1999;75:1421–1427. [DOI] [PubMed] [Google Scholar]
- 65. Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 2007;96:41–101. [DOI] [PubMed] [Google Scholar]
- 66. Perry AK, Chen G, Zheng D, Tang H, Cheng G. The host type I interferon response to viral and bacterial infections. Cell Res. 2005;15:407–422. [DOI] [PubMed] [Google Scholar]
- 67. Diamond MS, Kinder M, Matsushita H, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208:1989–2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. O’Neill LA, Golenbock D, Bowie AG. The history of toll-like receptors—redefining innate immunity. Nat Rev Immunol. 2013;13:453–460. [DOI] [PubMed] [Google Scholar]
- 69. Chen H, Sun H, You F, et al. Activation of STAT6 by STING is critical for antiviral innate immunity. Cell. 2011;147:436–446. [DOI] [PubMed] [Google Scholar]
- 70. Burdette DL, Vance RE. STING and the innate immune response to nucleic acids in the cytosol. Nat Immunol. 2012;14:19–26. [DOI] [PubMed] [Google Scholar]
- 71. Paludan SR, Bowie AG. Immune sensing of DNA. Immunity. 2013;38:870–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Liang Y, Peng H. STING-cytosolic DNA sensing: the backbone for an effective tumor radiation therapy. Ann Transl Med. 2016;4:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Parkes EE, Walker SM, Taggart LE, et al. Activation of STING-dependent innate immune signaling by s-phase-specific DNA damage in breast cancer. J Natl Cancer Inst. 2017;109:djw199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Bhattacharya S, Srinivasan K, Abdisalaam S, et al. RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res. 2017;45:4590–4605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Klein B, Loven D, Lurie H, et al. The effect of irradiation on expression of HLA class I antigens in human brain tumors in culture. J Neurosurg. 1994;80:1074–1077. [DOI] [PubMed] [Google Scholar]
- 76. Santin AD, Hiserodt JC, Fruehauf J, DiSaia PJ, Pecorelli S, Granger GA. Effects of irradiation on the expression of surface antigens in human ovarian cancer. Gynecol Oncol. 1996;60:468–474. [DOI] [PubMed] [Google Scholar]
- 77. Santin AD, Hermonat PL, Hiserodt JC, et al. Effects of irradiation on the expression of major histocompatibility complex class I antigen and adhesion costimulation molecules ICAM-1 in human cervical cancer. Int J Radiat Oncol Biol Phys. 1997;39:737–742. [DOI] [PubMed] [Google Scholar]
- 78. Qinfeng S, Depu W, Xiaofeng Y, Shah W, Hongwei C, Yili W. In situ observation of the effects of local irradiation on cytotoxic and regulatory T lymphocytes in cervical cancer tissue. Radiat Res. 2013;179:584–589. [DOI] [PubMed] [Google Scholar]
- 79. Peranzoni E, Zilio S, Marigo I, et al. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol. 2010;22:238–244. [DOI] [PubMed] [Google Scholar]
- 80. Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer J. 2010;16:348–353. [DOI] [PubMed] [Google Scholar]
- 81. Dolcetti L, Peranzoni E, Ugel S, et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur J Immunol. 2010;40:22–35. [DOI] [PubMed] [Google Scholar]
- 82. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol. 2008;181:5791–5802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ostrand-Rosenberg S. Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother. 2010;59:1593–1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol. 2010;184:3106–3116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh GR, Brown JM. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest. 2010;120:694–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Waring P, Mullbacher A. Cell death induced by the Fas/Fas ligand pathway and its role in pathology. Immunol Cell Biol. 1999;77:312–317. [DOI] [PubMed] [Google Scholar]
- 87. Vatner RE, Formenti SC. Myeloid-derived cells in tumors: effects of radiation. Semin Radiat Oncol. 2015;25:18–27. [DOI] [PubMed] [Google Scholar]
- 88. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. [DOI] [PubMed] [Google Scholar]
- 89. Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev. 2006;25:315–322. [DOI] [PubMed] [Google Scholar]
- 90. Leblond MM, Gerault AN, Corroyer-Dulmont A, et al. Hypoxia induces macrophage polarization and re-education toward an M2 phenotype in U87 and U251 glioblastoma models. Oncoimmunology. 2015;5:e1056442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Song CW, Payne JT, Levitt SH. Vascularity and blood flow in X-irradiated walker carcinoma 256 of rats. Radiology. 1972;104:693–697. [DOI] [PubMed] [Google Scholar]
- 92. Song CW, Levitt SH. Vascular changes in walker 256 carcinoma of rats following X irradiation. Radiology. 1971;100:397–407. [DOI] [PubMed] [Google Scholar]
- 93. Wong HH, Song CW, Levitt SH. Early changes in the functional vasculature of walker carcinoma 256 following irradiation. Radiology. 1973;108:429–434. [DOI] [PubMed] [Google Scholar]
- 94. Kocher M, Treuer H, Voges J, Hoevels M, Sturm V, Muller RP. Computer simulation of cytotoxic and vascular effects of radiosurgery in solid and necrotic brain metastases. Radiother Oncol. 2000;54:149–156. [DOI] [PubMed] [Google Scholar]
- 95. Maeda A, Chen Y, Bu J, Mujcic H, Wouters BG, DaCosta RS. In vivo imaging reveals significant tumor vascular dysfunction and increased tumor hypoxia-inducible factor-1α expression induced by high single-dose irradiation in a pancreatic tumor model. Int J Radiat Oncol Biol Phys. 2017;97:184–194. [DOI] [PubMed] [Google Scholar]
- 96. Solesvik OV, Rofstad EK, Brustad T. Vascular changes in a human malignant melanoma xenograft following single-dose irradiation. Radiat Res. 1984;98:115–128. [PubMed] [Google Scholar]
- 97. Dings RP, Williams BW, Song CW, Griffioen AW, Mayo KH, Griffin RJ. Anginex synergizes with radiation therapy to inhibit tumor growth by radiosensitizing endothelial cells. Int J Cancer. 2005;115:312–319. [DOI] [PubMed] [Google Scholar]
- 98. Emami B, Ten Haken RK, Nussbaum GH, Hughes WL. Effects of single-dose irradiation in tumor blood flow studied by 15O decay after proton activation in situ. Radiology. 1981;141:207–209. [DOI] [PubMed] [Google Scholar]
- 99. Chen FH, Chiang CS, Wang CC, et al. Radiotherapy decreases vascular density and causes hypoxia with macrophage aggregation in TRAMP-C1 prostate tumors. Clin Cancer Res. 2009;15:1721–1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kim DW, Huamani J, Niermann KJ, et al. Noninvasive assessment of tumor vasculature response to radiation-mediated, vasculature-targeted therapy using quantified power Doppler sonography: implications for improvement of therapy schedules. J Ultrasound Med. 2006;25:1507–1517. [DOI] [PubMed] [Google Scholar]
- 101. Kobayashi H, Reijnders K, English S, et al. Application of a macromolecular contrast agent for detection of alterations of tumor vessel permeability induced by radiation. Clin Cancer Res. 2004;10:7712–7720. [DOI] [PubMed] [Google Scholar]
- 102. Kuwahara Y, Mori M, Kitahara S, et al. Targeting of tumor endothelial cells combining 2 Gy/day of X-ray with Everolimus is the effective modality for overcoming clinically relevant radioresistant tumors. Cancer Med. 2014;3:310–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Dewhirst MW, Oliver R, Tso CY, Gustafson C, Secomb T, Gross JF. Heterogeneity in tumor microvascular response to radiation. Int J Radiat Oncol Biol Phys. 1990;18:559–568. [DOI] [PubMed] [Google Scholar]
- 104. Sonveaux P, Dessy C, Brouet A, et al. Modulation of the tumor vasculature functionality by ionizing radiation accounts for tumor radiosensitization and promotes gene delivery. FASEB J. 2002;16:1979–1981. [DOI] [PubMed] [Google Scholar]
- 105. Crokart N, Jordan BF, Baudelet C, et al. Early reoxygenation in tumors after irradiation: determining factors and consequences for radiotherapy regimens using daily multiple fractions. Int J Radiat Oncol Biol Phys. 2005;63:901–910. [DOI] [PubMed] [Google Scholar]
- 106. Heissig B, Rafii S, Akiyama H, et al. Low-dose irradiation promotes tissue revascularization through VEGF release from mast cells and MMP-9-mediated progenitor cell mobilization. J Exp Med. 2005;202:739–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Sofia Vala I, Martins LR, Imaizumi N, et al. Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis. PLoS ONE. 2010;5:e11222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Lerman OZ, Greives MR, Singh SP, et al. Low-dose radiation augments vasculogenesis signaling through HIF-1-dependent and -independent SDF-1 induction. Blood. 2010;116:3669–3676. [DOI] [PubMed] [Google Scholar]
- 109. Thanik VD, Chang CC, Lerman OZ, et al. Cutaneous low-dose radiation increases tissue vascularity through upregulation of angiogenic and vasculogenic pathways. J Vasc Res. 2010;47:472–480. [DOI] [PubMed] [Google Scholar]
- 110. Chen FH, Fu SY, Yang YC, Wang CC, Chiang CS, Hong JH. Combination of vessel-targeting agents and fractionated radiation therapy: the role of the SDF-1/CXCR4 pathway. Int J Radiat Oncol Biol Phys. 2013;86:777–784. [DOI] [PubMed] [Google Scholar]
- 111. Wagner-Ecker M, Schwager C, Wirkner U, Abdollahi A, Huber PE. MicroRNA expression after ionizing radiation in human endothelial cells. Radiat Oncol. 2010;5:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Garcia-Barros M, Paris F, Cordon-Cardo C, et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science. 2003;300:1155–1159. [DOI] [PubMed] [Google Scholar]
- 113. Hawsawi NM, Ghebeh H, Hendrayani SF, et al. Breast carcinoma-associated fibroblasts and their counterparts display neoplastic-specific changes. Cancer Res. 2008;68:2717–2725. [DOI] [PubMed] [Google Scholar]
- 114. Rodier F, Coppe JP, Patil CK, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11:973–979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Hellevik T, Pettersen I, Berg V, et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat Oncol. 2012;7:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Papadopoulou A, Kletsas D. Human lung fibroblasts prematurely senescent after exposure to ionizing radiation enhance the growth of malignant lung epithelial cells in vitro and in vivo. Int J Oncol. 2011;39:989–999. [DOI] [PubMed] [Google Scholar]
- 117. Ohuchida K, Mizumoto K, Murakami M, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004;64:3215–3222. [DOI] [PubMed] [Google Scholar]
- 118. Qayyum MA, Insana MF. Stromal responses to fractionated radiotherapy. Int J Radiat Biol. 2012;88:383–392. [DOI] [PubMed] [Google Scholar]
- 119. Qayyum MA, Kwak JT, Insana MF. Stromal-epithelial responses to fractionated radiotherapy in a breast cancer microenvironment. Cancer Cell Int. 2015;15:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Grabham P, Hu B, Sharma P, Geard C. Effects of ionizing radiation on three-dimensional human vessel models: differential effects according to radiation quality and cellular development. Radiat Res. 2011;175:21–28. [DOI] [PubMed] [Google Scholar]
- 121. Karam SD, Bhatia S. The radiobiological targets of SBRT: tumor cells or endothelial cells? Ann Transl Med. 2015;3:290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Fuks Z, Kolesnick R. Engaging the vascular component of the tumor response. Cancer Cell. 2005;8:89–91. [DOI] [PubMed] [Google Scholar]
- 123. Martin BJ. Inhibiting vasculogenesis after radiation: a new paradigm to improve local control by radiotherapy. Semin Radiat Oncol. 2013;23:281–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Truman JP, Garcia-Barros M, Kaag M, et al. Endothelial membrane remodeling is obligate for anti-angiogenic radiosensitization during tumor radiosurgery. PLoS ONE. 2010;5:e12310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Lindblom E, Antonovic L, Dasu A, Lax I, Wersall P, Toma-Dasu I. Treatment fractionation for stereotactic radiotherapy of lung tumours: a modelling study of the influence of chronic and acute hypoxia on tumour control probability. Radiat Oncol. 2014;9:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Brown JM, Diehn M, Loo BW., Jr. Stereotactic ablative radiotherapy should be combined with a hypoxic cell radiosensitizer. Int J Radiat Oncol Biol Phys. 2010;78:323–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Carlson DJ, Keall PJ, Loo BW, Jr, Chen ZJ, Brown JM. Hypofractionation results in reduced tumor cell kill compared to conventional fractionation for tumors with regions of hypoxia. Int J Radiat Oncol Biol Phys. 2011;79:1188–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Overgaard J. Hypoxic radiosensitization: adored and ignored. J Clin Oncol. 2007;25:4066–4074. [DOI] [PubMed] [Google Scholar]
- 129. Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432:332–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401. [DOI] [PubMed] [Google Scholar]
- 131. Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 2006;5:1597–1601. [DOI] [PubMed] [Google Scholar]
- 132. Shimoda M, Mellody KT, Orimo A. Carcinoma-associated fibroblasts are a rate-limiting determinant for tumour progression. Semin Cell Dev Biol. 2010;21:19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Ksiazkiewicz M, Gottfried E, Kreutz M, Mack M, Hofstaedter F, Kunz-Schughart LA. Importance of CCL2-CCR2A/2B signaling for monocyte migration into spheroids of breast cancer-derived fibroblasts. Immunobiology. 2010;215:737–747. [DOI] [PubMed] [Google Scholar]
- 134. Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–348. [DOI] [PubMed] [Google Scholar]
- 135. Tseng D, Vasquez-Medrano DA, Brown JM. Targeting SDF-1/CXCR4 to inhibit tumour vasculature for treatment of glioblastomas. Br J Cancer. 2011;104:1805–1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Mishra P, Banerjee D, Ben-Baruch A. Chemokines at the crossroads of tumor-fibroblast interactions that promote malignancy. J Leukoc Biol. 2011;89:31–39. [DOI] [PubMed] [Google Scholar]
- 137. Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9:1392–1400. [DOI] [PubMed] [Google Scholar]
- 138. Duda DG, Duyverman AM, Kohno M, et al. Malignant cells facilitate lung metastasis by bringing their own soil. Proc Natl Acad Sci U S A. 2010;107:21677–21682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Tachiiri S, Katagiri T, Tsunoda T, Oya N, Hiraoka M, Nakamura Y. Analysis of gene-expression profiles after gamma irradiation of normal human fibroblasts. Int J Radiat Oncol Biol Phys. 2006;64:272–279. [DOI] [PubMed] [Google Scholar]
- 140. Velarde MC, Demaria M, Campisi J. Senescent cells and their secretory phenotype as targets for cancer therapy. Interdiscip Top Gerontol. 2013;38:17–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Liu D, Hornsby PJ. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007;67:3117–3126. [DOI] [PubMed] [Google Scholar]
- 142. Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health and disease. J Immunol. 2005;174:4453–4460. [DOI] [PubMed] [Google Scholar]
- 143. Haston CK, Begin M, Dorion G, Cory SM. Distinct loci influence radiation-induced alveolitis from fibrosing alveolitis in the mouse. Cancer Res. 2007;67:10796–10803. [DOI] [PubMed] [Google Scholar]
- 144. Barcellos-Hoff MH, Derynck R, Tsang ML, Weatherbee JA. Transforming growth factor-beta activation in irradiated murine mammary gland. J Clin Invest. 1994;93:892–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006;24:99–146. [DOI] [PubMed] [Google Scholar]
- 146. Straub JM, New J, Hamilton CD, Lominska C, Shnayder Y, Thomas SM. Radiation-induced fibrosis: mechanisms and implications for therapy. J Cancer Res Clin Oncol. 2015;141:1985–1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol. 2010;97:149–161. [DOI] [PubMed] [Google Scholar]
- 148. Barcellos-Hoff MH. Stromal mediation of radiation carcinogenesis. J Mammary Gland Biol Neoplasia. 2010;15:381–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Hellevik T, Pettersen I, Berg V, et al. Changes in the secretory profile of NSCLC-associated fibroblasts after ablative radiotherapy: potential impact on angiogenesis and tumor growth. Transl Oncol. 2013;6:66–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Mantoni TS, Lunardi S, Al-Assar O, Masamune A, Brunner TB. Pancreatic stellate cells radioprotect pancreatic cancer cells through β1-integrin signaling. Cancer Res. 2011;71:3453–3458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Crittenden M, Kohrt H, Levy R, et al. Current clinical trials testing combinations of immunotherapy and radiation. Semin Radiat Oncol. 2015;25:54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Tang C, Welsh JW, de Groot P, et al. Ipilimumab with stereotactic ablative radiation therapy: phase I results and immunologic correlates from peripheral T cells. Clin Cancer Res. 2017;23:1388–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Haymaker CL, Kim D, Uemura M, et al. Metastatic melanoma patient had a complete response with clonal expansion after whole brain radiation and PD-1 blockade. Cancer Immunol Res. 2017;5:100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Qian JM, Yu JB, Kluger HM, Chiang VL. Timing and type of immune checkpoint therapy affect the early radiographic response of melanoma brain metastases to stereotactic radiosurgery. Cancer. 2016;122:3051–3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Shaverdian N, Lisberg AE, Bornazyan K, et al. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol. 2017;18:895–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Cuneo KC, Geng L, Fu A, Orton D, Hallahan DE, Chakravarthy AB. SU11248 (sunitinib) sensitizes pancreatic cancer to the cytotoxic effects of ionizing radiation. Int J Radiat Oncol Biol Phys. 2008;71:873–879. [DOI] [PubMed] [Google Scholar]
- 157. Schueneman AJ, Himmelfarb E, Geng L, et al. SU11248 maintenance therapy prevents tumor regrowth after fractionated irradiation of murine tumor models. Cancer Res. 2003;63:4009–4016. [PubMed] [Google Scholar]
- 158. Crane CH, Eng C, Feig BW, et al. Phase II trial of neoadjuvant bevacizumab, capecitabine, and radiotherapy for locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. 2009;76:824–830. [DOI] [PubMed] [Google Scholar]
- 159. Das P, Eng C, Rodriguez-Bigas MA, et al. Preoperative radiation therapy with concurrent capecitabine, bevacizumab, and erlotinib for rectal cancer: a phase 1 trial. Int J Radiat Oncol Biol Phys. 2013;88:301–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Morganti AG, Mignogna S, Caravatta L, et al. FOLFIRI-bevacizumab and concurrent low-dose radiotherapy in metastatic colorectal cancer: preliminary results of a phase I-II study. J Chemother. 2014;26:353–358. [DOI] [PubMed] [Google Scholar]
- 161. Ney DE, Carlson JA, Damek DM, et al. Phase II trial of hypofractionated intensity-modulated radiation therapy combined with temozolomide and bevacizumab for patients with newly diagnosed glioblastoma. J Neurooncol. 2014;122:135–143. [DOI] [PubMed] [Google Scholar]
- 162. Fury MG, Xiao H, Sherman EJ, et al. Phase II trial of bevacizumab + cetuximab + cisplatin with concurrent intensity-modulated radiation therapy for patients with stage III/IVB head and neck squamous cell carcinoma. Head Neck. 2016;38:E566–E570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Zhuang HQ, Yuan ZY, Wang P. Research progress on the mechanisms of combined bevacizumab and radiotherapy. Recent Pat Anticancer Drug Discov. 2014;9:129–134. [DOI] [PubMed] [Google Scholar]
- 164. Lubelski D, Abdullah KG, Weil RJ, Marko NF. Bevacizumab for radiation necrosis following treatment of high grade glioma: a systematic review of the literature. J Neurooncol. 2013;115:317–322. [DOI] [PubMed] [Google Scholar]
- 165. Matuschek C, Bolke E, Nawatny J, et al. Bevacizumab as a treatment option for radiation-induced cerebral necrosis. Strahlenther Onkol. 2011;187:135–139. [DOI] [PubMed] [Google Scholar]
- 166. Ney DE, Carlson JA, Damek DM, et al. Phase II trial of hypofractionated intensity-modulated radiation therapy combined with temozolomide and bevacizumab for patients with newly diagnosed glioblastoma. J Neurooncol. 2015;122:135–143. [DOI] [PubMed] [Google Scholar]
- 167. Gordon MS, Mendelson DS, Kato G. Tumor angiogenesis and novel antiangiogenic strategies. Int J Cancer. 2010;126:1777–1787. [DOI] [PubMed] [Google Scholar]
- 168. Zwolak P, Dudek AZ, Bodempudi VD, et al. Local irradiation in combination with bevacizumab enhances radiation control of bone destruction and cancer-induced pain in a model of bone metastases. Int J Cancer. 2008;122:681–688. [DOI] [PubMed] [Google Scholar]
- 169. El Kaffas A, Giles A, Czarnota GJ. Dose-dependent response of tumor vasculature to radiation therapy in combination with sunitinib depicted by three-dimensional high-frequency power Doppler ultrasound. Angiogenesis. 2013;16:443–454. [DOI] [PubMed] [Google Scholar]
- 170. Kleibeuker EA, Ten Hooven MA, Verheul HM, Slotman BJ, Thijssen VL. Combining radiotherapy with sunitinib: lessons (to be) learned. Angiogenesis. 2015;18:385–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Tong CC, Ko EC, Sung MW, et al. Phase II trial of concurrent sunitinib and image-guided radiotherapy for oligometastases. PLoS ONE. 2012;7:e36979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Staehler M, Haseke N, Stadler T, et al. Feasibility and effects of high-dose hypofractionated radiation therapy and simultaneous multi-kinase inhibition with sunitinib in progressive metastatic renal cell cancer. Urol Oncol. 2012;30:290–293. [DOI] [PubMed] [Google Scholar]
- 173. Staehler M, Haseke N, Nuhn P, et al. Simultaneous anti-angiogenic therapy and single-fraction radiosurgery in clinically relevant metastases from renal cell carcinoma. BJU Int. 2011;108:673–678. [DOI] [PubMed] [Google Scholar]
- 174. Brooks C, Sheu T, Bridges K, et al. Preclinical evaluation of sunitinib, a multi-tyrosine kinase inhibitor, as a radiosensitizer for human prostate cancer. Radiat Oncol. 2012;7:154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Kao J, Packer S, Vu HL, et al. Phase 1 study of concurrent sunitinib and image-guided radiotherapy followed by maintenance sunitinib for patients with oligometastases: acute toxicity and preliminary response. Cancer. 2009;115:3571–3580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Wuthrick EJ, Kamrava M, Curran WJ, Jr, et al. A phase 1B trial of the combination of the antiangiogenic agent sunitinib and radiation therapy for patients with primary and metastatic central nervous system malignancies. Cancer. 2011;117:5548–5559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Barcellos-Hoff MH. Remodeling of the irradiated tumor microenvironment: the fifth R of radiobiology. In: Tofilon PJ, Camphausen K. eds. Increasing the Therapeutic Ratio of Radiotherapy. 2016:135–149. [Google Scholar]
- 178. Golden EB, Formenti SC. Is tumor (R)ejection by the immune system the “5th R” of radiobiology? Oncoimmunology. 2014;3(1):e28133. [DOI] [PMC free article] [PubMed] [Google Scholar]