Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 15.
Published in final edited form as: Clin Cancer Res. 2018 Nov 9;25(6):1709–1717. doi: 10.1158/1078-0432.CCR-18-2581

The reciprocity between radiotherapy and cancer immunotherapy

Yifan Wang 1,#, Zhi-gang Liu 2,#, Hengfeng Yuan 3,6, Weiye Deng 1, Jing Li 4, Yuhui Huang 5, Betty YS Kim 6, Michael D Story 1, Wen Jiang 1,4
PMCID: PMC6420874  NIHMSID: NIHMS1511721  PMID: 30413527

Abstract

The clinical success of immune checkpoint inhibitors in treating metastatic and refractory cancers has generated significant interest in investigating their role in treating locally advanced diseases, thus requiring them to be combined with standard treatments in hope to produce synergistic antitumor responses. Radiotherapy in particular, has long been hypothesized to have actions complementary to those of immune checkpoint blockade, and a growing body of evidence indicates that cancer immunotherapy may also have radiosensitizing effects, which would provide unique benefit for locoregional treatments. Recent studies have demonstrated that when immune cells are activated by immunotherapeutics, they can reprogram the tumor microenvironment in ways that may potentially increase the radiosensitivity of the tumor. In this review, we highlight the evidence that supports reciprocal interactions between cancer immunotherapy and radiotherapy, where in addition to the traditional notion that radiation serves to enhance the activation of antitumor immunity, an alternative scenario also exists in which T-cell activation by cancer immunotherapy may sensitize tumors to radiation treatment through mechanisms that include normalization of the tumor vasculature and tissue hypoxia. We describe the empirical observations from preclinical models that support such effects and discuss their implications for future research and trial design.

Keywords: Radiotherapy, immunotherapy, immune checkpoint blockade, vessel normalization, radiation sensitization

Introduction

Recent successes of immune checkpoint inhibitors for metastatic cancers have resulted in their increased investigation in treating locally advanced diseases, often in combination with other treatment modalities such as radiation. Radiation is the standard of care therapy for many types of cancer. Although ionizing radiation is classically known to induce tumor cell killing via direct or indirect damage to the cellular DNA, a growing list of evidences begins to suggest that radiation may also eliminate tumors via activation of local and/or systemic immune responses, particularly when it is combined with immune stimulating agents such as immune checkpoint inhibitors (Table 1) (18). Despite these exciting observations, the precise mechanism of how radiation and immunotherapy benefit one another remains unclear. Although ionizing radiation can induced immunological changes within the tumor microenvironment including facilitate tumor antigen release (9), increase effector T cell infiltration (10), and up-regulate MHC-1 molecule on tumor cells (11), recent evidences suggest that cancer immunotherapy such as immune checkpoint inhibitors may also have radiosensitization effects (12). The latter concept is especially important and clinically relevant because larger primary tumors often respond poorly to immunotherapies and require local therapies such as radiation. In this review, we summarize the evidences that support radiation’s immune stimulating effects and propose the mechanisms by which immunotherapies such as immune checkpoint blockade may serve as novel radiosensitizers. Finally, we discuss the implications of this concept for clinical studies and future trial design.

Table 1:

Selected studies using combination of radiotherapy and immunotherapy

Study Type Disease Sequence RT IT Brief results Reference
Clinical: stage III trial Lung cancer RT, IT Definitive RT (54 to 66 Gy) durvalumab PFS improvement with durvalumab, similar side effects 56. Antonia et al. 2017
Clinical: stage I-II trial Various Concurrent 35 Gy in 10 fractions GM-CSF Abscopal responses in 27.6% patients 7. Golden et al. 2015
Clinical Melanoma IT, RT, IT 2850 cGy in 3 fractions over 7 days ipilimumab Abscopal effect, peripheral-blood immune cell changes 1. Postow et al. 2012
Clinical Lung cancer RT, IT, RT Fractionation RT to primary and meta tumors nivolumab Abscopal effect 3. Schoenhals et al. 2016
Clinical Pancreatic cancer Concurrent 45 Gy in 15 fractions GM-CSF Abscopal effect, survival benefit 6. Shi et al. 2017
Clinical Metastatic melanoma IT, RT, IT WBRT 30 Gy in 10 fractions ipilimumab, pembrolizumab Status improvement, long-term survival 4. Haymaker et al. 2017
Clinical Various Various Various doses anti-CTLA4, anti-PD-1/PD-L1 Induction immunotherapy begun more than 30 days before radiation resulted in longer OS 57. Samstein et al. 2017
Clinical Brain metastasis Various Various, 18 – 30 Gy ipilimumab, pembrolizumab, or nivolumab Immunotherapy increased radiation necrosis 59. Martin et al. 2018
Preclinical Melanoma NA 15 Gy vs. 15 Gy in 3 fractions NA 15 Gy single-dose generated more tumor-infiltrating T cells 62. Lugade et al. 2005
Preclinical Colon cancer RT, IT 10 Gy T cell adoptive transfer RT increased antigen presentation and enhanced IT efficacy 11. Reits et al. 2006
Preclinical Melanoma NA 20 Gy vs. 20 Gy in 4 fractions NA Immune response triggered by ablative radiation doses 61. Lee et al. 2009
Preclinical Prostate cancer NA 1 Gy × 10 vs. 10 Gy NA Multifraction radiation induced more DAMP release 66. Aryankalayil et al. 2014
Preclinical Pancreatic cancer and melanoma Various 20 Gy, 8 Gy anti-CTLA4, anti-PD-L1 When combined with radiation, anti-CTLA4 and anti-PD-L1 promotes response through different mechanisms 9. Twyman-Saint Victor et al. 2015
Preclinical Colon cancer NA 30 Gy vs. 30 Gy in 10 fractions NA Ablative dose changed tumor immune microenvironment 10. Filatenkov et al. 2015
Preclinical Breast cancer RT, IT 6 Gy × 5 anti-TGF-beta, anti-PD-1 Anti-PD-1 prolonged survival of mice treated with RT and TGF-beta blockade 43. Vanpouille-Box et al. 2015
Preclinical Pancreatic cancer Concurrent 10 Gy cyclic dinucleotides STING activator and RT controlled both local and distant tumors synergistically 34. Baird et al. 2016
Preclinical Colon cancer IT, RT vs. RT, IT 20 Gy anti-CTLA4 Anti-CTLA4 was most effective when given prior to radiation 55. Young et al. 2016
Preclinical Colon cancer IT, RT vs. RT, IT 20 Gy anti-OX40 Anti-OX40 was more effective when given 1 day post radiation 55. Young et al. 2016
Preclinical Colon and breast cancer RT, IT 8 Gy × 3 vs. 20 Gy anti-CTLA4 Anti-CTLA4 therapy only synergize with low dose radiation to induce an abscopal effect 65. Vanpouille-Box et al. 2017
Preclinical Breast and colon cancer IT, RT 0.5 Gy × 6 indoleamine 2,3-dioxygenase inhibitor RT and IT induced rejection of both irradiated and non-irradiated tumors 8. Lu et al. 2018

Abbreviations: RT: radiotherapy; IT: immunotherapy; WBRT; whole brain radiotherapy; GM-CSF: Granulocyte-macrophage colony-stimulating factor; DAMP; Damage-associated molecular patterns; PFS: progression free survival; OS: overall survival; NA: not applicable.

Immune-modulating effects of radiation

Radiation increases antigen visibility

The ability to evade immune surveillance is one of the hallmarks of cancer (13). A tumor escapes from the host’s immune surveillance by several mechanisms, including evading detection (i.e., by downregulating MHC class I expression) and avoiding destruction (e.g., by creating an immunosuppressive tumor microenvironment) (14). However, radiation may render tumor cells antigens more visible to the immune system. Several mechanisms are involved in this “unmasking” process. First, radiation-induced DNA damage may lead to increased release of neoantigens by tumor cells for immune recognition (15). Given that tumor cells often accumulate DNA mutations as the result of DNA repair deficiencies, a higher burden of mutant nucleic acids and proteins has increased propensity for trigger immunogenic responses against tumors treated with immune checkpoint blockade (1517). Ionizing radiation itself may also induce many DNA damages, including base modification, single strand break, and double strand break (18). These DNA alterations generated by radiation may act as a rich source of neoantigens that increase immune surveillance. In addition to modifying antigen release, radiation could also upregulate the expression of MHC molecules on cancer cells to facilitate presentation of tumor antigen to cytotoxic T cells. Activation of T cells is dependent on the recognition and binding of T cell receptor with peptide-bound MHC molecules. However, MHC-I is often downregulated by tumors to evade immune recognition (19). Radiation could increase the surface expression of MHC-I on tumor cells, making them more visible to cytotoxic T cells (11). Finally, radiation promotes the phagocytosis of damaged tumor cells by antigen-presenting cells, leading to increased priming of tumor-specific T cells. The pro-phagocytic effect of radiation is mediated by the release of damage-associated molecular pattern (DAMP) by tumor cells upon irradiation, including the translocation of the ER chaperone calreticulin, to the cell membrane. This immunogenic cell death process is also characterized by the release of high mobility group box 1 (HMGB1) and ATP, which together promotes tumor cell phagocytosis by macrophages and dendritic cells (20,21). These immune modulatory effects of radiation appear to be further augmented when it is combined with immune checkpoint blockers (22,23).

Radiation activates the cGAS-STING innate immune response

In addition to enhance the release and presentation of tumor-associated antigens, radiation may also activate antitumor immune responses by triggering the Stimulator of Interferon Genes (STING)-mediated DNA-sensing pathway (24). STING is part of the innate immune response that protects hosts from DNA pathogens (25) and is essential for host immunity (26). Radiation damage releases nuclear DNA into the cytosol (24). The presence of mutant DNA within the cytoplasma results in cyclin GMP-AMP (cGAMP), the product of cyclic GMP-AMP synthase (cGAS), to upregulate the transcription of type I interferon (IFN-I) genes via the STING-NFκB signaling pathway (27,28). Alternatively, STING and type I IFN activation can occur through ATM and IFI16 in a cGAS-independent manner (29). Type I IFN has important roles in regulating dentritic cell function and helper T-cell differentiation (30). The cGAS-STING pathway is essential for dendritic cells to sense the irradiated cancer cells and induce adaptive immune responses (31). However, the precise molecular mechanism governing STING activation in the presence of radiation is highly complex. A recent study found that both canonical and non-canonical NFκB signaling pathways were involved in radiation-induced type I IFN responses in dendritic cells (32). In murine colon cancer and melanoma models, the canonical NFκB pathway was required for the therapeutic effect of radiation, however, radiation also activated non-canonical NFκB pathway through cGAS-STING DNA sensing pathway to inhibit expression of IFN-β in dendritic cells. The inhibition of non-canonical NFκB pathway enhanced dendritic cell priming and the treatment efficacy of radiation (32). This study not only highlights the critical function of cGAS-STING in the immune responses after radiation, but also reveals the intriguing signal network downstream of cGAS-STING pathway.

Recognizing the importance of the STING pathway in antitumor immunity has led to several studies investigating the therapeutic potential of STING agonists in anticancer therapy. The direct activition of STING by agonists, either as monotherapy or in combination with radiation, has shown to induce both local and systemic antitumor immune response in murine cancer models (33,34). Currently, a phase I clinical trial (NCT02675439) is evaluating the safety and efficacy of a STING agonist, MIW815, for patients with advanced solid tumors or lymphoma. However, the role of STING in cancer immunotherapy remains controversial. A recent study showed that STING activation after radiation recruited myeloid-derived suppressor cells to the tumor, may in fact induce radioresistance and immunosuppression (35). Furthermore, the STING pathway may also promote immune tolerance by modulating the tumor microenvironment (36,37). Therefore, additional studies are warranted to investigate the biology and the role of the STING pathway in immune regulation within solid tumors.

Radiation modulates the tumor microenvironment

In addition to direct cancer cell killing, radiation may also reprogram the tumor microenvironment from an immunosuppressive to an immunostimulatory phenotype (38). Radiation can increase infiltration of CD8+ T cells while decreasing myeloid-derived suppressor cells, depending on the presence of cross-presenting dendritic cells and IFN-γ (10). Low-dose radiation promotes normalization of the tumor vasculature and polarization of M2-like macrophages toward an M1-like iNOS+ phenotype (39). The iNOS+ macrophages induce the expression of Th1 chemokines that recruit CD8+ and CD4+ T cells to the tumor, promoting T-cell mediated antitumor effects and prolonging survival in murine models of pancreatic cancer (39). On the other hand, radiation also stimulates secretion of cytokines that induce immunosuppression. For example, transforming growth factor-β (TGF-β) is upregulated shortly after radiation. TGF-β induces the suppression of CD8+ T cells and promotes regulatory T-cell transformation, causing immunosuppression in the tumor (20,40,41). In preclinical models, TGF-β neutralization regulated T-cells to promote anti-tumor immunity of dendritic cells (42). When combined with radiation, TGF-β neutralization increased IFN-γ-associated gene signatures and enhanced CD8+ T-cell response (43). Moreover, the combination of radiation and TGF-β neutralization controlled both local and distant tumor growth more effectively than did either treatment given alone (43).

Radiation regulates immune checkpoint expression

Radiation may also indirectly regulate the expression levels of immune checkpoint molecules on the surfaces of both cancer cells and immune cells within the tumor microenvironment through IFN-γ (12). Alternatively, a recent study suggested that radiation-induced DNA double-strand breakage upregulates the expression of PD-L1 on tumor cells via ATM/ATR/Chk1 kinases (44). Given that over-expression of PD-L1 within tumors is associated with improved responses to anti-PDL1 therapy (45), radiation may serve as an effective neoadjuvant treatment to increase the effectiveness of immune checkpoint blockade. However, the exact relationship between radiation and PDL1 induction has not been completely elucidated, and it remains to be seen whether it can be applied to difference tumors. The currently known mechanisms by which radiation enhances immunotherapy are summarized in Figure 1.

Figure 1. Mechanisms by which radiation enhances immunotherapy.

Figure 1.

Cancer-specific peptides released from damaged cancer cells facilitate antigen uptake and presentation by dendritic cells. Radiation also decreases an anti-phagocytosis signal (CD47) and increases a pro-phagocytosis signal (calreticulin) to enhance calreticulin-LRP/CD91-mediated phagocytosis by macrophages, thereby increasing antigen presentation and priming of T cells. Upregulation of MHC-I expression on tumor cells after irradiation increases recognition of cancer cells by T cells. After radiation, damaged DNA is released from nucleus to cytosol, which triggers the cGAS-STING pathway to activate interferon gene transcription. Several proteins (such as TGF-β and HMGB1) secreted by cancer cells also modulate the immune microenvironment.

Radiosensitization by immune checkpoint blockades

Hypoxic tumor microenvironment induces radioresistance

The abnormal and dysfunctional nature of the tumor vasculature causes hypoxia, limits drug delivery, and contributes to an overall immunosuppressive microenvironment (46,47). Meanwhile, hypoxic microenvironments induce excessive secretion of proangiogenic signals, such as vascular endothelial growth factor (VEGF), resulting in rapid but abnormal tumor vessel formation. Hypoxia directly and indirectly induces radioresistance via several mechanisms. Firstly, the lack of oxygen within hypoxic tumors results in fewer DNA damages from the same dose of radiation as compared to well oxygenated ones (48,49). Additionally, hypoxia also drives radioresistance by accumulating and stabilizing hypoxia-inducible factor-1 (HIF1) (48,50). HIF1 activation increases the expression of key enzymes that drive glycolysis leading to the accumulation of lactate and pyruvate as well as the anti-oxidants glutathione and NADPH. The latter molecules scavenge reactive oxygen species (ROS) generated by radiation exposure to limit DNA damage (49). Lactate, on the other hand, affects immune cell maturation leading to immune escape and immunotherapy resistance (51). Lactate also upregulates the HIF1 pathway creating a futile cycle of radio- and immune resistance (49,52). Collectively, hypoxia is one of the major drivers of tumor radioresistance; therefore, targeting tumor hypoxia may increase tumor radiosensitivity.

Tumor vascular normalization and hypoxia reduction may sensitize tumors to radiation

Recent studies suggest that immune checkpoint blockade not only unleashes T cells to attack tumor cells but may also modulate the tumor microenvironment by normalizing tumor vessels (47,53,54). This novel interaction creates the potential opportunity for immunotherapy to reduce tumor hypoxia and increase radiosensitivity. One recent stud showed that cancer immunotherapy induced tumor-vascular normalization in a T-cell-dependent manner (54). Specifically, when murine breast and colon tumor models were treated by anti-CTLA4 or anti-PD1 agents, the responding tumors showed strong growth inhibition as well as increased vessel perfusion and decreased intratumoral hypoxia. These immunotherapies also normalized the tumor vasculature through the accumulation of IFN-γ producing CD8+ cells. Importantly, the extent of tumor vascular normalization by immune checkpoint inhibitor is strongly associated with its therapeutic efficacy (54). Neutralization of IFN-γ or CD8+ T cell depletion abrogated both the vascular normalization and antitumor effect of immune checkpoint blockade, suggesting that both processes are intricately related to one another. A related study using bioinformatics analysis also identified a correlation between immunostimulatory pathways and vascular normalization-related genes. This study found that type 1 T-helper (TH1) cells were involved in the normalization of the tumor vasculature (53). Upon blockade of immune checkpoint, IFN-γ secretion of CD4+ T cells is prompted, which promotes vessel normalization and reduces hypoxia (53). Based on these evidences, it is highly plausible that the tumor vascular normalizing effects of immune checkpoint blockade not only create a feedback loop to reprogram the tumor immune microenvironment and enhance the efficacy of immunotherapy but also may sensitize tumors to radiation (Figure 2). With the combination of immune checkpoint inhibitors and radiation been tested in multiple clinical trials, this notion that cancer immunotherapy may promote radiosensitivity via normalization of tumor vasculature and hypoxia carries increased clinical significance. Detailed characterization of the intricate reciprocal relationships between radiation, vascular remodeling and immune regulation is critical to develop optimal combination immunotherapy-radiotherapy treatment for cancer patients.

Figure 2. Immunotherapy sensitizes tumors to radiation by modulating the tumor microenvironment.

Figure 2.

Immunotherapy increases T-cell infiltration into tumors. The infiltrated T cells secrete cytokines such as IFN-γ, which normalize the tumor vasculature. This vessel normalization increases perfusion and oxygen supply in the tumor microenvironment, and therefore could sensitize tumors to radiation.

Implications for clinical translation

Optimizing the timing of radiation in combination with immunotherapy

Considering the complexities of the interplay between radiation and immunotherapy, careful planning as to the timing of radiation to be given with immunotherapy could be crucial to optimizing treatment efficacy (20). One preclinical study indicated that the best timing relative to radiation was different for anti-CTLA4 and anti-OX40, suggesting that the optimal treatment sequences could be specific to the type of immunotherapy (55). Interestingly, analysis of a randomized clinical trial (56) and a retrospective analysis of clinical data (57) suggest that patient outcome seems to be better if immunotherapy is given concurrently or begun soon after radiation, as compared with starting immunotherapy later after the radiation. This finding potentially reflects vascular normalization prompted by immunotherapy (53,54) and radiotherapy (39), which would initiate the feedback loop of vascular normalization and immune stimulation and thus potentiate both radiotherapy and immunotherapy. Mechanistically, the potential radiosensitizing function of immunotherapy would be effective only when it is administered concurrently with radiation or before radiation, to precondition the tumor microenvironment and thereby increase tumor radiosensitivity. Thus, synergistic effects of the two therapies may best be realized by alternating immunotherapy and radiotherapy, or vice versa. Whether this is true requires further preclinical and clinical investigations. Meanwhile, despite the favorablr antitumor effects in some patients, immunotherapy often induce adverse events and side effects, particular when combined with other treatment modalities such as radiation (58). Therefore, toxicity may be a hurdle for combining the two modalities, given the observed increase in adverse events from immunotherapy combined with radiation (59,60). The balance between benefits and risks needs further exploration in future clinical trials.

Optimizing the dose of radiation in combination with immunotherapy

The radiation dose and dosing schedule are other critical factors to consider (20) when given with immunotherapies. Conventionally fractionated radiation (i.e., once-daily 2-Gy fractions) mainly targets vulnerabilities in the tumor’s DNA damage repair and cell cycle arrest. Advances in radiotherapy techniques have enabled the delivery of large doses of radiation, also given daily, to similar or lower total doses in fewer fractions. The radiation dose per fraction not only affects direct cancer cell killing but also influences modulation of the tumor microenvironment. Compared with conventional fractionation, a single high dose of radiation may facilitate the maturation of antigen-presenting cells (61) and increase the infiltration of immune cells into the tumor (62). Low-dose radiation has been shown to normalize the tumor vasculature (39), but high-dose radiation could cause more vessel damage, thereby reducing blood flow and perfusion (63). Meanwhile, the lack of reoxygenation during the course of hypofractionated radiation therapy means that hypoxic tumors are more resistant to hypofractionated radiation (64). Theoretically, normalization of the tumor vasculature by immunotherapy may overcome the radioresistance of hypoxic tumors to hypofractionated radiation while maintaining its immune-stimulatory effects. However, the optimal dose of radiation to be given with immunotherapy is unknown, as some preclinical studies suggested that hypofractionated radiation led to a greater immune response (10,61,62) but others have found conventional fractionation to be more favorable when combined with immunotherapy (65,66). The intriguing interactions between immune cells and the tumor vasculature in response to different doses of radiation warrant further preclinical and clinical investigation.

Developing novel biomarkers of treatment response

Despite some remarkable successes in cancer immunotherapy, the response rates are still far from satisfying (67). Identification of biomarkers with which to predict and evaluate response to treatment is an unmet need for precision immunotherapy. Although several such markers have been tested, including DNA repair deficiencies (17,68), mutational burden (69), PDL1 expression (45), and the gut microbiome (70) have been tested, several limitations remain. Firstly, immunotherapy responses tend to emerge later than conventional therapies. In addition, tumor volume variations during immunotherapy do not necessarily reflect treatment responses (67,71). Thus novel markers are needed with which to evaluate responsiveness (or lack thereof) to immunotherapy early in the course of treatment, ideally even before tumor regression can be detected. Tumor-vessel normalization or increased perfusion are being explored as early markers of immunotherapy responsiveness (54). In a murine breast cancer model, tumor vessel perfusion, measured by 3D and color Doppler ultrasonography at 6 days after treatment with anti-CTLA4, indicated that high perfusion levels correlated strongly with tumor regression at day 14, with more than 90% sensitivity and specificity (54). Because tumor vessel perfusion can be visualized noninvasively by imaging early in the course of treatment, this may be a promising marker for predicting the efficacy of immunotherapy for a given patient (Figure 3). Whether this marker remains valid when radiation is added to the immunotherapy and whether it can be extended to clinical management require carefully designed exploratory and confirmatory studies of potential biomarkers for use in clinical trials.

Figure 3. Identification of novel biomarkers to guide radiation and immunotherapy treatment.

Figure 3.

Pretreatment markers may be useful for identifying patients who may benefit from immunotherapy. For tumors that are resistant or not completely eradiated by immunotherapy, consolidation therapy such as radiation may be used. Noninvasive means to evaluate vascular-related changes within a solid tumor at earlier time points post immunotherapy treatments may further help guide clinical decision-making for personalized cancer care. CT, computed tomography; MRI, magnetic resonance imaging.

Can immunotherapy modulate intrinsic radiosensitivity of tumors?

Several lines of evidence suggest that pathways related to radiosensitivity may also modulate a tumor’s immunogenicity and predict its response to immunotherapies. For example, deficiencies in the DNA repair machinery could increase neoantigen release and trigger an immune response (16), and responsiveness to immunotherapy could also be predicted by the presence of these DNA repair deficiencies (15,68). To date, several common regulators of DNA repair and immune checkpoints have been discovered. PARP inhibitors have well-studied radiosensitizing effects (72), and interestingly they also upregulate PDL1 expression in breast cancer cell lines and animal models (73). An siRNA screen by Sato et al. showed that BRCA2 or Ku70/80 deletion was associated with enhanced PD-L1 expression after x-ray radiation of cancer cells (44). Another important DNA repair regulator, p53, was shown to regulate PD-L1 expression through miR-34 (74). In patients with ovarian cancer, expression levels of PD-1 and PD-L1 were found to be associated with BRCA1/2 and p53 mutation status (75). Growing evidence supports the notion that DNA repair deficiencies modulate tumor immune checkpoints, but whether immune checkpoint blockade conversely regulates DNA repair pathways is still unclear. This potential novel mechanism by which immunotherapy regulates the intrinsic radiosensitivity of tumors may prove to be a promising avenue for future investigation.

Conclusion

In summary, in addition to the orthodox understanding that radiation promotes the efficacy of immunotherapy, immunotherapy may also radiosensitize tumors to achieve better local control when combined with radiotherapy. Immune checkpoint blockers can promote normalization of tumor blood vessels, improve tissue perfusion, and decrease intratumoral hypoxia in a T cell-dependent manner through IFN-mediated signaling between T cells and endothelial cells (53,54). These changes within the local tumor microenvironment subsequently may sensitize the tumor to ionizing radiation by reducing tissue hypoxia, decreasing acidosis and improving the production of reactive oxygen species needed for radiation-induced tumor cell killing (49,51,53,54). In depth understanding of the reciprocal relationship between ionizing radiation and immune re-programming thus has significant implications for designing future clinical trials of combination immunotherapies. For example, incorporation of new biomarkers that correlate with intratumoral vascularity, and how radiation should be given in terms of dose and timing with respect to immunotherapy becomes highly important, especially considering the transient nature of immunotherapy-induced vascular normalization effects. With cancer immunotherapies play an ever-increasing role in the treatment of solid tumors, taking advantage of the mutual benefit between ionizing radiation and immunotherapies may open up new avenues for designing immune combination treatments to achieve improved therapeutic efficacy with minimal toxicity in cancer patients.

Acknowledgements

This work was supported in part by the American Society of Clinical Oncology (ASCO) Conquer Cancer Foundation Young Investigator Award (10804; W.J.), the Cancer Prevention and Research Institute of Texas CPRIT (RR180017; W.J.), the Mayo Clinic Center for Regenerative Medicine (B.Y.S.K.), the Jorge and Leslie Bacardi fund for the study of Regenerative Medicine (B.Y.S.K.), the National Institute of Neurological Disorders and Stroke Grant (R01 NS104315; B.Y.S.K.), National Natural Science Foundation of China (NO. 81572500, Z.L.), and Hunan Young Talents (NO. 2016RS3036; Z.L.). The authors thank Christine Wogan of the Division of Radiation Oncology at MD Anderson Cancer Center for editorial assistance. The authors thank Jordan Pietz of Creative Services, MD Anderson Cancer Center, and Cailian Wen of Xmagine Studio (Shenzhen, China) for artistic assistance.

Footnotes

Conflict of Interest: The authors declare no conflict of interests

References

  • 1.Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. The New England journal of medicine 2012;366(10):925–31 doi 10.1056/NEJMoa1112824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer 2016;4:51 doi 10.1186/s40425-016-0156-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schoenhals JE, Seyedin SN, Tang C, Cortez MA, Niknam S, Tsouko E, et al. Preclinical Rationale and Clinical Considerations for Radiotherapy Plus Immunotherapy: Going Beyond Local Control. Cancer journal 2016;22(2):130–7 doi 10.1097/PPO.0000000000000181. [DOI] [PubMed] [Google Scholar]
  • 4.Haymaker CL, Kim D, Uemura M, Vence LM, Phillip A, McQuail N, 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(2):100–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immunotherapy: a beneficial liaison? Nature reviews Clinical oncology 2017;14(6):365–79 doi 10.1038/nrclinonc.2016.211. [DOI] [PubMed] [Google Scholar]
  • 6.Shi F, Wang X, Teng FF, Kong L, Yu JM. Abscopal effect of metastatic pancreatic cancer after local radiotherapy and granulocyte-macrophage colony-stimulating factor therapy. Cancer Biol Ther 2017;18(3):137–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 2015;16(7):795–803 doi 10.1016/S1470-2045(15)00054-6. [DOI] [PubMed] [Google Scholar]
  • 8.Lu K, He C, Guo N, Chan C, Ni K, Lan G, et al. Low-dose X-ray radiotherapy–radiodynamic therapy via nanoscale metal–organic frameworks enhances checkpoint blockade immunotherapy. Nature Biomedical Engineering 2018;2(8):600–10 doi 10.1038/s41551-018-0203-4. [DOI] [PubMed] [Google Scholar]
  • 9.Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015;520(7547):373–7 doi 10.1038/nature14292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Filatenkov A, Baker J, Mueller AM, Kenkel J, Ahn GO, Dutt S, et al. Ablative Tumor Radiation Can Change the Tumor Immune Cell Microenvironment to Induce Durable Complete Remissions. Clinical cancer research : an official journal of the American Association for Cancer Research 2015;21(16):3727–39 doi 10.1158/1078-0432.CCR-14-2824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 2006;203(5):1259–71 doi 10.1084/jem.20052494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol 2015;16(13):e498–509 doi 10.1016/S1470-2045(15)00007-8. [DOI] [PubMed] [Google Scholar]
  • 13.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646–74 doi 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 14.Kalbasi A, June CH, Haas N, Vapiwala N. Radiation and immunotherapy: a synergistic combination. The Journal of clinical investigation 2013;123(7):2756–63 doi 10.1172/JCI69219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mouw KW, Goldberg MS, Konstantinopoulos PA, D’Andrea AD. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer discovery 2017;7(7):675–93 doi 10.1158/2159-8290.CD-17-0226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Germano G, Lamba S, Rospo G, Barault L, Magri A, Maione F, et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017. doi 10.1038/nature24673. [DOI] [PubMed] [Google Scholar]
  • 17.Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357(6349):409–13 doi 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lomax ME, Folkes LK, O’Neill P. Biological consequences of radiation-induced DNA damage: relevance to radiotherapy. Clin Oncol (R Coll Radiol) 2013;25(10):578–85 doi 10.1016/j.clon.2013.06.007. [DOI] [PubMed] [Google Scholar]
  • 19.Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Advances in immunology 2000;74:181–273. [DOI] [PubMed] [Google Scholar]
  • 20.Wang Y, Deng W, Li N, Neri S, Sharma A, Jiang W, et al. Combining Immunotherapy and Radiotherapy for Cancer Treatment: Current Challenges and Future Directions. Frontiers in pharmacology 2018;9:185 doi 10.3389/fphar.2018.00185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Onishi M, Okonogi N, Oike T, Yoshimoto Y, Sato H, Suzuki Y, et al. High linear energy transfer carbon-ion irradiation increases the release of the immune mediator high mobility group box 1 from human cancer cells. J Radiat Res 2018;59(5):541–6 doi 10.1093/jrr/rry049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rudqvist NP, Pilones KA, Lhuillier C, Wennerberg E, Sidhom JW, Emerson RO, et al. Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells. Cancer Immunol Res 2018;6(2):139–50 doi 10.1158/2326-6066.CIR-17-0134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, et al. Stereotactic Radiation Therapy Augments Antigen-Specific PD-1-Mediated Antitumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol Res 2015;3(4):345–55 doi 10.1158/2326-6066.CIR-14-0196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Li T, Chen ZJ. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med 2018;215(5):1287–99 doi 10.1084/jem.20180139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Watson RO, Manzanillo PS, Cox JS. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell 2012;150(4):803–15 doi 10.1016/j.cell.2012.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Xia TL, Konno H, Barber GN. Recurrent Loss of STING Signaling in Melanoma Correlates with Susceptibility to Viral Oncolysis. Cancer Res 2016;76(22):6747–59. [DOI] [PubMed] [Google Scholar]
  • 27.Sun LJ, Wu JX, Du FH, Chen X, Chen ZJJ. Cyclic GMP-AMP Synthase Is a Cytosolic DNA Sensor That Activates the Type I Interferon Pathway. Science 2013;339(6121):786–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009;461(7265):788–92 doi 10.1038/nature08476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Dunphy G, Flannery SM, Almine JF, Connolly DJ, Paulus C, Jonsson KL, et al. Non-canonical Activation of the DNA Sensing Adaptor STING by ATM and IFI16 Mediates NF-kappaB Signaling after Nuclear DNA Damage. Mol Cell 2018;71(5):745–60 doi 10.1016/j.molcel.2018.07.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gonzalez-Navajas JM, Lee J, David M, Raz E. Immunomodulatory functions of type I interferons. Nature reviews Immunology 2012;12(2):125–35 doi 10.1038/nri3133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014;41(5):843–52 doi 10.1016/j.immuni.2014.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hou Y, Liang H, Rao E, Zheng W, Huang X, Deng L, et al. Non-canonical NF-kappaB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy. Immunity 2018;49(3):490–503 doi 10.1016/j.immuni.2018.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep 2015;11(7):1018–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Baird JR, Friedman D, Cottam B, Dubensky TW, Jr., Kanne DB, Bambina S, et al. Radiotherapy Combined with Novel STING-Targeting Oligonucleotides Results in Regression of Established Tumors. Cancer Res 2016;76(1):50–61 doi 10.1158/0008-5472.CAN-14-3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liang H, Deng L, Hou Y, Meng X, Huang X, Rao E, et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nature communications 2017;8(1):1736 doi 10.1038/s41467-017-01566-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lemos H, Mohamed E, Huang L, Ou R, Pacholczyk G, Arbab AS, et al. STING Promotes the Growth of Tumors Characterized by Low Antigenicity via IDO Activation. Cancer Res 2016;76(8):2076–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ahn J, Xia TL, Konno H, Konno K, Ruiz P, Barber GN. Inflammation-driven carcinogenesis is mediated through STING. Nature communications 2014;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jiang W, Chan CK, Weissman IL, Kim BYS, Hahn SM. Immune Priming of the Tumor Microenvironment by Radiation. Trends in cancer 2016;2(11):638–45 doi 10.1016/j.trecan.2016.09.007. [DOI] [PubMed] [Google Scholar]
  • 39.Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer cell 2013;24(5):589–602 doi 10.1016/j.ccr.2013.09.014. [DOI] [PubMed] [Google Scholar]
  • 40.Thomas DA, Massague J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer cell 2005;8(5):369–80. [DOI] [PubMed] [Google Scholar]
  • 41.Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med 2001;7(10):1118–22. [DOI] [PubMed] [Google Scholar]
  • 42.Pu N, Zhao G, Gao S, Cui Y, Xu Y, Lv Y, et al. Neutralizing TGF-beta promotes anti-tumor immunity of dendritic cells against pancreatic cancer by regulating T lymphocytes. Cent Eur J Immunol 2018;43(2):123–31 doi 10.5114/ceji.2018.77381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, et al. TGF beta Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res 2015;75(11):2232–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sato H, Niimi A, Yasuhara T, Permata TBM, Hagiwara Y, Isono M, et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nature communications 2017;8(1):1751 doi 10.1038/s41467-017-01883-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515(7528):563–7 doi 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Park JS, Kim IK, Han S, Park I, Kim C, Bae J, et al. Normalization of Tumor Vessels by Tie2 Activation and Ang2 Inhibition Enhances Drug Delivery and Produces a Favorable Tumor Microenvironment. Cancer cell 2016;30(6):953–67 doi 10.1016/j.ccell.2016.10.018. [DOI] [PubMed] [Google Scholar]
  • 47.Huang Y, Kim BYS, Chan CK, Hahn SM, Weissman IL, Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nature reviews Immunology 2018;18(3):195–203 doi 10.1038/nri.2017.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rockwell S, Dobrucki IT, Kim EY, Marrison ST, Vu VT. Hypoxia and radiation therapy: past history, ongoing research, and future promise. Current molecular medicine 2009;9(4):442–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Meijer TW, Kaanders JH, Span PN, Bussink J. Targeting hypoxia, HIF-1, and tumor glucose metabolism to improve radiotherapy efficacy. Clinical cancer research : an official journal of the American Association for Cancer Research 2012;18(20):5585–94 doi 10.1158/1078-0432.CCR-12-0858. [DOI] [PubMed] [Google Scholar]
  • 50.Carlson DJ, Yenice KM, Orton CG. Tumor hypoxia is an important mechanism of radioresistance in hypofractionated radiotherapy and must be considered in the treatment planning process. Medical physics 2011;38(12):6347–50 doi 10.1118/1.3639137. [DOI] [PubMed] [Google Scholar]
  • 51.Scott KE, Cleveland JL. Lactate Wreaks Havoc on Tumor-Infiltrating T and NK Cells. Cell Metab 2016;24(5):649–50 doi 10.1016/j.cmet.2016.10.015. [DOI] [PubMed] [Google Scholar]
  • 52.Lee DC, Sohn HA, Park ZY, Oh S, Kang YK, Lee KM, et al. A lactate-induced response to hypoxia. Cell 2015;161(3):595–609 doi 10.1016/j.cell.2015.03.011. [DOI] [PubMed] [Google Scholar]
  • 53.Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 2017;544(7649):250–4 doi 10.1038/nature21724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zheng X, Fang Z, Liu X, Deng S, Zhou P, Wang X, et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. The Journal of clinical investigation 2018;128(5):2104–15 doi 10.1172/JCI96582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, et al. Optimizing Timing of Immunotherapy Improves Control of Tumors by Hypofractionated Radiation Therapy. PloS one 2016;11(6):e0157164 doi 10.1371/journal.pone.0157164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. The New England journal of medicine 2017. doi 10.1056/NEJMoa1709937. [DOI] [Google Scholar]
  • 57.Samstein R, Rimner A, Barker CA, Yamada Y. Combined Immune Checkpoint Blockade and Radiation Therapy: Timing and Dose Fractionation Associated with Greatest Survival Duration Among Over 750 Treated Patients. International Journal of Radiation Oncology*Biology*Physics 2017;99(2, Supplement):S129–S30 doi 10.1016/j.ijrobp.2017.06.303. [DOI] [Google Scholar]
  • 58.Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. The New England journal of medicine 2018;378(2):158–68 doi 10.1056/NEJMra1703481. [DOI] [PubMed] [Google Scholar]
  • 59.Martin AM, Cagney DN, Catalano PJ, Alexander BM, Redig AJ, Schoenfeld JD, et al. Immunotherapy and Symptomatic Radiation Necrosis in Patients With Brain Metastases Treated With Stereotactic Radiation. JAMA oncology 2018. doi 10.1001/jamaoncol.2017.3993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Tang C, Jiang W, Yap TA. Efficacy and Toxic Effects of Cancer Immunotherapy Combinations-A Double-Edged Sword. JAMA oncology 2018. doi 10.1001/jamaoncol.2017.4606. [DOI] [PubMed] [Google Scholar]
  • 61.Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 2009;114(3):589–95 doi 10.1182/blood-2009-02-206870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.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(12):7516–23. [DOI] [PubMed] [Google Scholar]
  • 63.Kim MS, Kim W, Park IH, Kim HJ, Lee E, Jung JH, et al. Radiobiological mechanisms of stereotactic body radiation therapy and stereotactic radiation surgery. Radiation oncology journal 2015;33(4):265–75 doi 10.3857/roj.2015.33.4.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.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. International journal of radiation oncology, biology, physics 2011;79(4):1188–95 doi 10.1016/j.ijrobp.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nature communications 2017;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Aryankalayil MJ, Makinde AY, Gameiro SR, Hodge JW, Rivera-Solis PP, Palayoor ST, et al. Defining Molecular Signature of Pro-Immunogenic Radiotherapy Targets in Human Prostate Cancer Cells. Radiat Res 2014;182(2):139–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nature reviews Clinical oncology 2017. doi 10.1038/nrclinonc.2017.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine 2015;372(26):2509–20 doi 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Khagi Y, Goodman AM, Daniels GA, Patel SP, Sacco AG, Randall JM, et al. Hypermutated Circulating Tumor DNA: Correlation with Response to Checkpoint Inhibitor-Based Immunotherapy. Clinical cancer research : an official journal of the American Association for Cancer Research 2017;23(19):5729–36 doi 10.1158/1078-0432.CCR-17-1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2017. doi 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid O, Lebbe C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clinical cancer research : an official journal of the American Association for Cancer Research 2009;15(23):7412–20 doi 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
  • 72.Alotaibi M, Sharma K, Saleh T, Povirk LF, Hendrickson EA, Gewirtz DA. Radiosensitization by PARP Inhibition in DNA Repair Proficient and Deficient Tumor Cells: Proliferative Recovery in Senescent Cells. Radiat Res 2016;185(3):229–45 doi 10.1667/RR14202.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jiao S, Xia W, Yamaguchi H, Wei Y, Chen MK, Hsu JM, et al. PARP Inhibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clinical cancer research : an official journal of the American Association for Cancer Research 2017;23(14):3711–20 doi 10.1158/1078-0432.CCR-16-3215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, et al. PDL1 Regulation by p53 via miR-34. Journal of the National Cancer Institute 2016;108(1) doi 10.1093/jnci/djv303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wieser V, Gaugg I, Fleischer M, Shivalingaiah G, Wenzel S, Sprung S, et al. BRCA1/2 and TP53 mutation status associates with PD-1 and PD-L1 expression in ovarian cancer. Oncotarget 2018;9(25):17501–11 doi 10.18632/oncotarget.24770. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES