Abstract
Immunotherapy combinations are being investigated to expand the benefit of immune checkpoint blockade across many cancer types. Radiation combinations, in particular using stereotactic body radiotherapy, are of keen interest because of underlying mechanistic rationale, safety, and availability as a standard of care in certain cancers. In addition to direct tumor cytotoxicity, radiation therapy has immunomodulatory effects such as induction of immunogenic cell death, enhancement of antigen presentation, and expansion of the T-cell receptor repertoire as well as recruitment and increased activity of tumor-specific effector CD8+ cells. Combinations of radiation with cytokines and/or chemokines and anti-programmed death 1 and anticytotoxic T-lymphocyte antigen 4 therapies have demonstrated safety and feasibility, as well as the potential to improve long-term outcomes and possibly induce out of irradiated field or abscopal responses. Novel immunoradiotherapy combinations represent a promising therapeutic approach to overcome radioresistance and further enhance systemic immunotherapy. Potential benefits include reversing CD8+ T-cell exhaustion, inhibiting myeloid-derived suppressor cells, and reversing M2 macrophage polarization as well as decreasing levels of colony-stimulating factor-1 and transforming growth factor-β. Here, we discuss current data and mechanistic rationale for combining novel immunotherapy agents with radiation therapy.
Immune checkpoint blockade (ICB), with anti-programmed death 1 (PD1) receptor or anti-PD ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte antigen 4 (CTLA4) antibodies, have improved clinical outcomes across several cancer types (1). However, many patients still do not derive clinical benefit and eventually have disease progression. Thus, improving the efficacy of ICB is an ongoing unmet clinical need.
One promising strategy involves combining radiation therapy (RT) with immune-oncology (IO) agents to improve responses, potentially in a synergistic manner. This strategy is particularly attractive as RT is standard of care in many malignancies, allowing for a greater ease of study in clinical trials (2-5). Indeed, prior to the widespread use of ICB, clinical trials combining cytokines such as interleukin-2 (IL-2) and chemokines such as granulocyte-macrophage colony-stimulating factor (GMCSF) with high-dose RT suggested feasibility for IO-RT approaches, with preliminary evidence for improved treatment outcomes (6-9). In this article we discuss characteristics of RT such as dose, schedule, and delivery mode in relation to its immunomodulatory effects, review safety, and efficacy results of existing IO-RT clinical trials and propose novel IO-RT combinations based on mechanistic rationale of IO to overcome RT-induced immunosuppression and further enhance immunostimulant effects.
Radiation characteristics and potential impact on immunotherapy efficacy
In the majority of disease settings, the current standard, curative RT treatment involves small, fractionated doses of RT (generally 1.8-2.2 Gy) given daily to cumulative doses of 50-70 Gy depending on clinical context (Table 1). This schedule is used because of the necessity of treating both the primary tumor and commonly at-risk draining lymphatics while limiting normal tissues toxicity. This approach, although leading to cure in many patients, may be disadvantageous when used in immunotherapy combinations because of a potential immunosuppressive effect (10-12). Indeed, recent data suggest that elective nodal RT may be antagonistic to immunotherapy, at least in preclinical models (13).
Table 1.
Radiation characteristics and potential impact on immunotherapy efficacya
Radiation type | Description | Immunomodulatory effect |
---|---|---|
Conventional |
|
|
|
|
|
Proton radiation treatment |
|
|
Carbon ion radiotherapy |
|
Immunogenic cell death |
|
|
|
FLASH-RT |
|
|
CTL = cytotoxic T-lymphocyte infiltration; IFN = interferon; RT = radiation therapy; SABR = stereotactic ablative radiation therapy; SBRT = stereotactic body radiation therapy; SBRT-PATHY = stereotactic body radiation therapy for Partial tumor irradiation of unresectable bulky tumors targeting exclusively their hypoxic segment; SFRT = spatially fractionated radiation therapy (also known as GRID therapy); SRS = stereotactic radiosurgery; TNA = tumor necrosis factor; TRAIL = TNF-related apoptosis-inducing ligand.
Conversely, approaches treating small volumes to high doses over few treatments—referred to by several names such as hypofractionated RT, stereotactic body RT (SBRT), stereotactic ablative RT (SABR), or stereotactic radiosurgery—are likely associated with immune-stimulatory effects. For example, in murine preclinical models, high-dose RT has been shown to increase tumor-associated macrophages (TAM) and CD8+ T cells and enhance expression of T-cell activation and interferon (IFN)-γ–associated gene expression (14-16).
These distinct and opposed outcomes following the combination of RT and immunotherapy based on the type of RT given (eg, conventional vs hypofractionated RT) complicate the inclusion of immunotherapy into the curative management of many solid tumors. Indeed, at the time of this publication, the only randomized clinical trial of combined conventionally fractionated chemoRT and immunotherapy that improved survival is the PACIFIC trial in non-small cell lung cancer (NSCLC), which held immunotherapy until the completion of RT (17-19). This approach would seem at least to partially avoid the immunosuppressive effects of conventionally fractionated RT while reaping some of the immune-stimulatory effects of tumor cell death due to RT. Similar trials that gave immunotherapy concurrent with RT have had no beneficial effects on outcome (20-22). Ongoing trials will determine the short- and intermediate-term sequencing of current definitive conventionally fractionated RT and immunotherapy for clinical use.
In the metastatic setting, where RT may be an effective immune adjunct as opposed to a curative therapy, utilizing hypofractionated regimens is likely a better approach. In addition to its immune-stimulatory effects, hypofractionated RT has the added benefit of potentially ablating 1 or more larger tumors, which are typically more resistant to immune-checkpoint inhibitors (ICI) with a minimal added toxicity (23). Increased tumor bulk has been associated with poor response to anti-PD1 (24-26), whereas the tumor microenvironment in large tumors is associated with increased infiltration of myeloid lineage cells, systemic immunosuppressive cytokines, and regulatory T-cell (Treg) infiltration (27-29).
There are several aspects of RT delivery that impact the interaction between RT and IO beyond dose, schedule, and combination with specific IO agents. These include heavy particle therapy such as proton, carbon ion, and π meson and spatially fractionated radiotherapy (SFRT) such as SFRT (GRID therapy) and SBRT PArtial Tumor irRT of unresectable bulky tumors targeting exclusively their HYpoxic segment (SBRT-PATHY), as well as an emerging paradigm of understanding the effect of dose rates with FLASH radiotherapy.
Photon-based RT treatment utilizes x-ray and γ-rays that deposit energy to tissue that causes single- and double-stranded DNA breaks to damage tumor cell DNA. Should enough DNA strand breaks occur in key regions of the genome, RT damage ultimately results in mitotic catastrophe and tumor cell death. The deposition of this energy is characterized by linear energy transfer (LET) or the energy transfer per unit length of the track of ionizing RT. Heavier ionizing particles have a greater chance to interact and transfer energy with the surrounding tissue. These heavier ionizing particles such as protons, carbon ion, and π meson are considered densely ionizing RT with a high LET. Lighter particles such x-ray and γ-rays classically are low LET and are considered sparsely ionizing RT. To properly compare the biological effects of each form of ionizing RT, we use the relative biologic effectiveness (RBE), defined as the ratio of a normalized dose of x-ray RT to a test dose (proton, carbon ion, etc) thus allowing one-to-one comparisons of the effects of different forms of RT treatment. Larger ion particles tend to have a higher RBE compared with traditional RT treatment. RBE is affected by LET, dose, fraction, dose rate, and biological system (30,31).
Proton RT treatment has unique properties that render it intriguing for pairing with IO. It has a higher LET and slightly higher RBE of 1.1 compared to photon treatment. It has unique properties in which the majority of its ionizing RT is given off at a specific location known as the Bragg peak, after which energy deposition promptly drops off and RT dose to deeper tissue is largely spared. One of the main reasons for excitement for proton therapy was the utilization of this Bragg peak phenomena to limit dose deposition to normal tissue. There are some reports of surprising results of increased tissue toxicity.
Some preclinical evidence suggests a difference in affecting tumors and tumor microenvironment with proton therapy. In vitro experiments suggested higher cell surface levels for the damage-associated molecular pattern (DAMP) molecule calreticulin, which might lead to increased immunogenicity and recognition by CTLs (32,33). Additionally, the lower dose bath of RT from proton therapy decreases the time and/or level of exposure of lymphocytes to RT ultimately decreasing the level of lymphopenia (34,35). Other studies simply suggest in vitro that proton therapy can produce unique antitumor responses, albeit this study did not directly compare proton with photon effects (36).
Carbon ion radiotherapy is another form of particle RT treatment that is not widely used today given the current expense and extremely large size facility needed to operate this. Carbon ions operate under the similar principles for proton therapy, however, the RBE is even higher than protons likely because of more dose deposition due to particle fragmentation into the surrounding tissue (37). Interestingly, some preclinical models with carbon ion radiotherapy suggest an enhancement of immune responsiveness to combinatorial RT and immunotherapy via enhanced immunologic cell death and increased tumor immunogenicity (38).
Pion therapy (negatively charged π mesons) is another modality that was previously explored because of its Bragg peak and increased effectiveness in hypoxic environments (39). Pions are heavy ions that are produced in cyclotrons or linear accelerators using protons to hit a beryllium target. Pion therapy Bragg peak is higher than the other heavy particles because of its negative charge being attracted to nucleus in materials resulting ultimately into nuclear fragmentation, alpha particle production, and protons and neutrons, which all interact with tissue. Pion therapy is not widely used because of cost and availability as well as the loss of conformality to tumor targets because of increased lateral scatter and worse penumbra due to the other charged particles it produces. Two smaller clinical trials compared overall survival (OS) in prostate and high-grade astrocytoma, however, pion therapy improves OS compared with conventional photon therapy (40,41),
The role of partial-tumor RT is being currently explored and may hold specific promise for RT-IO combinations. Two techniques are currently being used: SFRT, also known as GRID therapy, and SBRT-PATHY. The SFRT principle is to deliver large, heterogenous doses to large, bulky tumors utilizing a grid of pencil beams dose distribution to the tumor. The premise of GRID therapy relies on sparing some portions of the overlying skin to reduce the risk of skin necrosis. Originally, a metallic block with small beam holes was used to create this treatment pattern, but with modern techniques, this can be recreated with standard multileaf collimator blocks in contemporary radiotherapy machines. GRID traditionally is used for palliative purposes typically with photons, although proton and carbon ion therapy have been used as well. The excitement with SFRT for RT-IO combinations is that it has been shown to induce bystander effect to kill tumor cells in the lower dose region adjacent to the high-dose pencil beam areas, which is not typically seen in conventional fractionated treatment. This bystander effect might be mediated by induction of cytokines such as tumor necrosis factor alpha (TNA-α) and TNF-related apoptosis-inducing ligand (TRAIL), destruction of tumor microvasculature endothelium by induction of cermamide, or induction of immune response leading to abscopal effects, seen mainly in preclinical animal models in nonirradiated tumors (42). SBRT-PATHY takes the inhomogeneity concept and applies it to certain areas of the tumor microenvironment. SBRT-PATHY specifically treats only part of a large, bulky unresectable tumor and targets hypoxic areas of the tumor microenvironment. These hypoxic cores of tumors tend to be more radioresistant to conventional RT treatment, thus SBRT-PATHY is an attractive treatment that may trigger bystander and abscopal effects on tumors (42).
Another exciting technology or technique that may lend itself useful to the combinatorial IO-RT treatment paradigm is FLASH radiotherapy (FLASH-RT). FLASH-RT delivers a standard dose of RT treatment in a matter of milliseconds, a dramatic reduction from the typical length measured in minutes. In the 1960s, FLASH-RT showed increased normal cell survival in vivo when compared with more typical lower dose rates (43). A few interesting theories exist as to why normal tissue seems to be spared. The first is that FLASH-RT might cause the immediate consumption of oxygen in the environment, and thus normal tissue becomes hypoxic, and RT damage is most optimal in an oxygenated environment, therefore normal tissue is not as easily damaged. Tumors that are already hypoxic are not affected by this sudden change of oxygenation. The second theory that is of greater interest here is that by delivering dose so rapidly, circulating lymphocytes are no longer irradiated, and participants do not become lymphopenic. Additionally, FLASH may cause improved CTL infiltration into the tumor microenvironment, less chronic neuroinflammation, and differential expression of cytokines (43). Most of our knowledge with FLASH comes from preclinical animal models; however, in the past few years, the first patient, who had multiply recurrent CTCLs, was treated with FLASH therapy (44). Results were excellent with limited skin toxicity. It was noted that there was an increase in edema, which is speculated to be due to a bigger immune-mediated response.
In summary, the impact of these techniques or modalities on RT-IO interactions beyond conventional photon-based RT approaches is the subject of ongoing investigations, and despite great enthusiasm, definitive conclusions on the contributions of specific RT parameters remain largely uncertain. Only systematic and rigorous analysis of the different aspects of RT will help investigators identify the optimal RT approaches to pair with IO.
Immunomodulatory effects of RT
Immunostimulant effects of RT
Beyond direct cytocidal effect in large—and potentially ICI-resistant—tumors, hypofractionated RT has immunomodulatory effects including but not limited to 1) induction of immunogenic cell death (ICD), 2) enhancement of antigen presentation by dendritic cells (DCs), 3) expansion of the T-cell receptor (TCR) repertoire, and 4) recruitment and increased activity of tumor-specific effector CD8+ cells (45-48). The process of ICD includes a series of steps that can induce antitumor immunity and collectively shape RT-induced responses (49,50). After RT, ICD includes the release of adenosine triphosphate and high mobility group box 1 protein, which binds to toll-like receptor (TLR) 4 on DCs, and translocation of calreticulin to the cell surface. Calreticulin then serves as a potent prophagocytic signal (51,52). Potential immunostimulant effects of RT are summarized in Figure 1.
Figure 1.
Immunostimulant and immunosuppressant effects of radiation therapy (RT). The figure illustrates the mechanisms underlying immunostimulant effects of radiation therapy, which include enhancement of tumor-associated antigen (TAA) shedding, immunogenic cell death, increase in TAA presentation to antigen-presenting cells, major histocompatibility complex (MHC) class I expression, increase in expression of NKG2D ligand, upregulation of adhesion molecules. The mechanisms underlying immunosuppressant effects include TGF-beta signaling activation, M2 macrophage polarization, increase in infiltration of myeloid-derived suppressor cell infiltration, activation of colony-stimulating factor-1 (CSF1) pathway, hypoxia inducible factor 1 (HIF1), T-regulatory cell (Treg) recruitment, programmed-death ligand-1 (PD-L1) upregulation, and vascular endothelial growth factor (VEGF) upregulation.
Because of its immunomodulatory effects, hypofractionated RT may induce responses in nonirradiated lesions, a phenomenon known as the abscopal effect. Initially observed preclinically as early as 1953, isolated case reports of this abscopal effect pepper the RT literature but are rarely observed in clinical practice as their existence has been questioned (53).
Favorable initial data for abscopal effects were observed in initial studies combining RT with GM-CSF (8), however, follow-up trials have shown mixed results for abscopal or distant effects following the combination of hypofractionated RT with ICI, underscoring the lack of understanding of this phenomenon (54-57). Elucidating any distant immunomodulatory effects of hypofractionated RT is even more critical in light of several trials showing the survival benefit of consolidation hypofractionated RT to multiple oligometastatic sites (48,58-60).
Immunosuppressant effects of RT
In addition to the potentially immunosuppressant effects of RT targeting lymphocyte-producing organs, and circulating lymphocytes, intratumoral immunosuppression by RT has been described as well, particularly following conventionally fractionated RT. These effects can include 1) promoting MDSCs infiltration (61), 2) enhancing CSF1 gene expression and CSF1 levels in tumor (62), 3) increasing tissue growth factor (TGF)-β (63) and Tregs (64), 4) upregulating hypoxia-inducible factors (HIF) and vascular endothelial growth factor (VEGF) (65), upregulating PD-L1 expression (66,67), and M2 macrophage polarization (Figure 1) (68). Targeting each of those pathways can be promising to improve response to RT and expand the role of RT as a therapeutic approach in patients with metastatic cancer. Here we will discuss current data and mechanistic rationale of combining novel immunotherapy agents with RT.
Optimal approach to combine immunotherapy and RT
Many unanswered questions remain regarding the optimal approach to combine IO with RT (Table 2). Firstly, the optimal timing and sequencing of RT and immunotherapy is unknown. It is established that responses following the combination of PD-L1 monoclonal antibody (mAb) and hypofractionated RT vary dramatically in preclinical models based on the timing of either treatment in relation to the other (66,69,70). This importance is underscored, albeit in the conventionally fractionated setting, based on the aforementioned positive clinical trial for ICI consolidation after chemoRT vs several negative concurrent RT-ICI trials (17-20). With appropriate caveats, these data provide tantalizing clinical insight highlighting the potential importance of RT and ICI sequencing.
Table 2.
Optimal approach to combine immunotherapy and radiation therapya
Key question | Approach and considerations | Supportive findings |
---|---|---|
Sequencing immunotherapy with RT | Concurrent vs sequential | Sequential
|
Dose and fractionation of RT | Single vs multiple fractions | Fractionated RT
|
Number of lesions for RT | Single vs multisite RT | Multisite RT
|
Irradiated tumor location | Clinically relevant and/or large tumor vs targeting and/or not targeting specific location | None to TDLN
|
Ab = antibody; CRT = chemoradiation therapy; CTLA4 = cytotoxic T-lymphocyte antigen 4; HN = head and neck; IFN = interferon; OS = overall survival; RT = radiation therapy; TDLN = tumor-draining lymph node; TME = tumor microenvironment; TREX1 = Three prime repair exonuclease 1.
Secondly, the optimal dose and fractionation of RT as an ICI adjunct is worthy of further study. At least one study showed that a fractionated regimen (8 Gy x 3 fractions [fx]) vs a single fraction of 30 Gy resulted in upregulation of type I IFN genes mediated by stimulator of IFN genes (STING) pathway activation and induced abscopal responses in TSA breast and MCA38 colon mice models (71,72). Single RT dose above 12 Gy induced activation of 3 prime repair exonuclease 1 (TREX1), which decreased cytosolic DNA level aborting activation of STING pathway, nominating TREX1 as a key playing factor in RT effect (73). However, regimens such as that pursued in NRG-BR001 recommended anatomically driven doses of 30-50 Gy in 3-5 fractions resulted in induction of IFN-linked gene expression, exemplified by Deoxyribonuclease I (DNASE1), despite induction of DNASEIII (TREX1) (57,74).
Thirdly, the optimal number of tumor lesions to target via RT is unclear. As mentioned above, targeting multiple oligometastatic sites with RT has been associated with improved survival in several small trials. However, the majority of IO-RT trials performed to date have pursued single-site irradiation approaches with the goal of inducing abscopal responses (75-77). Moreover, in at least 1 trial, OS of patients receiving RT-IO was correlated with irradiated tumor response (74). This highlights the potential importance of multisite irradiation, as targeting several tumor lesions may further alter the local tumor immunosuppressive microenvironment.
Fourthly, the unique role of irradiated tumor location in improving IO-RT combination efficacy is an area of investigation. In most RT-IO trials, when choosing metastatic sites to irradiate, preference has been given to the most clinically relevant and/or larger lesions, although there are reports about the differential immunogenic potential of irradiated sites in enhancing peripheral CD8+ T-cell response (76). Another consideration for metastatic site targeting is the sparing of lymphocyte-producing organs to prevent RT-induced lymphopenia (78). The irradiation to tumor-draining lymph nodes (TDLN) may create local immunosuppressive microenvironment with the effect being dependent on RT dose (13,79,80,81). In the MC38 murine colon carcinoma model, elective irradiation to TDLN downregulated CXCR3-CXCR5–associated T-cell chemoattractant chemokines resulting in decreased effector CD8 T cells in lymph node and reduced efficacy of IO-RT (80). RT to TDLN reduced abscopal effect and decreased count of stem-like and total CD8 T cells in B16 murine melanoma model (81). Elective nodal irradiation led to a decrease in antigen-specific T cells in tumor microenvironment of Head and Neck (HN) cancer model and resulted in low numbers of circulating T lymphocytes supporting further the strategy to spear lymph node (LN) irradiation (82). However, prospective clinical data regarding beneficial effect of TDLN-sparing RT are lacking.
Prior clinical results of RT with anti-PD1 and/or anti-CTLA4 therapy
There is a strong scientific rationale to combine RT with ICI therapy not only to enhance the therapeutic effect of irradiation and immunotherapy alone but also to overcome the resistance to each therapy individually. Increased PD-L1 expression and T-cell exhaustion are established resistance mechanisms to RT, which might be overcome by combining RT with anti-PD1 or anti–PD-L1 therapies (83). In human papilloma virus (HPV)–negative head and neck squamous cell cancer (HNSCC), upregulation of PD-L1 was associated with Axl-PI3K/Akt expression and led to locoregional failure for patients after RT (84).
Safety results of RT with anti-PD1 and/or anti-CTLA4 therapy
Despite conceptual promise, there were early clinical concerns surrounding the potential safety of RT combined with ICI given the possibility of overlapping toxicities. The safety profile may vary between using combination of anti-PD1 and/or L1 vs anti-CTLA4 and/or anti-PD1/L1 plus anti-CTLA4 with PD1/-L1 targeting appearing to be associated with fewer adverse events. The central nervous system and the possibility of treatment-associated brain necrosis (TABN) and/or radionecrosis is a particularly important anatomic consideration for IO-RT combination toxicity. The incidence of TABN is reported to be approximately 15%-30% with studies reporting association of IO with hypofractionated RT with both radiological and clinical TABN (85-88). The association was especially strong in patients with melanoma (85). The incidence of TABN observed in 2 single-institutional studies was 13.5%-15% in melanoma patients. In contrast, another group reported the association between immunotherapy and TABN when combined with stereotactic radiosurgery in patients with melanoma (hazard ratio [HR] = 4.02, 95% confidence interval [CI]= 1.17 to 13.82; P = .03) (85).
Anti-CTLA4
In a retrospective, single-center study of patients with metastatic melanoma, the combination of ipilimumab administered before fractionated RT resulted in 18% immune-related adverse events, which required holding or delaying ipilimumab administration (89). In patients with metastatic castrate-resistant prostate cancer, bone-directed RT at the dose of 8 Gy in 1 fraction followed by ipilimumab resulted in high-grade AEs in 26% of patients (75).
Anti-PD1 and/or L1
In a safety analysis of combining anti-PD1 therapy with RT, one study reported a drug limiting toxicity (DLT) rate of less than 10% with using a multisite hypofractionated RT approach (57), whereas combination of single-site hypofractionated RT and pembrolizumab resulted in no DLTs (90). In the former study, all patients with toxicities had at least 2 irradiated lesions. The use of concurrent avelumab with chemoRT resulted in G3 and higher treatment-related AEs in approximately one-third of patients, which was similar to chemoRT alone (20). Finally, the most robust analysis to date to address this issue was the recent pooled analysis of patients enrolled in prospective clinical trials of ICI (anti-PD1, anti–PD-L1, CTLA4) with RT within 90 days of initiation of ICI. The study revealed no increased rate of G3 and higher treatment-related AEs in comparison with those who received immunotherapy alone (91).
Concurrent vs sequential
Another important consideration for safety evaluation is whether IO was given concurrently with hypofractionated RT or sequentially after RT. The randomized phase I trial of Concurrent or Sequential Ipilimumab, Nivolumab and Stereotactic Body Radiotherapy in Patients with Stage IV NSCLC Study (COSINR) study combined anti-PD1 with anti-CTLA4 therapy with hypofractionated RT randomized to concurrent and sequential multisite SBRT. In the sequential arm, the combination of all 3 anti-CTLA4 with anti-PD1 and RT resulted in increased risk of pneumonitis and prompted dose de-escalation of hypofractionated RT in patients with NSCLC during central lung radiation (92). There were no DLTs observed in the concurrent arm supporting the concept of safe initiation of IO during RT. The US Food and Drug Administration database pooled analysis of clinical trials combining ICB with RT revealed that ICB therapy within 90 days of RT was not associated with an increased risk of serious adverse events.
In summary, anti-PD1 and/or anti-CTLA4 and RT combination approach was found to be generally safe in terms of immune-related adverse events with only minor concern for increased incidence of pneumonitis with only anti-PD1 with anti-CTLA4 and RT combination and radionecrosis especially among patients with metastatic melanoma (86,92).
Efficacy results of RT with anti-PD1 and/or anti-CTLA4 therapy
The anti-CTLA4 and/or anti-PD1 or L1 plus RT trials provided informative data about sequencing of IO and RT, preferential anatomic site of RT, and IO-RT–induced immune profile changes.
Sequencing IO and RT
The efficacy of sequential vs concurrent IO-RT has not been formally evaluated and compared in large clinical trials, and it remains unknown whether one is more beneficial over another. It is noteworthy to mention that individual clinical trial data may support sequential over concurrent IO-RT in terms of efficacy endpoint. In patients with metastatic liver and lung (excluding melanoma) advanced tumors, therapy with ipilimumab and sequential or concurrent SBRT (total 50 Gy in 4 fx or total 60 Gy in 10 fx) resulted in a clinical benefit rate of 26% and reached to 42% in patients who received sequential RT and irradiation to lung lesions. There was a trend toward improved responses while using sequential as opposed to concurrent RT as well as radiating lung as opposed to liver lesions (55). ECOG-ACRIN is an ongoing randomized phase III trial that aims to evaluate concomitant vs sequential durvalumab with chemoRT for unresectable stage III NSCLC (93). Concurrent avelumab with chemoRT vs chemoRT alone failed to improve PFS in patients with locally advanced HNSCC cancers. In fact, the hazard ratio was equal to 1.21, raising the question about proper combination strategy of IO and chemoRT and the hypothesis that concurrent design may have impacted the beneficial effect of adding avelumab (20). In the cohort of patients with intermediate risk HPV-positive and locally advanced HPV-negative HNSCC, sequential pembrolizumab with cisplatin and RT resulted in numerically higher 1- and 2-year progression-free survival (PFS) in comparison with the concurrent approach (94). Pooled analysis of patients with NSCLC treated with pembrolizumab and RT revealed statistically significant improvement in abscopal response rate (odds ratio [OR] = 2.96; P = .0039), median PFS (HR = 0.67; P = .045), and OS (HR = 0.67; P = .0004) in combination of pembrolizumab with RT vs RT alone (95).
IO-RT–induced immune profile changes
A growing body of evidence supports IO-RT combinations as inducing improved immune responses, and several of those changes may be linked to clinical benefit. In patients with metastatic castrate-resistant prostate cancer, ipilimumab with RT (8 Gy x 1) resulted in increased peripheral blood CD8+ T cells, and high ratio of CD8+ to CD4+ T-cell ratio was associated with improved clinical benefit (75). In patients with chemotherapy-resistant NSCLC, concurrent RT (total 30 Gy in 5 fx or total 27 Gy in 3 fx) with ipilimumab resulted in expansion of TCR clones. In addition, increases in IFN-β were associated with response to therapy, whereas baseline tumor PD-L1 expression, CD8+ T-cell infiltration, and increased proliferation of peripheral blood CD4+CD8+ cells were not (96). Pembrolizumab combined with multisite SBRT (total 30-50 Gy in 3-5 fx) resulted in increased expression of IFN-γ–linked genes as well as a decrease in those associated with the cell cycle and DNA repair (57). Single-site RT regimens (total 24 Gy in 3 fx or 17 Gyx1 fx) with pembrolizumab resulted in increase in Ki67+ PD1+ CTLA+CD8 T cells in peripheral blood mononuclear cells (PBMCs) (90,97). Concurrent RT (total 30 Gy in 10 fx or total 27 Gy in 3 fx) resulted in an increase in peripheral blood TCR diversity in patients with advanced melanoma (98).
Does IO-RT improve response rates and/or PFS/OS over IO or RT alone?
Even though above-mentioned trials demonstrated evidence of enhanced immune responses after IO-RT, these changes have not translated into consistent improvement in efficacy. In a phase III trial of patients with metastatic castrate-resistant prostate cancer who progressed on docetaxel, the addition of ipilimumab to RT (8 Gyx1) did not improve OS in comparison to placebo (75). In a phase II randomized trial of patients with advanced adenoid cystic carcinoma, the addition of RT (total 30 Gy in 5 fx) to pembrolizumab did not improve PFS in comparison with pembrolizumab alone (99). In a phase II trial of patients with metastatic renal cell carcinoma (RCC) who progressed on prior anti-angiogenic therapies combination of nivolumab with RT (total 30 Gy in 3 fx) resulted in an objective response rate (ORR) of 17% and did not meet the prespecified ORR of 40% (100,101). Concurrent RT (total 30 Gy in 10 fx or total 27 Gy in 3 fx) with ipilimumab and nivolumab did not demonstrate improved efficacy over immunotherapy alone in patients with advanced melanoma who received prior immunotherapy and/or targeted therapy (98). An ongoing large, randomized phase II trial aims to evaluate the efficacy of ipilimumab, nivolumab, and RT in melanoma patients with brain metastases (102). The combination of ipilimumab-nivolumab with SBRT (total 45-50 Gy in 3-5 fx) demonstrated ORR of 45% in treatment-naïve NSCLC patients, which is numerically higher than ipilimumab-nivolumab ORR of 33% in metastatic NSCLC patients unselected for PD-L1 status (92,103). Noteworthy, in patients with microsatellite stable colorectal and pancreatic cancers who progressed on first-line therapy, the combination of ipilimumab-nivolumab and RT (total 24 Gy in 3 fx) resulted in an encouraging disease control rate of 25% and 20% in colorectal and pancreatic groups, respectively (104). Table 3 further summarizes randomized trials exploring these combinations.
Table 3.
Clinical trial results of RT with anti-PD1 (PD-L1) and/or anti-CTLA4 therapya
Study | Immunotherapy | RT | Study population | Key finding(s) |
---|---|---|---|---|
Qin et al. (89) | Ipilimumab (Ipi) |
|
Advanced melanoma | Ipi before RT vs Ipi after RT resulted in longer duration of irradiated tumor response. |
Kwon et al. (75) | Ipilimumab | Total 8 Gy in 1 fraction | mCRPC with bone Mets | Ipi vs placebo after bone directed RT did not improve OS. |
Welsh et al. (55), Tang et al. (76) | Ipilimumab | Total 50 Gy divided in 4 fractions or total 60 Gy in 10 fractions | Advanced solid tumors with liver and/or lung Mets |
|
Formenti et al. (96) | Ipilimumab | Total 30 Gy in 5 fractions or total 27 Gy in 3 fractions |
|
|
Luke et al. (57,74) | Pembrolizumab | Total 30-50 Gy in 3-5 fractions | Advanced solid tumors |
|
Mahmood et al. (99) | Pembrolizumab | Total 30 Gy in 5 fractions | mACC |
|
Maity et al. (90) | Pembrolizumab |
|
|
|
Lee et al. (20) | Avelumab | Total 70 Gy in 35 fractions | Locally advanced SCHNC |
|
Masini et al. (100,101) | Nivolumab | Total 50 Gy in 5 fractions | mRCC |
|
Postow et al. (98) |
|
Total 30 Gy in 10 fractions or total 27 Gy in 3 fractions | Advanced melanoma |
|
Hammers et al. (97) |
|
50 Gy in 5 fractions | mRCC |
|
Patel et al. (105) |
|
|
|
|
Parikh et al. (104) |
|
Total 24 Gy in 3 fractions | MSS CRC and PDAC | DCR 25% and 20% |
Kim et al. (106) |
|
Total 24 Gy in 2 fractions | Merkel cell carcinoma |
|
Clump et al. (94) | Pembrolizumab | Total 70 Gy in 35 fractions | SCHNC |
|
AE = adverse effect; a/w = associated with; CRC = colorectal cancer; DCR = disease control rate; DLT = dose-limiting toxicity; G3 = grade 3; HR = hazard ratio; ICB = immune checkpoint blockade; IFN = interferon; IO = immune-oncology; mACC = metastatic adenoid cystic carcinoma; mCRPC = metastatic castrate resistant prostate cancer; Mets = metastases; mPFS = median progression free survival; mRCC = metastatic renal cell carcinoma; MSS = microsatellite stable; Nivo = nivolumab; NSCLC = non-small cell lung cancer; ORR = objective response rate; OS = overall survival; PBMC = peripheral blood mononuclear cell; PDAC = pancreatic ductal adenocarcinoma; PD-L1 = programmed death ligand 1; PFS = progression-free survival; RT = radiation therapy; SBRT = stereotactic body radiation therapy; SCHNC = squamous cell head and neck cancer; TRAE = treatment-related adverse event.
In summary, clinical studies combining anti-PD1 or PD-L1 and/or anti-CTLA4 therapies with RT demonstrate immunomodulation triggered by RT but efficacy is yet required to be investigated in further large-scale studies.
RT in combination with novel immunotherapy agents
The introduction of novel immunotherapy agents may be a promising approach to overcome RT-induced immunosuppression and synergize with the immunostimulatory effects of RT. Although many approaches might be considered, near-term possibilities may include but are not necessarily limited to innate immune targets to enhance tumor-associated antigen shedding and presentation, cytokines and/or chemokines, T-cell checkpoints, and anti-CSF1 to overcome immunosuppressive macrophages.
Below, we review current data on combination therapy of RT with innate immune targets such as TLR, STING agonists, cytokines/chemokines, and IFN-γ–linked mechanisms.
Targeting innate immunity
RT with TLR agonist therapy
TLRs are a class of pattern recognition receptor, which recognize damage-associated molecular patterns and play an essential role in innate immunity (107-109). Targeting TLR pathway along with RT may have synergistic activity in increasing tumor antigenicity, activation of DCs, and induction of tumor-specific cytotoxic T lymphocytes (110,111). The TLR/MyD88 pathway has been described to play an important role in RT-induced antitumor activity (51).
The activity of TLR agonists, such as TLR3, TLR7, and TLR9 combined with RT, has been tested via various preclinical models. In the Lewis lung carcinoma model, RT with TLR3 agonist polyinosinic‐polycytidylic acid (poly [I:C])–induced tumor growth inhibition was dependent on Batf3-positive DCs, CD8 T-cell activation, and TNF-α production. Pretreatment with poly(I:C) as opposed to after RT showed more robust antitumor efficacy mainly driven by decreased infiltration of MDSCs (112). The efficacy of intratumorally as well as intravenously administered TLR7 agonists combined with RT was demonstrated in murine colorectal, lymphoma, and melanoma models. The combination resulted in autophagic cell death, increase in CD8+ T cells, and decrease in Tregs and MDSCs with subsequent clearance of tumor cells and reduction of metastatic potential of tumor cells in melanoma and pancreatic models (113,114). This antitumor effect was more potent with fractionated RT (2 Gyx2) as opposed to single 10 Gy RT (113,115,116). TLR9 agonists and RT have been found to have a synergistic activity in increasing tumor antigenicity. They activated nuclear factor-kB and mitogen-activated protein kinases and enhanced both innate and adaptive immune system and the immune system response against tumor-associated antigens (117-119).
Preclinical data support combining TLR agonists with RT and has led to multiple early phase trials evaluating the role of TLR agonists to enhance responsiveness to RT (115,117,120-124). Two phase II clinical trials have studied RT with TLR3 agonist combinations in patients with glioblastoma multiforme (123,125). Polyinosinic-Polycytidylic (Poly-ICLC) with RT (2 Gyx30) was well tolerated and resulted in median OS of approximately 15 months, which is comparable with standard-of-care RT plus temozolomide. Adjuvant poly-ICLC and chemoRT was well tolerated but provided only mild improvement in median OS (17.2 months). The combination of intratumorally injected TLR9 agonists and RT (2 Gyx2) was safe in patients with low grade B-cell lymphoma patients and resulted in an overall response rate of 26.7%. This combination was later evaluated in patients with mycosis fungoides and was well tolerated resulting in clinically meaningful results in approximately 30% of patients (126-128). The injection of class C CpG (TLR9 agonist) SD-101 with RT was safe and resulted in tumor burden reduction in approximately 90% of patients with indolent lymphoma. The treatment was associated with an increase in the number of CD4+ effector and CD8+ cells and a decrease in Treg cells in the injected lesions (126,129,130,131). These studies were done in a small cohort of patients, and despite demonstrating safety and potentially clinically meaningful profiles along with promising immune reinvigoration, they did not reach to large phase II-III trials to officially evaluate the clinical benefit.
RT with STING agonist therapy
STING is another pattern recognition receptor that drives immune responses via activation of IFN-1, vascular normalization, and infiltration of CD8 T cells, as well as induction of nonclassical tertiary lymphoid structures (132-134). cyclic GMP-AMP synthase (cGAS)-STING pathway plays an important role in RT-induced antitumor immunity. Radiation-induced IFN-1 signaling and DC sensing of irradiated tumor cells are dependent on STING protein (135). In STING-deficient mice, RT fails to induce increases in IFN-β and CXCL10. STING is also a mediator for RT-induced adaptive immunity. RT induction of STING increases in tumor-associated antigen-specific CD8+ T cells in wild-type mice, whereas in STING-deficient Tmem173-/-, mice failed to demonstrate increases in CD8+ T cells (135). In HNSCC cell lines, STING was shown to regulate genes involved in reactive oxygen homeostasis with STING-deficient cells demonstrating decreased reactive oxygen species. Intravenously administered STING agonist SB11285 demonstrated synergistic activity with RT resulting in prolonged tumor growth inhibition in HNSCC (136). Although there are no results from human clinical studies that evaluated the combination of RT with STING agonists, this approach is promising to investigate (137).
RT with cytokine therapy
The combination of RT with cytokine therapy has been demonstrated to induce immunostimulatory changes and antitumor activity in preclinical models. The interaction of TNF-α and RT has been described since the 1990s, reporting radiosensitizing effects of TNF-α while using it before irradiation (138). Low-dose total body irradiation with IL-2 resulted in reduction of tumor burden in comparison to IL-2 alone and increased intratumoral natural killer cells and macrophages in a murine melanoma model (139). Hypofractionated RT at the dose of 6 Gyx4 combined with IL-12 microsphere resulted in greater tumor reduction in orthotopic pancreatic ductal carcinoma model in comparison to hypofractionated RT alone. The treatment resulted in intratumoral increase in IFN-γ and CXCL10, upregulation of M1, and downregulation of immunosuppressive M2 genes (140). RT at the dose of 10 Gyx1 combined with intratumorally injected IL-12 resulted in tumor regression in orthotopic hepatocellular carcinoma model. The therapy resulted in a decrease in MDSCs and increase in CD40+, CD80+, MHC II expressed DCs, CD8+, and natural killer cells (141). The RT at the dose of 8 Gyx3 in contrast to 20 Gyx1 combined with subcutaneous IL-15 vs IL-15 or RT alone resulted in long-term irradiated and nonirradiated tumor growth control in TSA mammary cancer cell lines. The RT with IL-15 resulted in an increase in the number of CD103 dendritic cells (cDCs), expression of CD40, CD80, and CD86 costimulatory molecules, and intratumoral infiltration of CD8+ T cells (142,143).
While thinking about combining cytokines with RT in the clinical setting, careful evaluation of safety data must be considered. Cytokine therapies have historically been associated with adverse events limiting their broad use in clinical settings. Small case series of melanoma patients raised concerns regarding safety of IFN with RT combination because of observed RT necrosis in brain and subcutaneous tissue (144). In stage III melanoma, IFN with RT (6 Gyx5) combination showed acceptable safety profile with the most common dermatological severe toxicity being witnessed in 9% of patients (145). Other combinations including IL-2, GM-CSF, and TNF-α demonstrated no dose-limiting toxicities and were overall well tolerated in patients with melanoma, renal cell carcinoma, and pancreatic cancer (6,8,146,147,148). In summary, the studies combining cytokines with RT have not demonstrated significant increases in toxicities in comparison with each modality alone.
With the introduction of IL-2 as a therapeutic modality for patients with melanoma and RCC, several studies then investigated combining IL-2 with RT aiming to increase antitumor responses. In a small study of 12 patients with melanoma and RCC, the combination of SBRT (20 Gy per fx) administered before IL-2 resulted in ORRs of 71% and 60%, respectively. The responders demonstrated higher peripheral blood CD4+ effector memory T cells than nonresponders (6). In a randomized phase II trial of SBRT (1 or 2 doses, 20 Gy per fx) with IL-2 in 44 patients with advanced melanoma, SBRT with IL-2 resulted in 54% ORR in contrast to 35% in IL-2 alone cohort. However, enhanced ORR did not translate to improvement in PFS or OS. The study showed no difference in ORR in radiating 1 vs more than 1 tumor sites (7).
Golden et al. (8) combined GM-CSF with RT (35 Gy in 10 fx) targeting rate of abscopal responses as a primary objective of their study. The combination resulted in nonirradiated tumor response in 27.6% of patients with improved OS among abscopal responders. Patients with progressive disease had higher neutrophil to lymphocyte ratio at baseline in comparison with those in responders. The abscopal response was also described in a patient with metastatic pancreatic cancer patients when combining GM-CSF with local RT (45 Gy in 15fx) (54).
In the initial phase I study, intratumoral injection of TNFerade (adenovector containing TNF-α with a RT-inducible promoter) was combined with RT (20-70 Gy) and demonstrated safety of this approach along with ORR of 70% in patients with advanced solid tumors (149,150). However, follow-up study did not demonstrate with recurrent head and neck cancer and pancreatic cancer did not demonstrate superiority in OS (148,151).
In contrast to the above-mentioned cytokines, which are not widely used in clinical practice, targeting IL-6 and IL-8 are of particular interest for studying in investigational settings. IL-6 and IL-8 promote recruitment of immunosuppressive MDSCs, and overcoming RT-induced expansion of MDSCs and M2 macrophage is an important area of investigation to expand the clinical benefit of RT.
Overcoming immunosuppressive effects of RT
MDSCs induced immunosuppression and M2 macrophage polarization
Various preclinical studies proposed targets such as colony-stimulating factor 1 receptor (CSFR1), IL-6, and IL-8 to be combined with RT to address this immunosuppressive effect (62,141,152-154). RT resulted in an increase in CSF1 gene expression in irradiated tumor and serum CSF1 cytokine levels in patients with prostate cancer. The addition of CSF1R kinase inhibitor PLX3397 to RT resulted in a decrease in intratumoral and splenic MDSCs in RM-1 prostate cancer mouse model (62). In GL261 glioma cells, this combination also resulted in an increase in number of tumor-infiltrating lymphocytes, CD8+ and CD4+ T cells, and tumor-specific IFN-γ production (153). In vitro studies of RT-resistant esophageal squamous cell carcinoma cells demonstrate increased expression of IL-8 levels, suggesting that elevated IL-8 is one of the mechanisms of RT resistance (155). IL-6 enhanced recruitment of MDSCs in irradiated prostate tumor microenvironment and inhibition of IL-6 resulted in antitumor effect (156). The elevation of IL-6 after irradiation has been reported in C6 glioma cells resulting in radioresistance (157).
In the clinical setting, the safety and efficacy of combination of cabiralizumab (anti-CSF1R mAb) with nivolumab and hypofractionated RT was evaluated in patients with advanced solid tumors. The therapy was safe, resulting in a DLT rate of 12% and requiring no dose reduction of hypofractionated RT. The ORR was 22%, and responses were driven mainly by a decrease in size of irradiated tumor (158,159). IL-8 levels prior to irradiation were inversely associated with response to IO-RT in this study. The phase I clinical trial of anti–IL-8 therapy with RT (30-50 Gy in 3-5 fx) and anti-PD1 therapy (NCT04572451) is ongoing in patients with advanced solid tumors and melanoma. The trial of anti–IL-6 tocilizumab with ipilimumab and nivolumab and RT is ongoing in patients with advanced pancreatic cancer (NCT04258150).
RT with anti-TGF-β therapy
TGF-β signaling pathway activation is one of major immunosuppressive TGF-β 1 in irradiated lesions, which can create an immunosuppressive environment limiting activity of RT in addition to enhancing fibrosis and inducing chronic organ damage (63,160).TGF-β creates immunosuppressive environment via inhibition of dendritic cell activity, cytotoxic T-lymphocyte function, decreased production of IFN-γ, and activation of Treg cells, along with decreasing the effectiveness of RT via inducing DNA damage response (161-164).Targeting TGF-β pathway can be promising in not only increasing efficacy of RT but also preventing RT-induced pulmonary and rectal fibrosis (165,166).The inhibition of TGF-β with TGF-β kinase receptor 1 inhibitor increased radiosensitivity of glioblastoma cells, resulted in increase in DNA double-strand breaks, and reduced DNA damage repair. Inhibition of TGF-β pathway impacted RT-induced gene expression changes with the largest effect of combination therapy being on cellular growth and proliferation (167). In 4T1 mammary breast cancer mouse model, TGF-β inhibition along with RT resulted in increase in CD8+ T cells and CD4+ T cells, gene expression profile of IFN-γ network and immune effector functions, and regression of irradiated and nonirradiated pulmonary metastatic lesions. The combination therapy resulted in increased expression of PD-L1 and PD-L2 in some proportion of 4T1 cell lines, and further anti-PD1 blockade resulted in complete regression of tumor (168). In murine CT26 colorectal cancer (CRC) model, oral inhibitor of TGF-β SM16 was given prior to RT resulting in CD8+ T cells in the tumor and CD4+ T cells in TDLNs. Preclinical data provide evidence of combination of RT with TGF-β inhibitors, but safety and efficacy are yet to be confirmed in human clinical trials.
Targeting Indoleamine 2,3-dioxygenase (IDO) pathway
Several preclinical studies support the enhanced immune activation while combining RT with indoleamine 2,3-dioxygenase pathway targeting molecules. The main function of IDO1 is the regulation of acquired local and peripheral immune tolerance in normal and pathological scenarios (169). As such, IDO1 activity induced the Treg compartment of the immune system, with concomitant inhibition of the effector T-cell compartment (170,171). Combination of IDO inhibitor with RT resulted in upregulation of CD80, CD86, and the major histocompatibility complex II in spleen DCs and the concurrent downregulation of CD4, CD25, and FOXP3 Tregs in Lewis lung cancer model (172). IDO1 enzyme inhibitor combination with anti-PD1mAb (PD1- monoclonal antibody) and RT resulted in tumor regression in a preclinical glioblastoma model (173). In the CRC model, IDO1 inhibition sensitized CRC to RT-induced cell death, whereas the IDO1 metabolite kynurenine promoted radioprotection. IDO1 inhibition also potentiated Th1 cytokines and myeloid cell-modulating factors in the tumor microenvironment and promoted an abscopal effect on tumors outside the RT field. Conversely, IDO1 blockade protected the normal small intestinal epithelium from RT toxicity and accelerated recovery from RT-induced weight loss, indicating a role in limiting side effects. These data demonstrated that IDO1 inhibition potentiates RT effectiveness in CRC (174). Overall, there are no clinical data about combining RT with IDO inhibitors other than a study in children with diffuse intrinsic pontine glioma combination of IDO pathway inhibitor indoximod with RT (54 Gy in 30 fx), and chemotherapy was well tolerated and improved OS in comparison with historical control (175).
Future directions and summary
With the rapid development of novel cancer immunotherapies, a number of new immune mechanisms are coming into the clinical setting, which may have the potential to enhance and overcome resistance mechanisms of RT. In particular, the introduction of a new generation of ICB agents in clinical trials such as T-cell immunoglobulin and ITIM domain (TIGIT), lymphocyte activation gene 3 (LAG3), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) brings a new potential for combining these agents with hypofractionated RT. In a CT26 colon cancer mouse model, the combination of anti-PD1 and anti-TIGIT with 8 Gy RT in 3 fractions resulted in complete regression of tumor in 90% of cases. The antitumor effect of anti-TIGIT was not seen while combining with 2 Gy RT in 18 fractions. The results highlight that different regimens of RT may be required while combining with various ICBs (10). In a murine hepatocellular cancer model, RT induced increased expression of TIM3 on CD4+ and CD8+ TILs. The combination of RT and anti-TIM3 resulted in improved responses mainly driven via CD8+ T-cell activation (176). In a murine glioma model, the combination of RT with anti-TIM3 and anti-PD1 resulted in increase in CD8+ to Treg ratio, IFN-γ–secreting CD8+ cells and improved survival in comparison with anti-TIM3, anti-PD1, and RT monotherapies (177). Another promising combination therapy includes RT with targeting myeloid checkpoint “don’t-eat-me” molecule CD47. In a mouse KP1 small-cell lung cancer model, anti-CD47 antibody enhanced antitumor activity of local RT as well as abscopal responses in a macrophage-mediated T-cell independent manner (178). In addition to novel IO-based therapies, an intriguing combination approach may be using radioconjugates, which deliver RT with tumor-targeting agents to facilitate new IO-RT combinations. In ovarian and breast preclinical models, 225Ac-anti-HER2 and anti-CD47 antibodies demonstrated improved efficacy over each single agent alone. The underlying mechanism of this enhanced response is potentially driven by upregulation of calreticulin and prophagocytic signaling activity (179). Another developing approach for broadening the systemic impact of RT is biology-guided RT (BgRT), which uses positron emission tomography as an imaging tool to characterize the biological characteristics of the tumor and to deliver RT. The fast-rotating linear accelerator obtains signals in a real-time fashion from different metastatic sites allowing treatment of multiple tumor locations during the same session with reduced RT dose. BgRT for lung SBRT has been demonstrated to reduce planned target volumes and organs at risk (180,181). Lastly, nanoparticle-based drug delivery is an evolving approach with the goal to deliver the drug to the tumor region with less systemic toxic effects. This has gained a particular importance in non–T-cell inflamed tumors and for drugs that demonstrated systemic toxicities otherwise. Zn-CDA is a tumor-targeted STING agonist that demonstrated improved efficacy in combination with RT and anti–PD-L1 over either strategy alone in T-cell noninflammed pancreatic cancer and glioma models. The treatment was associated with enhanced gene expression related to antigen presentation and M1 polarization (182).
In summary, current data support the safety of combination therapy with RT and immunotherapy along with biological correlates showing enhanced immune response to combinatorial therapy. Furthermore, preclinical data support further efficacy investigation of novel ICB agents with RT along with targeting TGF-β, anti–IL-8, IDO, TLRs, and STING. Apart from incorporation of novel IO treatments, approaches such as radioconjugates, BgRT, and nanoparticles are advanced strategies to use for future combinatorial therapies. These as well as other RT combinatorial partners represent a bright future for clinical investigation of novel RT-IO clinical trials.
Acknowledgements
Funding sources provide infrastructure support for investigation and team coordination surrounding clinical trial pursuit.
Contributor Information
Lilit Karapetyan, Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
Uzoma K Iheagwara, Department of Medicine, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, USA; Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
Adam C Olson, Department of Medicine, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, USA; Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
Steven J Chmura, Department of Radiation Oncology, University of Chicago, Chicago, IL, USA.
Heath K Skinner, Department of Medicine, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, USA; Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
Jason J Luke, Department of Medicine, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
Data availability
No new data were generated or analyzed in support of this research.
Author contributions
Lilit Karapetyan, MD, MS, FACP (Conceptualization; Writing—original draft; Writing—review & editing), Uzoma K. Iheagwara, MD, PhD (Writing—original draft; Writing—review & editing), Adam C. Olson, MD, MS (Writing—original draft; Writing—review & editing), Steven J. Chmura, MD, PhD (Writing—review & editing), Heath K. Skinner, MD, PhD (Writing—original draft; Writing—review & editing), and Jason J. Luke, MD, FACP (Conceptualization; Writing—original draft; Writing—review & editing)
Funding
This work was supported by the National Institutes of Health (UM1CA186690-06, P50CA254865-01A1, P30CA047904-32, R01DE031729-01A1 to JJL).
Conflicts of interest
Jason J. Luke reports: Outside of submitted work: Data and Safety Monitoring Board: Abbvie, Immutep, Evaxion; Scientific Advisory Board: (no stock) 7 Hills, Affivant, Bright Peak, Exo, Fstar, Inzen, RefleXion, Xilio (stock) Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, physIQ, Pyxis, Saros, STipe, Tempest; Consultancy with compensation: Abbvie, Agenus, Alnylam, Atomwise, Bayer, Bristol-Myers Squibb, Castle, Checkmate, Codiak, Crown, Cugene, Curadev, Day One, Eisai, EMD Serono, Endeavor, Flame, G1 Therapeutics, Genentech, Gilead, Glenmark, HotSpot, Kadmon, KSQ, Janssen, Ikena, Inzen, Immatics, Immunocore, Incyte, Instil, IO Biotech, Macrogenics, Merck, Mersana, Nektar, Novartis, Partner, Pfizer, Pioneering Medicines, PsiOxus, Regeneron, Replimmune, Ribon, Roivant, Servier, STINGthera, Synlogic, Synthekine; Research Support: (all to institution for clinical trials unless noted) AbbVie, Astellas, Astrazeneca, Bristol-Myers Squibb, Corvus, Day One, EMD Serono, Fstar, Genmab, Ikena, Immatics, Incyte, Kadmon, KAHR, Macrogenics, Merck, Moderna, Nektar, Next Cure, Numab, Palleon, Pfizer, Replimmune, Rubius, Servier, Scholar Rock, Synlogic, Takeda, Trishula, Tizona, Xencor; Patents: (both provisional) Serial #15/612,657 (Cancer Immunotherapy), PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof). Steven J Chmura: Outside of submitted work: Royalties from Wolters Kluwer (UpToDate). Adam C. Olson reports: Outside of submitted work: research funding from Varian Medical Systems, Reflexion Medical and serves as a consultant for RenovoRx.
The other authors (LK, UKI, HKS) declare no disclosures or conflicts of interest.
References
- 1. Ribas A, Wolchok JD.. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350-1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Spiotto M, Fu Y-X, Weichselbaum RR.. The intersection of radiotherapy and immunotherapy: mechanisms and clinical implications. Sci Immunol. 2016;1(3):eaag1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Weichselbaum RR, Liang H, Deng L, Fu Y-X.. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol. 2017;14(6):365-379. [DOI] [PubMed] [Google Scholar]
- 4. Grassberger C, Ellsworth SG, Wilks MQ, Keane FK, Loeffler JS.. Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol. 2019;16(12):729-745. [DOI] [PubMed] [Google Scholar]
- 5. Pitroda SP, Chmura SJ, Weichselbaum RR.. Integration of radiotherapy and immunotherapy for treatment of oligometastases. Lancet Oncol. 2019;20(8):e434-e42. [DOI] [PubMed] [Google Scholar]
- 6. Seung SK, Curti BD, Crittenden M, et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2—tumor and immunological responses. Sci Transl Med. 2012;4(137):137ra74. [DOI] [PubMed] [Google Scholar]
- 7. Curti B, Crittenden M, Seung SK, et al. Randomized phase II study of stereotactic body radiotherapy and interleukin-2 versus interleukin-2 in patients with metastatic melanoma. J Immunother Cancer. 2020;8(1):e000773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Golden EB, Chhabra A, Chachoua A, 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] [PubMed] [Google Scholar]
- 9. Barker CA, Postow MA.. Combinations of radiation therapy and immunotherapy for melanoma: a review of clinical outcomes. Int J Radiat Oncol Biol Phys. 2014;88(5):986-997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Grapin M, Richard C, Limagne E, et al. Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: a promising new combination. J Immunother Cancer. 2019;7(1):160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kaur P, Asea A.. Radiation-induced effects and the immune system in cancer. Front Oncol. 2012;2:191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Chen D, Patel RR, Verma V, et al. Interaction between lymphopenia, radiotherapy technique, dosimetry, and survival outcomes in lung cancer patients receiving combined immunotherapy and radiotherapy. Radiother Oncol. 2020;150:114-120. [DOI] [PubMed] [Google Scholar]
- 13. Saddawi-Konefka R, O’Farrell A, Faraji F, et al. Lymphatic-preserving treatment sequencing with immune checkpoint inhibition unleashes cDC1-dependent antitumor immunity in HNSCC. Nat Commun. 2022;13(1):4298. [DOI] [PMC free article] [PubMed]
- 14. 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(6):2345-2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Pandey R, Shankar BS, Sharma D, Sainis KB.. Low dose radiation induced immunomodulation: effect on macrophages and CD8+ T cells. Int J Radiat Biol. 2005;81(11):801-812. [DOI] [PubMed] [Google Scholar]
- 16. Coates PJ, Rundle JK, Lorimore SA, Wright EG.. Indirect macrophage responses to ionizing radiation: implications for genotype-dependent bystander signaling. Cancer Res. 2008;68(2):450-456. [DOI] [PubMed] [Google Scholar]
- 17. Antonia SJ, Villegas A, Daniel D, et al. ; for the PACIFIC Investigators. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379(24):2342-2350. [DOI] [PubMed] [Google Scholar]
- 18. Antonia SJ, Villegas A, Daniel D, et al. ; for the PACIFIC Investigators. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377(20):1919-1929. [DOI] [PubMed] [Google Scholar]
- 19. Spigel DR, Faivre-Finn C, Gray JE, et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J Clin Oncol. 2022;40(12):1301-1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Lee NY, Ferris RL, Psyrri A, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol 2021;22(4):450-462. [DOI] [PubMed] [Google Scholar]
- 21. Bourhis J, Sire C, Tao Y, et al. LBA38 pembrolizumab versus cetuximab, concomitant with radiotherapy (RT) in locally advanced head and neck squamous cell carcinoma (LA-HNSCC): results of the GORTEC 2015-01 “PembroRad” randomized trial. Ann Oncol. 2020;31:S1168. [DOI] [PubMed] [Google Scholar]
- 22. Mell LK, Torres-Saavedra P, Wong S, et al. Radiotherapy with durvalumab vs. cetuximab in patients with locoregionally advanced head and neck cancer and a contraindication to cisplatin: phase II results of NRG-HN004. In: Presented at the 2022 American Society of Radiation Oncology Annual Meeting; October 23–26, 2022; San Antonio, TX. Abstract LBA02. Accessed October 24, 2022.
- 23. Salama JK, Hasselle MD, Chmura SJ, et al. Stereotactic body radiotherapy for multisite extracranial oligometastases: final report of a dose escalation trial in patients with 1 to 5 sites of metastatic disease. Cancer. 2012;118(11):2962-2970. [DOI] [PubMed] [Google Scholar]
- 24. Joseph RW, Elassaiss-Schaap J, Kefford R, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab. Clin Cancer Res. 2018;24(20):4960-4967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Katsurada M, Nagano T, Tachihara M, et al. Baseline tumor size as a predictive and prognostic factor of immune checkpoint inhibitor therapy for non-small cell lung cancer. Anticancer Res. 2019;39(2):815-825. [DOI] [PubMed] [Google Scholar]
- 26. Moynihan KD, Opel CF, Szeto GL, et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med. 2016;22(12):1402-1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kim SI, Cassella CR, Byrne KT.. Tumor burden and immunotherapy: impact on immune infiltration and therapeutic outcomes. Front Immunol. 2021;11(3762). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Guisier F, Cousse S, Jeanvoine M, Thiberville L, Salaun M.. A rationale for surgical debulking to improve anti-PD1 therapy outcome in non small cell lung cancer. Sci Rep. 2019;9(1):16902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Huang AC, Postow MA, Orlowski RJ, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545(7652):60-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Hall EG. Radiobiology for the Radiologist. 8th ed. Philadelphia, PA: Wolters Kluwer; 2019. [Google Scholar]
- 31. Shinde A, Li R, Kim J, Salgia R, Hurria A, Amini A.. Stereotactic body radiation therapy (SBRT) for early-stage lung cancer in the elderly. Semin Oncol. 2018;45(4):210-219. [DOI] [PubMed] [Google Scholar]
- 32. Gameiro SR, Malamas AS, Bernstein MB, et al. Tumor cells surviving exposure to proton or photon radiation share a common immunogenic modulation signature, rendering them more sensitive to T cell-mediated killing. Int J Radiat Oncol Biol Phys. 2016;95(1):120-130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Durante M, Reppingen N, Held KD.. Immunologically augmented cancer treatment using modern radiotherapy. Trends Mol Med. 2013;19(9):565-582. [DOI] [PubMed] [Google Scholar]
- 34. Welsh J, Gomez D, Palmer MB, et al. Intensity-modulated proton therapy further reduces normal tissue exposure during definitive therapy for locally advanced distal esophageal tumors: a dosimetric study. Int J Radiat Oncol Biol Phys. 2011;81(5):1336-1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Davuluri R, Jiang W, Fang P, et al. Lymphocyte nadir and esophageal cancer survival outcomes after chemoradiation therapy. Int J Radiat Oncol Biol Phys. 2017;99(1):128-135. [DOI] [PubMed] [Google Scholar]
- 36. Mirjolet C, Nicol A, Limagne E, et al. Impact of proton therapy on antitumor immune response. Sci Rep. 2021;11(1):13444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Malouff TD, Mahajan A, Krishnan S, Beltran C, Seneviratne DS, Trifiletti DM.. Carbon ion therapy: a modern review of an emerging technology. Front Oncol. 2020;10:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zhou H, Tu C, Yang P, et al. Carbon ion radiotherapy triggers immunogenic cell death and sensitizes melanoma to anti-PD-1 therapy in mice. Oncoimmunology. 2022;11(1):2057892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Palcic B, Korbelik M, Trotter M, Revesz L.. Oxygen enhancement ratio of fractionated regimens in vitro. Radiat Res. 1989;117(3):409-418. [PubMed] [Google Scholar]
- 40. Pickles T, Goodman GB, Fryer CJ, et al. Pion conformal radiation of prostate cancer: results of a randomized study. Int J Radiat Oncol Biol Phys. 1999;43(1):47-55. [DOI] [PubMed] [Google Scholar]
- 41. Pickles T, Goodman GB, Rheaume DE, et al. Pion radiation for high grade astrocytoma: results of a randomized study. Int J Radiat Oncol Biol Phys. 1997;37(3):491-497. [DOI] [PubMed] [Google Scholar]
- 42. Yan W, Khan MK, Wu X, et al. Spatially fractionated radiation therapy: history, present and the future. Clin Transl Radiat Oncol. 2020;20:30-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Zhang Y, Ding Z, Perentesis JP, Khuntia D, Pfister SX, Sharma RA.. Can rational combination of ultra-high dose rate FLASH radiotherapy with immunotherapy provide a novel approach to cancer treatment? Clin Oncol (R Coll Radiol). 2021;33(11):713-722. [DOI] [PubMed] [Google Scholar]
- 44. Bourhis J, Sozzi WJ, Jorge PG, et al. Treatment of a first patient with FLASH-radiotherapy. Radiother Oncol. 2019;139:18-22. [DOI] [PubMed] [Google Scholar]
- 45. Demaria S, Formenti SC.. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol. 2012;2:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Formenti SC, Demaria S.. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10(7):718-726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Golden EB, Pellicciotta I, Demaria S, Barcellos-Hoff MH, Formenti SC.. The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol. 2012;2:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Phillips R, Shi WY, Deek M, et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol. 2020;6(5):650-659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Fucikova J, Kepp O, Kasikova L, et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Galluzzi L, Vitale I, Warren S, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 2020;8(1):e000337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. 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(9):1050-1059. [DOI] [PubMed] [Google Scholar]
- 52. Fucikova J, Spisek R, Kroemer G, Galluzzi L.. Calreticulin and cancer. Cell Res. 2021;31(1):5-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Abuodeh Y, Venkat P, Kim S.. Systematic review of case reports on the abscopal effect. Curr Probl Cancer. 2016;40(1):25-37. [DOI] [PubMed] [Google Scholar]
- 54. Shi F, Wang X, Teng F, Kong L, Yu J.. Abscopal effect of metastatic pancreatic cancer after local radiotherapy and granulocyte-macrophage colony-stimulating factor therapy. Cancer Biol Ther. 2017;18(3):137-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Welsh JW, Tang C, de Groot P, et al. Phase II trial of ipilimumab with stereotactic radiation therapy for metastatic disease: outcomes, toxicities, and low-dose radiation–related abscopal responses. Cancer Immunol Res. 2019;7(12):1903-1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Foster CC, Fleming GF, Karrison TG, et al. Phase I study of stereotactic body radiotherapy plus nivolumab and urelumab or cabiralizumab in patients with advanced solid tumors. Clin Cancer Res. 2021;27(20):5510-5518. [DOI] [PubMed] [Google Scholar]
- 57. Luke JJ, Lemons JM, Karrison TG, et al. Safety and clinical activity of pembrolizumab and multisite stereotactic body radiotherapy in patients with advanced solid tumors. J Clin Oncol 2018;36(16):1611-1618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Gomez DR, Tang C, Zhang J, et al. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol 2019;37(18):1558-1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): a randomised, phase 2, open-label trial. Lancet 2019;393(10185):2051-2058. [DOI] [PubMed] [Google Scholar]
- 60. Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: a prospective, randomized, multicenter phase II trial. J Clin Oncol 2018;36(5):446-453. [DOI] [PubMed] [Google Scholar]
- 61. Demaria S, Golden EB, Formenti SC.. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol. 2015;1(9):1325-1332. [DOI] [PubMed] [Google Scholar]
- 62. 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(9):2782-2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Barcellos-Hoff MH, Derynck R, Tsang ML, Weatherbee JA.. Transforming growth factor-beta activation in irradiated murine mammary gland. J Clin Invest. 1994;93(2):892-899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Muroyama Y, Nirschl TR, Kochel CM, et al. Stereotactic radiotherapy increases functionally suppressive regulatory T cells in the tumor microenvironment. Cancer Immunol Res 2017;5(11):992-1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Moeller BJ, Cao Y, Li CY, Dewhirst MW.. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5(5):429-441. [DOI] [PubMed] [Google Scholar]
- 66. 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(19):5458-5468. [DOI] [PubMed] [Google Scholar]
- 67. Menon H, Ramapriyan R, Cushman TR, et al. Role of radiation therapy in modulation of the tumor stroma and microenvironment. Front Immunol. 2019;10:193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Brown JM, Thomas R, Nagpal S, Recht L.. Macrophage exclusion after radiation therapy (MERT): a new and effective way to increase the therapeutic ratio of radiotherapy. Radiother Oncol. 2020;144:159-164. [DOI] [PubMed] [Google Scholar]
- 69. Harris TJ, Hipkiss EL, Borzillary S, et al. Radiotherapy augments the immune response to prostate cancer in a time-dependent manner. Prostate. 2008;68(12):1319-1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Young KH, Baird JR, Savage T, et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 2016;11(6):e0157164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. 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(17):5379-5388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Vanpouille-Box C, Formenti SC, Demaria S.. TREX1 dictates the immune fate of irradiated cancer cells. Oncoimmunology. 2017;6(9):e1339857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. 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]
- 74. Luke JJ, Onderdonk BE, Bhave SR, et al. Improved survival associated with local tumor response following multi-site radiotherapy and pembrolizumab: secondary analysis of a phase I trial. Clin Cancer Res. 2020;26(24):6437-6444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Kwon ED, Drake CG, Scher HI, et al. ; for the CA184-043 Investigators. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15(7):700-712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. 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(6):1388-1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Ratnayake G, Reinwald S, Shackleton M, et al. Stereotactic radiation therapy combined with immunotherapy against metastatic melanoma: long-term results of a phase 1 clinical trial. Int J Radiat Oncol Biol Phys. 2020;108(1):150-156. [DOI] [PubMed] [Google Scholar]
- 78. Lambin P, Lieverse RIY, Eckert F, et al. Lymphocyte-sparing radiotherapy: the rationale for protecting lymphocyte-rich organs when combining radiotherapy with immunotherapy. Semin Radiat Oncol. 2020;30(2):187-193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Battaglia A, Buzzonetti A, Martinelli E, et al. Selective changes in the immune profile of tumor-draining lymph nodes after different neoadjuvant chemoradiation regimens for locally advanced cervical cancer. Int J Radiat Oncol Biol Phys. 2010;76(5):1546-1553. [DOI] [PubMed] [Google Scholar]
- 80. Marciscano AE, Ghasemzadeh A, Nirschl TR, et al. Elective nodal irradiation attenuates the combinatorial efficacy of stereotactic radiation therapy and immunotherapy. Clin Cancer Res. 2018;24(20):5058-5071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Buchwald ZS, Nasti TH, Lee J, et al. Tumor-draining lymph node is important for a robust abscopal effect stimulated by radiotherapy. J Immunother Cancer. 2020;8(2):e000867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Darragh LB, Gadwa J, Pham TT, et al. Elective nodal irradiation mitigates local and systemic immunity generated by combination radiation and immunotherapy in head and neck tumors. Nat Commun. 2022;13(1):7015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. 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(7547):373-377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Skinner HD, Giri U, Yang LP, et al. Integrative analysis identifies a novel AXL–PI3 kinase–PD-L1 signaling axis associated with radiation resistance in head and neck cancer. Clin Cancer Res. 2017;23(11):2713-2722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Martin AM, Cagney DN, Catalano PJ, et al. Immunotherapy and symptomatic radiation necrosis in patients with brain metastases treated with stereotactic radiation. JAMA Oncol. 2018;4(8):1123-1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Hwang WL, Pike LRG, Royce TJ, Mahal BA, Loeffler JS.. Safety of combining radiotherapy with immune-checkpoint inhibition. Nat Rev Clin Oncol. 2018;15(8):477-494. [DOI] [PubMed] [Google Scholar]
- 87. Du Four S, Janssen Y, Michotte A, et al. Focal radiation necrosis of the brain in patients with melanoma brain metastases treated with pembrolizumab. Cancer Med. 2018;7(10):4870-4879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Choi S, Hong A, Wang T, et al. Risk of radiation necrosis after stereotactic radiosurgery for melanoma brain metastasis by anatomical location. Strahlenther Onkol. 2021;197(12):1104-1112. [DOI] [PubMed] [Google Scholar]
- 89. Qin R, Olson A, Singh B, et al. Safety and efficacy of radiation therapy in advanced melanoma patients treated with ipilimumab. Int J Radiat Oncol Biol Phys. 2016;96(1):72-77. [DOI] [PubMed] [Google Scholar]
- 90. Maity A, Mick R, Huang AC, et al. A phase I trial of pembrolizumab with hypofractionated radiotherapy in patients with metastatic solid tumours. Br J Cancer. 2018;119(10):1200-1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Anscher MS, Arora S, Weinstock C, et al. Association of radiation therapy with risk of adverse events in patients receiving immunotherapy: a pooled analysis of trials in the US food and drug administration database. JAMA Oncol. 2022;8(2):232-240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Bestvina CM, Pointer KB, Karrison T, et al. A phase 1 trial of concurrent or sequential ipilimumab, nivolumab, and stereotactic body radiotherapy in patients with stage IV NSCLC study. J Thorac Oncol. 2022;17(1):130-140. [DOI] [PubMed] [Google Scholar]
- 93. Varlotto JM, Sun Z, Ramalingam SS, et al. Randomized phase III Trial of MEDI4736 (durvalumab) as concurrent and consolidative therapy or consolidative therapy alone for unresectable stage 3 NSCLC: a trial of the ECOG-ACRIN Cancer Research Group (EA5181). J Clin Oncol 2021;39(suppl 15):TPS8584-TPS. [Google Scholar]
- 94. Clump DA, Zandberg DP, Skinner HD, et al. A randomized phase II study evaluating concurrent or sequential fixed-dose immune therapy in combination with cisplatin and intensity-modulated radiotherapy in intermediate- or high-risk, previously untreated, locally advanced head and neck cancer (LA SCCHN). J Clin Oncol. 2022;40(suppl 16):6007. [Google Scholar]
- 95. Theelen WSME, Chen D, Verma V, et al. Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Respir Med 2021;9(5):467-475. [DOI] [PubMed] [Google Scholar]
- 96. Formenti SC, Rudqvist N-P, Golden E, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 2018;24(12):1845-1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Hammers HJ, Vonmerveldt D, Ahn C, et al. Combination of dual immune checkpoint inhibition (ICI) with stereotactic radiation (SBRT) in metastatic renal cell carcinoma (mRCC) (RADVAX RCC). J Clin Oncol 2020;38(suppl 6):614. [Google Scholar]
- 98. Postow MA, Knox SJ, Goldman DA, et al. A prospective, phase 1 trial of nivolumab, ipilimumab, and radiotherapy in patients with advanced melanoma. Clin Cancer Res. 2020;26(13):3193-3201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Mahmood U, Bang A, Chen Y-H, et al. A randomized phase 2 study of pembrolizumab with or without radiation in patients with recurrent or metastatic adenoid cystic carcinoma. Int J Radiat Oncol Biol Phys. 2021;109(1):134-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Masini C, Iotti C, Giorgi UD, et al. Nivolumab (NIVO) in combination with stereotactic body radiotherapy (SBRT) in pretreated patients (pts) with metastatic renal cell carcinoma (mRCC): first results of phase II NIVES study. J Clin Oncol. 2020;38(suppl 6):613.31829907 [Google Scholar]
- 101. Masini C, Iotti C, De Giorgi U, et al. Nivolumab in combination with stereotactic body radiotherapy in pretreated patients with metastatic renal cell carcinoma. Results of the phase II NIVES study. Eur Urol. 2022;81(3):274-282. [DOI] [PubMed] [Google Scholar]
- 102. Gonzalez M, Hong AM, Carlino MS, et al. A phase II, open label, randomized controlled trial of nivolumab plus ipilimumab with stereotactic radiotherapy versus ipilimumab plus nivolumab alone in patients with melanoma brain metastases (ABC-X Trial). J Clin Oncol. 2019;37(suppl 15):TPS9600. [Google Scholar]
- 103. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non–small-cell lung cancer. N Engl J Med 2019;381(21):2020-2031. [DOI] [PubMed] [Google Scholar]
- 104. Parikh AR, Szabolcs A, Allen JN, et al. Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial. Nat Cancer. 2021;2(11):1124-1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Patel JD, Bestvina CM, Karrison T, et al. Randomized phase I trial to evaluate Concurrent or Sequential Ipilimumab, Nivolumab, and stereotactic body Radiotherapy in patients with stage IV non-small cell lung cancer (COSINR Study). J Clin Oncol. 2020;38(suppl 15):9616. [Google Scholar]
- 106. Kim S, Wuthrick E, Blakaj D, et al. Combined nivolumab and ipilimumab with or without stereotactic body radiation therapy for advanced Merkel cell carcinoma: a randomised, open label, phase 2 trial. Lancet. 2022;400(10357):1008-1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Rakoff-Nahoum S, Medzhitov R.. Toll-like receptors and cancer. Nat Rev Cancer. 2009;9(1):57-63. [DOI] [PubMed] [Google Scholar]
- 108. Akira S, Uematsu S, Takeuchi O.. Pathogen recognition and innate immunity. Cell. 2006;124(4):783-801. [DOI] [PubMed] [Google Scholar]
- 109. Kawai T, Akira S.. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat Immunol. 2010;11(5):373-384. [DOI] [PubMed] [Google Scholar]
- 110. Chamoto K, Takeshima T, Wakita D, et al. Combination immunotherapy with radiation and CpG-based tumor vaccination for the eradication of radio- and immuno-resistant lung carcinoma cells. Cancer Sci. 2009;100(5):934-939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Teitz-Tennenbaum S, Li Q, Okuyama R, et al. Mechanisms involved in radiation enhancement of intratumoral dendritic cell therapy. J Immunother. 2008;31(4):345-358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Yoshida S, Shime H, Takeda Y, et al. Toll-like receptor 3 signal augments radiation-induced tumor growth retardation in a murine model. Cancer Sci. 2018;109(4):956-965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Hyun Cho JL, Ko H, Yoon B, et al. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget. 2017;8(15):24932-24948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Schölch S, Rauber C, Tietz A, et al. Radiotherapy combined with TLR7/8 activation induces strong immune responses against gastrointestinal tumors. Oncotarget. 2015;6(7):4663-4676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Dovedi SJ, Melis MH, Wilkinson RW, et al. Systemic delivery of a TLR7 agonist in combination with radiation primes durable antitumor immune responses in mouse models of lymphoma. Blood. 2013;121(2):251-259. [DOI] [PubMed] [Google Scholar]
- 116. Adlard AL, Dovedi SJ, Telfer BA, et al. A novel systemically administered toll-like receptor 7 agonist potentiates the effect of ionizing radiation in murine solid tumor models. Int J Cancer. 2014;135(4):820-829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Li X, Liu D, Liu X, et al. CpG ODN107 potentiates radiosensitivity of human glioma cells via TLR9-mediated NF-κB activation and NO production. Tumour Biol. 2012;33(5):1607-1618. [DOI] [PubMed] [Google Scholar]
- 118. Brody JD, Ai WZ, Czerwinski DK, et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol. 2010;28(28):4324-4332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Yuan S, Qiao T, Li X, et al. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol Lett. 2018;15(4):5271-5279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Mason KA, Ariga H, Neal R, et al. Targeting toll-like receptor 9 with CpG oligodeoxynucleotides enhances tumor response to fractionated radiotherapy. Clin Cancer Res. 2005;11(1):361-369. [PubMed] [Google Scholar]
- 121. Milas L, Mason KA, Ariga H, et al. CpG oligodeoxynucleotide enhances tumor response to radiation. Cancer Res. 2004;64(15):5074-5077. [DOI] [PubMed] [Google Scholar]
- 122. Walshaw RC, Honeychurch J, Choudhury A, Illidge TM.. Toll-like receptor agonists and radiation therapy combinations: an untapped opportunity to induce anticancer immunity and improve tumor control. Int J Radiat Oncol Biol Phys. 2020;108(1):27-37. [DOI] [PubMed] [Google Scholar]
- 123. Butowski N, Chang SM, Junck L, et al. A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05). J Neurooncol. 2009;91(2):175-182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Dewan MZ, Vanpouille-Box C, Kawashima N, et al. Synergy of topical toll-like receptor 7 agonist with radiation and low-dose cyclophosphamide in a mouse model of cutaneous breast cancer. Clin Cancer Res. 2012;18(24):6668-6678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Rosenfeld MR, Chamberlain MC, Grossman SA, et al. A multi-institution phase II study of poly-ICLC and radiotherapy with concurrent and adjuvant temozolomide in adults with newly diagnosed glioblastoma. Neuro Oncol. 2010;12(10):1071-1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Frank MJ, Reagan PM, Bartlett NL, et al. In situ vaccination with a TLR9 agonist and local low-dose radiation induces systemic responses in untreated indolent lymphoma. Cancer Discov. 2018;8(10):1258-1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Hamid O, Ismail R, Puzanov I.. Intratumoral immunotherapy—update 2019. Oncologist. 2020;25(3):e423-e438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27(2):161-167. [DOI] [PubMed] [Google Scholar]
- 129. Kohrt HE, Chu J, Brody J, et al. Dose-escalated, intratumoral TLR9 agonist and low-dose radiation induce abscopal effects in follicular lymphoma. Blood. 2014;124(21):3092.25193870 [Google Scholar]
- 130. Levy R, Reagan PM, Friedberg JW, et al. SD-101, a novel class C CpG-Oligodeoxynucleotide (ODN) toll-like receptor 9 (TLR9) agonist, given with low dose radiation for untreated low grade B-cell lymphoma: interim results of a phase ½ trial. Blood. 2016;128(22):2974. [Google Scholar]
- 131. Meng Y, Carpentier AF, Chen L, et al. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. Int J Cancer. 2005;116(6):992-997. [DOI] [PubMed] [Google Scholar]
- 132. Flood BA, Higgs EF, Li S, Luke JJ, Gajewski TF.. STING pathway agonism as a cancer therapeutic. Immunol Rev. 2019;290(1):24-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Chelvanambi M, Fecek RJ, Taylor JL, Storkus WJ.. STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment. J Immunother Cancer. 2021;9(2):e001906., [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Ishikawa H, Ma Z, Barber GN.. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461(7265):788-792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. 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(5):843-852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Hayman TJ, Baro M, MacNeil T, et al. STING enhances cell death through regulation of reactive oxygen species and DNA damage. Nat Commun. 2021;12(1):2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Amouzegar A, Chelvanambi M, Filderman JN, Storkus WJ, Luke JJ.. STING agonists as cancer therapeutics. Cancers (Basel). 2021;13(11):2695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Hallahan DE, Beckett MA, Kufe D, Weichselbaum RR.. The interaction between recombinant human tumor necrosis factor and radiation in 13 human tumor cell lines. Int J Radiat Oncol Biol Phys. 1990;19(1):69-74. [DOI] [PubMed] [Google Scholar]
- 139. Safwat A, Aggerholm N, Roitt I, Overgaard J, Hokland M.. Low-dose total body irradiation augments the therapeutic effect of interleukin-2 in a mouse model for metastatic malignant melanoma. J Exp Ther Oncol. 2003;3(4):161-168. [DOI] [PubMed] [Google Scholar]
- 140. Mills BN, Connolly KA, Ye J, et al. Stereotactic body radiation and interleukin-12 combination therapy eradicates pancreatic tumors by repolarizing the immune microenvironment. Cell Rep. 2019;29(2):406-421.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Wu C-J, Tsai Y-T, Lee IJ, et al. Combination of radiation and interleukin 12 eradicates large orthotopic hepatocellular carcinoma through immunomodulation of tumor microenvironment. Oncoimmunology. 2018;7(9):e1477459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Pilones KA, Charpentier M, Garcia-Martinez E, Demaria S.. IL15 synergizes with radiotherapy to reprogram the tumor immune contexture through a dendritic cell connection. Oncoimmunology. 2020;9(1):1790716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Pilones KA, Charpentier M, Garcia-Martinez E, et al. Radiotherapy cooperates with IL15 to induce antitumor immune responses. Cancer Immunol Res. 2020;8(8):1054-1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Hazard LJ, Sause WT, Noyes RD.. Combined adjuvant radiation and interferon-alpha 2B therapy in high-risk melanoma patients: the potential for increased radiation toxicity. Int J Radiat Oncol Biol Phys. 2002;52(3):796-800. [DOI] [PubMed] [Google Scholar]
- 145. Finkelstein SE, Trotti A, Rao N, et al. The Florida melanoma trial I: a prospective multicenter phase I/II trial of postoperative hypofractionated adjuvant radiotherapy with concurrent interferon-alfa-2b in the treatment of advanced stage III melanoma with long-term toxicity follow-up. Int Sch Res Notices Immunol. 2012;2012:1. [Google Scholar]
- 146. Safwat A, Schmidt H, Bastholt L, et al. A phase II trial of low-dose total body irradiation and subcutaneous interleukin-2 in metastatic melanoma. Radiother Oncol. 2005;77(2):143-147. [DOI] [PubMed] [Google Scholar]
- 147. Lange JR, Raubitschek AA, Pockaj BA, et al. A pilot study of the combination of interleukin-2-based immunotherapy and radiation therapy. J Immunother (1991). 1992;12(4):265-271. [DOI] [PubMed] [Google Scholar]
- 148. Herman JM, Wild AT, Wang H, et al. Randomized phase III multi-institutional study of TNFerade biologic with fluorouracil and radiotherapy for locally advanced pancreatic cancer: final results. J Clin Oncol. 2013;31(7):886-894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Senzer N, Mani S, Rosemurgy A, et al. TNFerade biologic, an adenovector with a radiation-inducible promoter, carrying the human tumor necrosis factor alpha gene: a phase I study in patients with solid tumors. J Clin Oncol. 2004;22(4):592-601. [DOI] [PubMed] [Google Scholar]
- 150. Hallahan DE, Mauceri HJ, Seung LP, et al. Spatial and temporal control of gene therapy using ionizing radiation. Nat Med. 1995;1(8):786-791. [DOI] [PubMed] [Google Scholar]
- 151. Seiwert TY, Darga T, Haraf D, et al. A phase I dose escalation study of Ad GV.EGR.TNF.11D (TNFeradeTM Biologic) with concurrent chemoradiotherapy in patients with recurrent head and neck cancer undergoing reirradiation. Ann Oncol. 2013;24(3):769-776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Kroon P, Gadiot J, Peeters M, et al. Concomitant targeting of programmed death-1 (PD-1) and CD137 improves the efficacy of radiotherapy in a mouse model of human BRAFV600-mutant melanoma. Cancer Immunol Immunother. 2016;65(6):753-763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Newcomb EW, Lukyanov Y, Kawashima N, et al. Radiotherapy enhances antitumor effect of anti-CD137 therapy in a mouse Glioma model. Radiat Res. 2010;173(4):426-432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Alfaro C, Teijeira A, Oñate C, et al. Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs). Clin Cancer Res 2016;22(15):3924-3936. [DOI] [PubMed] [Google Scholar]
- 155. Hua Q, Gu X, Chen X, Song W, Wang A, Chu J.. IL-8 is involved in radiation therapy resistance of esophageal squamous cell carcinoma via regulation of PCNA. Arch Biochem Biophys. 2019;676:108158. [DOI] [PubMed] [Google Scholar]
- 156. Wu C-T, Chen M-F, Chen W-C, Hsieh C-C.. The role of IL-6 in the radiation response of prostate cancer. Radiat Oncol. 2013;8(1):159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Tamari Y, Kashino G, Mori H.. Acquisition of radioresistance by IL-6 treatment is caused by suppression of oxidative stress derived from mitochondria after γ-irradiation. J Radiat Res. 2017;58(4):412-420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Foster C, Luke JJ, Hseu R, Janisch L, Fleming G, Chmura SJ.. Phase I study of multisite stereotactic body radiotherapy plus nivolumab and urelumab or nivolumab and cabiralizumab in patients with advanced solid tumors. J ImmunoTher Cancer. 2019;7(suppl 1):282.31694725 [Google Scholar]
- 159. Foster CC, Fleming GF, Karrison TG, et al. Phase I study of stereotactic body radiotherapy plus nivolumab and urelumab or cabiralizumab in advanced solid tumors. Clin Cancer Res. 2021;27(20):5510-5518. [DOI] [PubMed] [Google Scholar]
- 160. Anscher MS, Thrasher B, Rabbani Z, Teicher B, Vujaskovic Z.. Antitransforming growth factor-beta antibody 1D11 ameliorates normal tissue damage caused by high-dose radiation. Int J Radiat Oncol Biol Phys. 2006;65(3):876-881. [DOI] [PubMed] [Google Scholar]
- 161. Barcellos-Hoff MH, Newcomb EW, Zagzag D, Narayana A.. Therapeutic targets in malignant glioblastoma microenvironment. Semin Radiat Oncol. 2009;19(3):163-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Hardee ME, Marciscano AE, Medina-Ramirez CM, et al. Resistance of glioblastoma-initiating cells to radiation mediated by the tumor microenvironment can be abolished by inhibiting transforming growth factor-β. Cancer Res. 2012;72(16):4119-4129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Bettini M, Vignali DA.. Regulatory T cells and inhibitory cytokines in autoimmunity. Curr Opin Immunol. 2009;21(6):612-618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limón P.. The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol. 2010;10(8):554-567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Flechsig P, Dadrich M, Bickelhaupt S, et al. LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-β and BMP-associated proinflammatory and proangiogenic signals. Clin Cancer Res. 2012;18(13):3616-3627. [DOI] [PubMed] [Google Scholar]
- 166. Liu Y, Kudo K, Abe Y, et al. Inhibition of transforming growth factor-beta, hypoxia-inducible factor-1alpha and vascular endothelial growth factor reduced late rectal injury induced by irradiation. J Radiat Res. 2009;50(3):233-239. [DOI] [PubMed] [Google Scholar]
- 167. Zhang M, Kleber S, Röhrich M, et al. Blockade of TGF-β signaling by the TGFβR-I kinase inhibitor LY2109761 enhances radiation response and prolongs survival in glioblastoma. Cancer Res. 2011;71(23):7155-7167. [DOI] [PubMed] [Google Scholar]
- 168. Vanpouille-Box C, Diamond JM, Pilones KA, et al. TGFβ is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 2015;75(11):2232-2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Sharma MD, Baban B, Chandler P, et al. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest. 2007;117(9):2570-2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ.. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res. 2017;77(24):6795-6811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Cheong JE, Sun L.. Targeting the IDO1/TDO2-KYN-AhR pathway for cancer immunotherapy - challenges and opportunities. Trends Pharmacol Sci. 2018;39(3):307-325. [DOI] [PubMed] [Google Scholar]
- 172. Liu M, Li Z, Yao W, et al. IDO inhibitor synergized with radiotherapy to delay tumor growth by reversing T cell exhaustion. Mol Med Rep. 2020;21(1):445-453. 10.3892/mmr.2019.10816. [DOI] [PubMed] [Google Scholar]
- 173. Ladomersky E, Zhai L, Lenzen A, et al. IDO1 inhibition synergizes with radiation and PD-1 blockade to durably increase survival against advanced glioblastoma. Clin Cancer Res 2018;24(11):2559-2573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Chen B, Alvarado DM, Iticovici M, et al. Interferon-induced IDO1 mediates radiation resistance and is a therapeutic target in colorectal cancer. Cancer Immunol Res. 2020;8(4):451-464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Johnson TS, Aguilera D, Al-Basheer A, et al. Results of the NLG2105 phase I trial using the IDO pathway inhibitor indoximod, in combination with radiation and chemotherapy, for children with newly diagnosed DIPG. Ann Oncol. 2019;30:xi38. [Google Scholar]
- 176. Kim K-J, Lee HW, Seong J.. Combination therapy with anti-T-cell immunoglobulin and mucin-domain containing molecule 3 and radiation improves antitumor efficacy in murine hepatocellular carcinoma. J Gastroenterol Hepatol. 2021;36(5):1357-1365. [DOI] [PubMed] [Google Scholar]
- 177. Kim JE, Patel MA, Mangraviti A, et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin Cancer Res. 2017;23(1):124-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Nishiga Y, Drainas AP, Baron M, et al. Radiotherapy in combination with CD47 blockade elicits a macrophage-mediated abscopal effect. Nat Cancer 2022;3(11):1351-1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Li J, McCloskey M, Jennings C, et al. Antibody radioconjugates in combination with CD47 immunotherapy improves antitumor efficacy in preclinical models of solid tumors. J Nuclear Med. 2022;63(suppl 2):2574. [Google Scholar]
- 180. Liang J, Da Silva A, Han C, et al. Biology-guided radiotherapy for lung SBRT reduces planning target volume and organs at risk doses. Int J Radiat Oncol Biol Phys. 2019;105(1):S254. [Google Scholar]
- 181. Yang J, Yamamoto T, Thielemans K, Mazin S, Graves E, Keall P.. SU-E-J-156: a feasibility study for real-time tumor tracking using positron emission tomography (PET). Med Phys. 2011;38(6Part9):3479. [Google Scholar]
- 182. Yang K, Han W, Jiang X, et al. Zinc cyclic di-AMP nanoparticles target and suppress tumours via endothelial STING activation and tumour-associated macrophage reinvigoration. Nat Nanotechnol. 2022;17(12):1322-1331. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analyzed in support of this research.