Abstract
Lately, the success of ICIs has drastically changed the landscape of cancer treatment, and several immune checkpoint inhibitors (ICIs) have been approved by the US Food and Drug Administration (FDA) for advanced non-small cell lung cancer (NSCLC). However, numerous patients are resistant to ICIs and require additional procedures for better efficacy results. Thus, combination therapy is urgently needed to strengthen the anti-tumor immunity. A variety of preclinical and clinical studies combining ICIs with radiotherapy (RT) have demonstrated that the combination could induce synergistic effects, as RT overcomes the resistance to ICIs. However, the underlying mechanism of the synergistic effect and the optimal arrangement of the combination therapy are indecisive now. Hence, this review was conducted to provide an update on the current clinical trial results and highlighted the ongoing trials. We also discussed the optimal parameters in clinical trials, including radiation dose, radiation fractionation, radiation target field, and sequencing of combination therapy. In this review, we found that combination therapy showed stronger anti-tumor immunity with tolerable toxicities in clinical trials. However, the best combination mode and potential biomarkers for the target patients in combination therapy are still unclear.
Keywords: immunotherapy, radiotherapy, combination, immune checkpoints, non-small cell lung cancer
Introduction
Lung cancer is one of the leading cause of deaths worldwide,1 with more than 85% of lung cancers being non-small cell lung cancers (NSCLC). Treatment approaches and prognosis of NSCLC patients have been evaluated based on the TNM staging system of the American Joint Committee on Cancer (AJCC).2 Surgery or stereotactic body radiation therapy (SBRT) offers prominent local control of tumors and is considered safe for patients with stage I or II NSCLC.3 However, patients with early stage NSCLC still have a higher risk of distant metastasis, and patients with resectable stage III NSCLC usually undergo surgery, while unresectable patients receive concurrent chemoradiation (cCRT) treatment. It has been shown that patients with stage III NSCLC have a lower 5-year OS rate of 15–20% after undergoing traditional curative therapies.4 However, previous studies focusing on increasing radiation dose or involving induction or consolidation chemotherapy failed to show any evidence of extra clinical benefit.4–6 Thus, improving local control and preventing metastatic spread are still significant challenges in stage III NSCLC patients. It has been reported that oncogene-driven stage IV NSCLC patients are stable within 1–2 years after treatment of tyrosine kinase inhibitors (TKIs),7 but patients with advanced NSCLC who had not received positive oncogenic drivers demonstrated poor prognosis with conventional therapies and warranted more powerful therapies.
Novel immunotherapeutic approaches aim to enhance the immunity of the host, by gathering together immune cells, and getting these cells to recognize and then eliminate the cancer cells. The US Food and Drug Administration (FDA) has approved various immune checkpoint inhibitors (ICIs) for treatment of advanced NSCLC, including nivolumab, pembrolizumab, atezolizumab, and durvalumab.8 However, several patients showed no benefit from treatment with ICIs, and are classified as having primary resistance. Ultimately, most primary responders develop acquired resistance after treatment with ICIs, and their conditions progress. Hence, great efforts are being made to improve the efficacy of ICIs. Several studies have reported combinations of ICIs with other therapies, such as chemotherapy, radiotherapy (RT), targeted therapy, or other immunotherapies. RT can augment the benefit of ICIs, providing local control of primary tumors and strengthening immune responses even outside the targeted site, this is known as the “abscopal effect.”9
In this review, we summarize current clinical trials and discuss the optimal combination modes of ICIs and RT. We also outline challenges and future directions for immunotherapy and RT.
Immunotherapy: resistance mechanisms
Several processes are involved in immune system attack on tumor cells, including tumor recognition, antigen presentation, and T-cell activation.10 However, cancer cells can escape immune surveillance by interaction with immune cells. In attempts to ensure this does not occur, there is current interest in ICIs as therapeutics, in particular the anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor, anti-programmed death 1 (PD-1) inhibitor, and anti-programmed death ligand 1 (PD-L1) inhibitor. CTLA-4 on T-cells binds to peripheral membrane B7 on antigen presenting cells (APC), and inhibits T-cell activation.11 Similarly to the CTLA-4/B7 complex, PD-L1 on tumor cells attaches to PD-1 on T-cells. Then, the PD-1/PD-L1 compound releases inhibitory signals and acts in the effector phase of T-cell immune response.12
Primary resistance to immunotherapy is related to various tumor intrinsic and extrinsic factors (Table 1).13 Some tumor cells containing non-synonymous somatic mutations can be recognized by the immune cells as neoantigens. The lack of antigenic proteins is considered to be one of the most significant intrinsic factors.14 The advancement of next-generation sequencing (NGS) techniques has made possible comprehensive detection of tumor mutation burden (TMB). Moreover, deficient DNA mismatch repair (MMR) can provoke a higher level of genomic instability and tumor-specific mutation.15 Consequently, NSCLC patients with an absence of antigenic proteins, including proficient DNA MMR, lower TMB, and lower neoantigen load show inferior responses to ICIs and have poor progression-free survival (PFS).16 Meanwhile, primary resistance is related to the absence of antigen presentation machinery that is caused by downregulation of major histocompatibility complex class 1 (MHC-1).17,18
Table 1.
Mechanisms of immune resistance.
Primary resistance | |
Intrinsic factor | Absence of antigen proteins |
Absence of antigen presentation machinery | |
Extrinsic factor | PD-L1 expression levels |
Insufficient tumor-infiltrating lymphocytes | |
Severe exhaustion of T-cells | |
Immunosuppressive cells, immunosuppressive cytokines, and inhibitory checkpoints | |
Metabolic status | |
Alterations of several signaling pathways | |
Acquired resistance | Loss of functional T-cells |
Escape mutations |
The major extrinsic factors include immunoregulatory factors within the tumor microenvironment (TME), such as low PD-L1 expression levels, insufficient tumor-infiltrating lymphocytes (TILs), and severe exhaustion of T-cells.19 Besides, the immunosuppressive cells, immunosuppressive cytokines and inhibitory checkpoints involved in resistance to ICIs include regulatory T-cells (Tregs), Th2 cells, myeloid-derived suppressor cells (MDSCs), M2 macrophages, and T-cell immunoglobulin domain and mucin-3 (TIM-3).20 The metabolic status of the TME is also considered to be a crucial factor, with ICIs more efficacious in patients with higher glycolytic rates.21 Alterations in several signaling pathways (MAPK, PI3K, WNT, IFN, EGFR, or ALK signaling pathways) can affect the function of T-cells and response to ICIs.22 For example, oncogene-driven NSCLC patients had lower levels of TMB, exhibiting insufficient responses to ICIs.15,23 Although oncogene drivers upregulate expression of PD-L1, such expression levels were reduced after TKI treatment, generating resistance to ICIs.24,25 On the other hand, the resistance may be correlated with decreased cytotoxic CD8+ T-cells (CTLs) and enhanced T-cell exhaustion induced by the oncogenic signals.23,24 Several host-related factors are related to ICI resistance, such as age, smoking, background infection or chronic disease, and gut microbiota.26
The mechanism of acquired resistance might be correlated with loss of functional T-cells and development of escape mutations, such as mutations of Janus kinase (JAK1/2). JAK1/2 reduces presentation of tumor antigens through the interferon (IFN)-γ signaling pathway.19
RT: overcoming resistance to immunotherapy
RT is widely used in treatment of lung cancer and can provide great local tumor control. It causes random point mutations and double-stranded breaks in the DNA, increasing TMB and neoantigens.27 Cell death induced by radiation can occur by apoptosis, necrosis, autophagic death, mitotic catastrophe, or proliferative senescence.28 Radiation-induced cell death results in release of adenosine triphosphate (ATP), high mobility group box 1 protein (HMGB-1) and calrecticulin (CALR), and a rise in production of IFN-I.28–30 HMGB-1 binds to toll-like receptor-4 (TLR-4) and the HMGB-1/TLR-4 interaction is involved in tumor antigen progression and presentation.31 RT also promotes activation and maturation of dendritic cells (DCs) through the above signaling pathways.32 Hence, the immunogenicity of tumor cells is strengthened by increasing the tumor antigens, activating dendritic cells (DCs), and enhancing antigen cross-presentation to T-cells.29 The radiation-induced cell death is defined as immunogenic cell death (ICD) as it leads to increased recognition of cancer cells by T-cells, and thus an improved immune response.28,33 Radiation also increases expression of NK group 2 member D (NKG2D) ligand and first apoptosis signal (FAS) on tumor cells, and promotes recognition and elimination of cancer cells by T-cells and NK cells.34,35
On the other hand, the RT could modify the immunosuppressive TME. The DNA damage-induced kinase ABL1 amplifies expression of macrophage colony-stimulating factor 1 (CSF1) in response to radiation, resulting in proliferation of circulating MDSCs.36 These MDSCs subsequently enhance tumor infiltration.37 Radiation expands and activates CD8+ T-cells in draining lymph nodes and also improves recruitment of T-cells to tumor sites by upregulating chemokines, such as CXCL10 and CXCL16.38,39 Meanwhile, RT increases expression of vascular adhesion molecule-1 (VCAM-1) on endothelial cells and permits extravasation of T-cells.37 RT reprograms macrophages to secrete nitric oxide (NO) and leads to enhanced T-cell infiltration and vascular normalization.29 MHC-I expression is upregulated on tumor cells and improved tumor recognition.40 Expression of PD-L1 is also increased on cancer cells after RT and is dependent on IFN-γ produced by CD8+ T-cells.41,42 Hence, radiation could change tumor metabolism status and enhance immunological response.27
RT can overcome resistance to ICIs via complicated mechanisms and can convert non-immunogenic tumors into immunogenic ones, making logical a combination of RT and ICIs (Fig. 1).
Figure 1.
Radiation activates the host immune system. Step 1. Release of neoantigen: Neoantigen increasing; Step 2. Neoantigen presentation: ATP/HMGB-1/CALR/IFN-I from tumor cells; Step 3. Proliferation and activation of dendritic cells and T cells: ATP/HMGB-1/CALR/IFN-I and CXCL10/CXCL 16 from tumor cells; Step 4. Recruiting of T cells to irradiated or unirradiated tumors: CXCL10/CXCL16 from tumor cells, increasing of VCAM-1 on endothelial cells; Step 5. Infiltration of T cells to tumors: CSF-1 from tumor cells, MDSC increasing; Step 6. Recognization and killing of tumor cells: PD-L1/MHC-1/NKG2D/FAS increasing on tumor cells. Vascular normalization: NO from macrophages.
Current studies of combination therapy
Previous studies have provided evidence that ICIs can improve survival of patients with advanced NSCLC. Currently, there are various clinical trials investigating combinations of ICIs and RT. Tables 2–4 were generated by searching the ClinicalTrials.gov database using the search terms “radiation,” “immunotherapy,” “NSCLC,” “CTLA-4 inhibitors,” “PD-1 inhibitors,” “PD-L1 inhibitors,” and several variations. The results were then manually filtered for inclusion of studies that were conducted on ICIs and RT. The clinical trial results highlighted the advantages of combinations in patients at different stages.
Table 2.
Active clinical trials involving the use of both SBRT and immunotherapy in early stage NSCLC.
NCT number | Reference | Radiation | Sequencing | Radiation dose | Immunotherapy | Stage | Phases | Enrollment |
---|---|---|---|---|---|---|---|---|
NCT03383302 | 45 | SBRT | Sequential | 54 Gy/3 fx, 55 Gy//5 fx | Nivolumab | I–II | 1/2 | 31 |
NCT03110978 | 46 | SBRT | Concurrent | 50 Gy/4 fx, 70 Gy/10 fx | Nivolumab | I/IIA (T2N0M0) | 2 | 140 |
NCT03148327 | 47 | SBRT | Concurrent | 54 Gy/3 fx, 50 Gy/4 fx, 65 y/10 fx | Durvalumab | I/IIA (T2bN0M0) | 1/2 | 105 |
NCT03446547 | 48 | SBRT | Sequential | 3–4 fx (dose NM) | Durvalumab | I | 2 | 216 |
NCT02599454 | 49 | SBRT | Induction | 50 Gy/4 fx, 50 Gy/5 fx | Atezolizumab | I | 1 | 33 |
NCT03050554 | 50 | SBRT | Concurrent | 48 Gy/4 fx, 50 Gy/5 fx | Avelumab | I | 1/2 | 56 |
Fx, fractions; Gy, gray; NM, not mentioned; NSCLC, non-small cell lung cancer; SBRT, stereotactic body radiation therapy. Note: all trials used consolidation immunotherapy after the combination therapy which is not shown in this table.
Table 4.
Clinical trials involving use of both radiotherapy and immunotherapy in advanced NSCLC.
NCT number | Reference | Radiation | Sequencing | Radiation dose | Immunotherapy | Phases | Enrollment |
---|---|---|---|---|---|---|---|
NCT02239900a | 66 | SBRT | Sequential/concurrent | 50 Gy/4 fx, 60 Gy/10 fx, 20 Gy/5 fx | Ipilimumab | 1/2 | 120 |
NCT02221739 | 67 | RT | Concurrent | 30 Gy/5 fx, 28.5 Gy/3 fx | Ipilimumab | 2 | 27 |
NCT02318771 | 68 | RT | Sequential/concurrent | 8 Gy/1 fx, 20 Gy/5 fx | Pembrolizumab | 1 | 40 |
NCT02492568 | 69 | SBRT | Sequential | 24 Gy/3 fx | Pembrolizumab | 2 | 92 |
NCT02608385 | 70 | SBRT | Sequential | 3–5 fx (dose NM) | Pembrolizumab | 1 | 35 |
NCT03004183 | 71 | SBRT | Sequential | 30 Gy/5 fx | Pembrolizumab | 2 | 57 |
NCT03307759 | 72 | SBRT | Sequential/induction | NM | Pembrolizumab | 1 | 32 |
NCT03245177 | 73 | TRT | Induction | 60–66 Gy/30–32 fx | Pembrolizumab | 1 | 25 |
NCT03368222 | 74 | SBRT | Induction | 30 Gy/3 fx, 54 Gy/3 fx | Pembrolizumab | 1 | 24 |
NCT03436056 | 75 | SBRT | Induction | 30 Gy/3 fx, 54 Gy/3 fx | Pembrolizumab | 1 | 24 |
NCT02444741a | 76 | SBRT/HFRT | Concurrent/induction | 50 Gy/4 fx, 45 Gy/15 fx | Pembrolizumab | 1/2 | 104 |
NCT02587455 | 77 | RT | Concurrent | NM | Pembrolizumab | 1 | 48 |
NCT02658097 | 78 | RT | Concurrent | 8 Gy/1 fx | Pembrolizumab | 2 | 48 |
NCT02831933 | 79 | SBRT | Sequential | 30 Gy/5 fx | Nivolumab | 2 | 25 |
NCT03223155 | 80 | TRT | Sequential/concurrent | 3–5 fx(dose NM) | Nivolumab/ipilimumab | 1 | 80 |
NCT03511391 | 81 | SBRT | Induction | 24 Gy/3 fx | Nivolumab/pembrolizumab | 2 | 97 |
NCT03044626 | 82 | RT | Concurrent | 20 Gy/5 fx | Nivolumab | 2 | 130 |
NCT03224871 | 83 | HFRT | Concurrent | 24 Gy3 fx | Nivolumab/pembrolizumab | 1 | 30 |
NCT03176173 | 84 | IGRT | Concurrent | 10 fx (dose NM) | Nivolumab/pembrolizumab/atezolizumab | 2 | 85 |
NCT03035890 | 85 | HFRT | Concurrent | 24–45 Gy/3 fx, 30–50 Gy/5 fx | Nivolumab/pembrolizumab/atezolizumab | NM | 33 |
NCT03313804 | 86 | SBRT/3D-CRT | Concurrent | SBRT: BED > 100 Gy, 3D-CRT: 30 Gy | Nivolumab/pembrolizumab/atezolizumab | 2 | 57 |
NCT03391869 | 87 | RT | Induction | NM | Ipilimumab+nivolumab | 3 | 270 |
NCT03168464 | 88 | RT | Concurrent | 30 Gy/5 fx | Ipilimumab+nivolumab | 1/2 | 45 |
NCT03431948 | 89 | SBRT | NM | 30–50 Gy | Nivolumab+ urelumab/cabiralizumab | 1 | 60 |
NCT03509584 | 90 | HFRT | NM | 24 Gy/3 fx | Nivolumab (+ipilimumab) | 1 | 24 |
NCT02400814 | 91 | SBRT | Sequential/induction/concurrent | 5 fx (dose NM) | Atezolizumab | 1 | 45 |
NCT02463994 | 92 | HIGRT | Sequential | NM | Atezolizumab | 1 | 12 |
NCT03050060 | 93 | HIGRT | Induction | NM | Atezolizumab | 2 | 120 |
NCT03275597 | 94 | SBRT | Sequential | 30–50 Gy/5 fx | Durvalumab+tremelimumab | 1b | 180 |
NCT02888743 | 95 | RT | Induction | NM | Durvalumab+tremelimumab | 2 | 180 |
NCT03212469 | 96 | SBRT | Induction | NM | Durvalumab+tremelimumab | 1/2 | 40 |
NCT02639026 | 97 | HFRT | Concurrent | 24 Gy/3 fx, 17 Gy/1 fx | Durvalumab+tremelimumab | 1 | 30 |
Fx, fractions; Gy, gray; HFRT, hypofractionated radiotherapy; HIGRT, hypofractionated image-guided radiotherapy; IGRT, image-guided Radiotherapy; NM, not mentioned; NSCLC, non-small lung cancer; RT, radiation therapy; SBRT, stereotactic body radiation therapy; TRT, thoracic radiotherapy; WFRT, wide-field radiation therapy; 3D-CRT, 3D conformal radiotherapy. Note: all trials used consolidation immunotherapy after the combination therapy which is not shown in this table.
aTrials with reported results.
Surgery or SBRT demonstrated outstanding local control in patients with early stage NSCLC, with a 5-year local control rate of 92% in stage IA NSCLC patients and of 73% in stage IB and IIA NSCLC patients.43 However, there is also a probability of relapse or metastasis in early stage NSCLC patients and hence improved treatments are required. Currently, there are six ongoing clinical trials on the combination of SBRT with ICIs, as presented in Table 2.44–49 The dose of SBRT ranges from 48 to 70 Grey (Gy) [3–10 fx (fractions)], and there is no consensus on administration with some trials administering SBRT before ICI administration and some after, or even concurrently with ICIs. We defined combination modes as 1. sequential therapy, where the patients received ICI after the final fraction of SBRT; 2. induction therapy, where the patients received SBRT after a few cycles of ICI; and 3. concurrent therapy, where the patients received SBRT and ICIs concurrently. The ICIs used in the clinical trials included nivolumab, durvalumab, atezolizumab, and avelumab, and patients received consolidation ICI after immunoradiation. All the trials are in phase I or II and are still recruiting patients without any published results (Table 2 and Fig. 2).
Figure 2.
Sequencing modes of combination therapy. Sequential therapy: patients receiving ICI after CRT; Induction therapy: patients receiving CRT after a few cycles of ICI; Concurrent therapy: patients receiving CRT and ICI concurrently.
In the phase I clinical trial KEYNOTE-001 (NCT01295827), 38 (39%) of 97 patients with locally advanced or metastatic NSCLC received extracranial RT, and 24 (25%) patients among them received thoracic RT before the first cycle of pembrolizumab. A secondary analysis of this trial compared the PFS and OS of patients with or without prior RT before the first cycle of pembrolizumab.50 Patients who had previously received extracranial RT had a significantly longer median PFS [6.3 vs. 2.0 months; hazard ratio (HR): 0.50 (95% CI: 0.30–0.84), P = 0.0084] and OS [11.6 vs. 5.3 months; HR: 0.59 (95% CI: 0.36–0.96), P = 0.034]. The study also noted that patients with prior thoracic RT had a higher risk of treatment-related pulmonary toxicity [13% (3/24) vs. (1/73) 1%], but had similar incidence of grade 3 or worse pulmonary toxicities (one patient in each group). The tolerable safety profile and promising efficacy in this trial highlighted the potential of combination therapy and the requirement for carefully monitoring the toxicity in patients.
Resectable locally advanced patients, typically stage IIIA (AJCC), normally undergo initial surgery before or after chemotherapy and RT. The latest clinical trial NCT02259621 reported use of neoadjuvant nivolumab (anti-PD-1inhibitor) in resectable NSCLC patients (stages I, II and III).51 The first dose of nivolumab was given to the patients about 4 weeks before the planned surgery. Patients were given conventional adjuvant chemotherapy or RT if necessary. Neoadjuvant nivolumab showed tolerable adverse effects (AEs), with only one grade 3 pneumonia and no grade 4 or 5 AEs among 21 patients, and there was no delays to any surgeries. The median interval between the second dose of nivolumab and surgery was 18 days (range 11-29 days). After neoadjuvant nivolumab, pathological down-staging occurred in eight patients (40%), and shrinkage of the tumors made the surgeries safer. The major pathologic response (MPR), defined as ≤10% residual viable tumor cells in surgical specimens viewed with hematoxylin and eosin staining, was seen in nine patients (45%). There are three other clinical trials regarding combination of ICIs and RT or chemoradiotherapy (CRT) before undergoing the surgery in resectable stage III NSCLC patients52–54 (Table 3). Another trial involves administration of adjuvant pembrolizumab after neoadjuvant concurrent chemoradiotherapy (cCRT) and surgery55 (Table 3). The adjuvant usage of ICIs after surgery is controversial, and further clinical trials are required to provide sufficient evidence for clinical decision-making.
Table 3.
Combination of radiotherapy and immunotherapy in stage III NSCLC.
NCT number | Reference | Radiation | Sequencing | Radiation dose | Immunotherapy | Stage | Phases | Enrollment |
---|---|---|---|---|---|---|---|---|
Resectable stage III NSCLC | ||||||||
NCT03217071 | 53 | SBRT | Induction | 12 Gy/1 fx | Pembrolizumab | I–IIIA | 2 | 40 |
NCT02987998 | 54 | cCRT | Concurrent | 45 Gy/25 fx | Pembrolizumab | IIIA | 1 | 20 |
NCT03053856 | 56 | cCRT | Adjuvant | 44 Gy//22 fx | Pembrolizumab | IIIA | 2 | 37 |
NCT03237377 | 55 | TRT | Concurrent | 45–50 Gy/25 fx | Durvalumab (+tremelimumab) | IIIA | 2 | 32 |
Unresectable stage III NSCLC | ||||||||
NCT02768558 | 60 | cCRT | Sequential | 60 Gy | Nivolumab | III | 3 | 13 |
NCT03285321 | 61 | cCRT | Sequential | 59.4–66.6 Gy | Nivolumab (+ipilimumab) | III | 2 | 108 |
NCT02434081a | 62 | cCRT | Concurrent | NM | Nivolumab | III | 2 | 78 |
NCT02525757a | 58 | cCRT | Sequential/concurrent | 60–66 Gy/30–32 fx | Atezolizumab | III | 2 | 52 |
NCT03102242 | 63 | cCRT | Induction | 60 Gy/30 fx | Atezolizumab | III | 2 | 63 |
NCT02125461a | 57 | cCRT | Sequential | 54–66 Gy | Durvalumab | III | 3 | 713 |
NCT03509012 | 64 | cCRT | Concurrent | NM | Durvalumab | III | 1 | 300 |
NCT02343952a | 59 | cCRT | Concurrent | 59.4–66.6 Gy | Pembrolizumab | III | 2 | 93 |
NCT02621398 | 65 | cCRT | Concurrent | 30 fx (dose NM) | Pembrolizumab | II–IIIB | 1 | 30 |
cCRT, concurrent chemoradiotherapy; Fx, fractions; Gy, gray; NM, not mentioned; NSCLC, non-small cell lung cancer; RT, radiation therapy; SBRT, stereotactic body radiation therapy; TRT, thoracic radiotherapy. Note: trials of resectable stage III NSCLC received surgery after combination therapy and trials of unresectable stage III NSCLC used consolidation immunotherapy after the combination therapy which is not shown in this table.
a Trials with reported results.
Conventional treatment for patients with unresectable locally advanced NSCLC is curative CRT. In the PACIFIC study (NCT02125461), a phase III study for patients with stage III unresectable lung cancer, patients received consolidation durvalumab (anti-PD-L1 inhibitor) until disease progression (up to 12 months) after initial cCRT.56 Durvalumab was given for no more than 6 weeks after CRT, with results showing an improvement of 11.6 months in median PFS when compared with the control group (17.2 vs. 5.6 months). These results accelerated approval of durvalumab by the FDA as consolidation therapy in stage III unresectable lung cancer patients after cCRT. The latest report even showed that the durvalumab group had a better OS (2-year OS rate: 66.3% vs. 55.6%).56 The phase II trial DETERRED (NCT02525757) on another anti-PD-L1inhibitor, atezolizumab, is an ongoing trial conducted in unresectable stage III NSCLC patients.57 After a resting period of 3-4 weeks followed by definitive cCRT, patients received atezolizumab in addition to chemotherapy for two cycles. Later, the patients received extra consolidation treatment with atezolizumab for 12 months. Of the seven patients, two showed progression after six and eight doses of atezolizumab. The LUN 14-179 (NCT02343952) study examined use of pembrolizumab in unresectable stage III NSCLC patients, reporting a similar median PFS of 15.4 months with a resting period between pembrolizumab and CRT of 4-8 weeks.58 The 1-year OS rate was 80.5% and the 2-year OS rate was 68.7%. A comparable trial, RTOG 3505 (NCT02768558), on nivolumab in unresectable stage III NSCLC patients is ongoing, but no results have yet been reported.59 The combination modes of CRT and ICIs are summarized as follows: 1. sequential therapy: patients receiving ICI after CRT; 2. induction therapy: patients receiving CRT after a few cycles of ICI; and 3. concurrent therapy: patients receiving CRT and ICI concurrently60–64 (Table 3 and Fig. 2).
In patients with advanced NSCLC, usage of combinations of ICIs and RT is more frequent and more complicated. There are several ongoing clinical trials using various ICIs, such as ipilimumab, tremelimumab, pembrolizumab, nivolumab, atezolizumab, and durvalumab. Different combination modes, including induction, sequential, or concurrent therapies have been used (Fig. 2). The clinical trials also adopted various radiation technologies, such as image-guided radiotherapy (IGRT), intensity modulated radiation therapy (IMRT), hypofractionated radiotherapy (HFRT), stereotactic body radiation therapy (SBRT), and wide-field radiation therapy (WFRT)65–96 (Table 4). The clinical trial NCT02239900 on ipilimumab with concurrent or sequential RT reported that three (10%) of 31 advanced NSCLC patients exhibited partial response (PR) outside the radiation field.65 For the 19 patients in NCT02444741, six patients (32%) showed PR, seven patients (36%) experienced stable disease, and six patients (32%) progressed after a combination of RT and pembrolizumab.75
With the rapidly increasing number of clinical trials regarding the combination therapy, we look forward to further discussions on their results.
Toxicities of combination therapy
Among studies reporting toxicity data, four trials are about unresectable stage III NSCLC and two trials are conducted with stage IV NSCLC patients. In the updated analyses of the PACIFIC study, grade 3 or 4 AEs were observed in 30.5% of patients who received sequential cCRT and durvalumab, and in 26.1% of patients in the placebo group.56 As the most frequent AE leading to treatment discontinuation in this trial, pneumonitis occurred in 32.8% (156/475) of patients. Among the 475 patients, 16 patients experienced grade 3–4 pneumonitis and five patients experienced grade 5 pneumonitis.56 The DETERRED study was designed for sequential and concurrent parts, but only the results of the sequential group have been published. The latest abstract published at the 2018 American Society of Clinical Oncology (ASCO) annual meeting, reported that one patient developed grade 2 radiation-induced pneumonitis which was manageable with steroids.57 The current combination of cCRT and pembrolizumab in LUN 14-179 reported 11 patients with grade 2 pneumonitis, five with grade 3–4 pneumonitis, and one pneumonitis-associated death from a total of 93 patients. The median interval for pneumonitis occurrence is 8.4 weeks.58 However, for the first 21 patients recruited in the trial ETOP NICOLAS, the most frequent AEs are fatigue and anemia and no pneumonitis grade ≥3 has been observed. The observation period is only 3 months after RT, so long-term toxicities are not available.61 The trial NCT02239900 on ipilimumab reported four patients with grade 3 toxicity from 13 patients in the concurrent group, and eight out of 22 patients in the sequential group. No patient experienced grade > 1 pneumonitis in this trial.65 As for current combination of pembrolizumab, NCT02444741 reported one grade 3 pneumonitis in 19 patients75 (Table 5). According to the reported data, ICIs remain tolerable when combined with RT in NSCLC patients.
Table 5.
Toxicities reported in the combination therapy.
NCT number | Reference | Radiation | Sequencing | Immunotherapy | Stage | Any grade AEs | Grade ≥3 AEs | Any grade pneumonitis |
---|---|---|---|---|---|---|---|---|
NCT02125461a | 57 | cCRT | Sequential | Durvalumab | III | 460/475 | 145/475 | Grade ≤2 (136/475); Grade 3–4 (16/475); Grade 5 (4/475) |
NCT02525757a | 58 | cCRT | Sequential | Atezolizumab | III | 3/7 | 2/7 | Grade 2 (1/7) |
NCT02343952a | 59 | cCRT | Concurrent | Pembrolizumab | III | NM | NM | Grade 2 (11/93),Grade 3–4 (5/93), pneumonitis associated death (1/93) |
NCT02434081a | 62 | cCRT | Concurrent | Nivolumab | III | NM | NM | No pneumonitis grade ≥3 |
NCT02239900a | 66 | SBRT | Concurrent | Ipilimumab | IV | 6/13 | 4/13 | No pneumonitis grade > 1 |
NCT02239900a | 66 | SBRT | Sequential | Ipilimumab | IV | 9/22 | 8/22 | No pneumonitis grade > 1 |
NCT02444741a | 76 | SBRT/HFRT | Concurrent | Pembrolizumab | IV | 11/19 | 3/19 | Grade 3 (1/19) |
AEs, adverse events; cCRT, concurrent chemoradiotherapy; Fx, fractions; Gy, gray; HFRT, hypofractionated radiotherapy; NM, not mentioned; NSCLC, non-small cell lung cancer; SBRT, stereotactic body radiation therapy.
a Trials with reported results.
Challenges in combination therapy
The traditional mechanism of RT includes four principles (“four Rs”): repair, reassortment, repopulation, and reoxygenation.97 Currently, the immunogenic effect of RT is noted, and the combination therapy is expected to deliver better clinical efficacy. However, there are still many questions regarding the optimal combinations to maximize the synergistic effects.
RT dose and fractionation
In the design of the current clinical trials, a wide range of RT dose and fractionation schedules are applied. The appropriate selection of RT dose and fractionation to elicit an effective anti-tumor immune response safely is unknown.98
Previous preclinical breast and colon cancer models suggest that 24 Gy/3 fx or 30 Gy/5 fx is better than 20 Gy/1f when combined with anti-CTLA4 inhibitor for metastatic lesions.99 Work on a preclinical model of prostate cancer also suggested that multi-fraction radiation is superior when compared to a single dose of radiation.100 Furthermore, in preclinical studies of lung cancer and melanoma models, ablative hypofractionated radiotherapy (AHFRT) (23 Gy/2 fx) showed better local control of primary tumors and a stronger systemic anti-tumor effect (abscopal effect) than conventional fractionated radiotherapy (CFRT) of 36 Gy/9 fx with the same biological equivalent dose (BED).101 At the cellular level, AHFRT suppressed recruitment of MDSCs into tumors and decreased expression of PD-L1, which then activated cytotoxic CD8+ T lymphocytes. AHFRT combines with anti-PD-L1 inhibitor to trigger a more effective anti-tumor effect, showing significant improvement in terms of tumor control and mouse survival.
RT target region
T-lymphocytes are radiosensitive. Radiation can not only damage TILs, but also affect T-cells in the peripheral blood that transit through the irradiated field.102 Larger radiation fields expose more lymphocytes to RT, which may in turn exhaust the T-cells. Methods to reduce RT-induced lymphopenia include reduction in the volume of the radiation field using highly conformal techniques such as SBRT and HFRT. A comment on keynote-001 trial even reported that it might not be necessary to include the entire tumor during RT.50,103 This is because RT of only a part of cancer might be sufficient to trigger the immune response.101 There are some clinical trials in which only a part of the large primary tumor (NCT02608385, NCT03217071, NCT03368222, NCT03436056) has been irradiated52,69,73,74 (Table 6). However, another report revealed that when RT is combined with ICI, it is necessary to avoid irradiation of the lymph node region, as this can affect generation of stronger immune responses.104 As it is common practice to irradiate the draining lymph nodes in NSCLC, there is a need for further research into this discrepancy in conclusions.
Table 6.
Different design of radiation field in clinical trials.
NCT number | Reference | Radiation | Sequencing | Immunotherapy | Radiation lesion enrollment |
---|---|---|---|---|---|
NCT02608385 | 70 | SBRT | Sequential | Pembrolizumab | Partially for large tumor or all lesions for oligometastatic patients |
NCT03217071 | 53 | SBRT | Induction | Pembrolizumab | 50% of primary tumor |
NCT03368222 | 74 | SBRT | Induction | Pembrolizumab | Part of a lung lesion |
NCT03436056 | 75 | SBRT | Induction | Pembrolizumab | Part of a lung lesion |
NCT02444741a | 76 | SBRT/WFRT | Concurrent/induction | Pembrolizumab | Primary tumor: 50 Gy/4 fx or 45 Gy/15 fx and metastatic lesion: 5–10 Gy |
NCT03223155 | 80 | TRT | Sequential/concurrent | Nivolumab/ipilimumab | 2–4 lesions |
NCT03511391 | 81 | SBRT | Induction | Nivolumab/pembrolizumab | Maximally 3 lesions |
NCT03431948 | 89 | SBRT | NM | Nivolumab+ urelumab/cabiralizumab | Metastatic lesion(s) |
NCT03275597 | 96 | SBRT | Sequential | Durvalumab+tremelimumab | All lesion |
NM, not mentioned; SBRT, stereotactic body radiation therapy; TRT, thoracic radiotherapy; WFRT, wide-field radiation therapy.
aTrials with reported results.
Several previous clinical trials used ICIs and single lesion radiation, but failed to provoke the abscopal effect and clinical benefits.105,106 In his latest review, Chang presented an opposite opinion.107 Firstly, that multisite radiation can induce more neoantigen and functional T-cells than single lesion radiation and can modulate the immunosuppressive TME. Secondly, the extent of anti-tumor immunity induced by radiation is not the same when irradiating different sites. Analysis of phase I trial NCT02239900 showed that irradiation of liver metastases in NSCLC patients caused stronger anti-tumor immunity than irradiation of pulmonary metastases.65 Hence, different organs are not equally immunogenic and irradiation of multisites can increase the probability of successful induction of anti-tumor response. Thirdly, multisite radiation can reduce the tumor burden and enhance efficacy of ICIs. In the subgroup analysis of a phase III trial NCT00861614, the prostate patients with smaller disease burden or who received irradiation to all lesions demonstrated good clinical benefits with combination therapy of ipilimumab and RT.105 Furthermore, in a preclinical model of melanoma, tumor burden was found to be associated with T-cell reinvigoration. The T-cell invigoration to tumor burden ratio was directly proportional to the PFS after the immunotherapy.108 Based on these preclinical and clinical studies, the review by Chang suggested enhancing the limited efficacy of combination therapy by delivering RT to as much of the tumor burden as could be safely irradiated.107 Compared with numerous clinical trials examining radiation of only a single lesion, there are only six clinical trials using multiple site radiation. The NCT03223155, NCT03431948, and NCT03511391 trials were designed to irradiate more than one lesion79,80,88 (Table 6). Clinical trial NCT02444741 was designed to irradiate metastatic lesions with low doses ranging from 5 to 10 Gy and to irradiate the primary tumor at a dose of 45–50 Gy/5 fx.73 The NCT02608385 and NCT03275597 trials were designed to conduct SBRT on all lesions for oligometastatic patients69,93 (Table 6).
RT sequencing
The sequencing of each ingredient is critical for the efficacy of combination therapy. As different ICIs affect different phases of the immune response and different immune cells, the sequencing must be carefully designed to produce the greatest synergistic effect. However, the optimum sequencing is as yet unknown, although concurrent therapy appeared to be superior to sequential therapy in a preclinical colorectal cancer model.109 The preclinical study reported that administration of an anti-PD-1 inhibitor on day one or five of RT gave better results than administration 1 week after RT.109 A similar phenomenon occurred when a combination of anti-CTLA-4 inhibitor and HFRT (20 Gy/1f) was administered in preclinical colorectal cancer models.110 The anti-CTLA-4 inhibitor showed most efficient results when given before the radiation.110 The difference between preclinical and clinical studies highlights the need to conduct studies to compare the concurrent mode with the sequential mode and the induction mode.
A retrospective analysis of 758 patients who received an ICI (anti-CTLA4 and/or anti-PD1/anti-PDL1) demonstrated that patients who received concurrent therapy had a better OS. Furthermore, the OS was longer when patients received induction ICI for more than 30 days before RT when compared with those receiving ICI within 30 days of RT (20 vs. 11 months).111 There is also a phase II clinical trial, NICOLAS (NCT02434081), regarding involvement of timepoint of nivolumab concurrently with cCRT in patients with unresectable locally advanced NSCLC.61 The results published at the 2018 ASCO annual meeting provided evidence that combining nivolumab and RT concurrently in cycle two of chemotherapy was safe and tolerable.
In contrast, the PACIFIC study reported that sequential use of durvalumab from several weeks (up to 6 weeks) after cCRT improved median PFS of 11.6 months when compared with the control group.56 However, the analysis suggested that beginning durvalumab usage within 14 days of completing cCRT seemed to have greater PFS than starting it after 14 days (up to 42 days).112 Hence, the synergistic effect is not only correlated with the sequencing, but also with the resting period between RT and ICIs. Various trials with different combination modes are ongoing, and the best combination that triggers a powerful immune response without exhausting the T-cells will be confirmed in the future.
Combination with other immunotherapies
Other non-ICI immunotherapies, such as interleukin-2 (IL-2), granulocyte-macrophage colony stimulating factor (GM-CSF), and cancer vaccines have been used experimentally to gather further information to improve the efficacy of combination therapy.
IL-2 is a cytokine produced by T-cells that promotes T-cell growth. It has been approved by the FDA for treatment of metastatic renal cell carcinoma (RCC) and metastatic melanoma.113 According to a phase I study that combined high dose IL-2 with SBRT for metastatic RCC and melanoma patients, combination therapy resulted in better tumor regression than IL-2 alone. The better efficacy is induced by increased activation of effector memory T-cells.114 Although IL-2 has shown promising efficacy, it has apparent toxicities, including flu-like symptoms, vascular leak syndrome (VLS), pulmonary edema, hypotension, and heart toxicities.115 Currently, a phase I pilot study NCT03224871 is being designed to add IL-2 to concurrent use of nivolumab or pembrolizumab and RT (24 Gy/3 fx).78
GM-CSF is a cytokine that can stimulate tumor cell migration from distant sites, promote maturation of DCs, and enable cross-presentation of tumor cell antigens within TME.116,117 It is normally produced by macrophages, T-cells, and fibroblasts and is secreted from the tumor cells after radiation.117 A proof-of-principle pilot study enrolled 41 patients with different cancer types receiving RT (35 Gy/10 fx) to a metastatic lesion and 14 days of GM-CSF. Among these patients, 11 (26.8%) exhibited a clinical response distant to the radiation site.118 The addition of GM-CSF or its gene vaccine (GVAX) to the combination therapy is being investigated in pancreatic cancer and other cancers (NCT02663440, NCT02677155, NCT02648282).119–121
As a critical approach to strengthen the anti-tumor immunity, cancer vaccines present tumor antigens on recombinant viral vectors. A phase II trial in 33 prostate cancer patients with prostate-specific antigen (PSA) ≥10 or a Gleason score of ≥7 or stage T2b to T3 receiving PSA-vaccine (adenoviral vector-mediated herpes simplex virus thymidine kinase, AdHSV-tk) and IMRT has been conducted.122 After treatment, a sustained long-term PSA-specific T-cell response occurred and persisted for up to 8–12 months. Other trials reported that combination of cancer vaccines with CRT showed no dose-limiting toxicities in pancreatic cancer (NCT00638612) and malignant gliomas (NCT00751270).123,124 There are various cancer vaccines applied in NSCLC, including BLP-25 anti-mucin 1 (MUC1), belagenpumatucel-L, TG4010 (modified virus of Ankara–MUC1–IL-2), CIMAvax epidermal growth factor (EGF), melanoma antigen-encoding gene A3 (MAGEA3), GM.CD40L, and NYESO.125 Currently, there are two ongoing clinical trials in NSCLC patients, NCT03004183 and NCT02831933, using an oncolytic virus (adenovirus-mediated expression of herpes simplex virus thymidine kinase, ADV/HSV-tk) together with combination therapy of SBRT (30 Gy/5 fx) and ICI.70,78
Patient selection biomarkers
Selection of suitable patients for combination therapy is crucial. Previous studies have indicated that PD-L1 expression, tumor-infiltrating lymphocytes (TILs), TMB, neoantigens, and DNA MMR deficiency are candidate biomarkers for ICIs. However, currently, there are no validated biomarkers for responses to combination therapy. In the PACIFIC trial, the unstratified HR for disease progression or death is significantly different between the durvalumab group and control group in people with >1% PD-L1 expression (HR: 0.46, 95% CI: 0.33–0.61) and the HR showed no significant differences in people with <1% PD-L1 expression.56 On the other hand, patients with higher TMB, particularly those with MMR deficiency could be particularly sensitive to radiotherapy, causing a stronger anti-tumor immune response.126 Furthermore, patients with lower tumor burden had better response with combination therapy.105 When selecting the patients, the tumor doubling time should be evaluated and patients with a short tumor doubling time may not be suitable for combination therapy. Other possible factors for patient selection are unknown and may involve multiple levels, including genome, transcriptome, proteome, immunome, or microbiome.
Conclusions
Emerging preclinical and clinical data support combination of ICIs with RT. The results of ongoing trials have reported the safety and efficacy data of combination therapy; however, the optimal arrangement of the two therapies has not been identified, especially the design of RT. Firstly, the studies show that multi-fraction is better than single-fraction, and AHFRT is better than CFRT. Secondly, irradiation of more immunogenic metastatic lesions rather than the primary tumor may bring better clinical benefits because of immunogenic differences between different organs. Otherwise, the importance of accurate gross tumor volume (GTV) has been weakened, as the irradiating part of the tumor still could trigger anti-tumor immunity. Thirdly, studies on sequencing of combination therapy have controversial results. Fourthly, development of biomarkers to predict the response to combination therapy would help to identify the target patients who are most likely to benefit from treatment, which would improve the cost-effectiveness of ICIs. More studies are needed to understand the synergistic effect of the combination therapy. Finding the best way to maximize the immunity of hosts is the next step in the area of immuno-oncology.
Acknowledgements
This work was supported by the National Natural Science Foundation of China (grant numbers: 81872478) and Sichuan Provincial Research Foundation for Basic Research (grant numbers: 2018JY0550).
Conflict of interest statement
None declared.
References
- 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin 2017;67:7–30. [DOI] [PubMed] [Google Scholar]
- 2. Goldstraw P, Chansky K, Crowley J, et al. The IASLC lung cancer staging project: proposals for revision of the TNM stage groupings in the forthcoming (eighth) edition of the TNM classification for lung cancer. J Thorac Oncol 2016;11:39–51. [DOI] [PubMed] [Google Scholar]
- 3. Chang JY, Liu YH, Zhu Z, et al. Stereotactic ablative radiotherapy: A potentially curable approach to early stage multiple primary lung cancer. Cancer 2013;119:3402–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Curran WJ Jr, Paulus R, Langer. et al. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst 2011;103:1452–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Bradley JD, Paulus R, Komaki R, et al. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): a randomised, two-by-two factorial phase 3 study. Lancet Oncol 2015;16:187–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hanna N, Neubauer M, Yiannoutsos C, et al. Phase III study of cisplatin, etoposide, and concurrent chest radiation with or without consolidation docetaxel in patients with inoperable stage III non-small-cell lung cancer: the Hoosier Oncology Group and U.S. Oncology. J Clin Oncol 2008;26:5755–60. [DOI] [PubMed] [Google Scholar]
- 7. Greenhalgh J, Dwan K, Boland A, et al. First-line treatment of advanced epidermal growth factor receptor (EGFR) mutation positive non-squamous non-small cell lung cancer. Cochrane Database Syst Rev 2016;28:918–89. [DOI] [PubMed] [Google Scholar]
- 8. Bianco A, Malapell U, Rocco D, et al. Targeting immune checkpoints in non small cell lung cancer. Curr Opin Pharmacol 2018:40:46–50. [DOI] [PubMed] [Google Scholar]
- 9. Formenti SC, Demaria S. Radiation therapy to convert the tumor into an in situ vaccine. Int J Radiat Oncol Biol Phys 2012;84:879–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Pardoll D, Allison J. Cancer immunotherapy: breaking the barriers to harvest the crop. Nat Med 2004;10:887–92. [DOI] [PubMed] [Google Scholar]
- 11. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56–61. [DOI] [PubMed] [Google Scholar]
- 12. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Pitt JM, Vétizou M, Daillère R, et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and-extrinsic factors. Immunity 2016;44:1255–69. [DOI] [PubMed] [Google Scholar]
- 14. Sharma P, Hu-Lieskovan S, Wargo JA, et al. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Festenstein H, Garrido F. MHC antigens and malignancy. Nature 1986;322:502–3. [DOI] [PubMed] [Google Scholar]
- 18. Ishizuka JJ, Manguso RT, Cheruiyot CK, et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 2019;565:43–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Wang Q, Wu X. Primary and acquired resistance to PD-1/PD-L1 blockade in cancer treatment. Int Immunopharmacol 2017;46:210–9. [DOI] [PubMed] [Google Scholar]
- 20. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell 2018;33:547–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Chang CH, Qiu J, O’Sullivan D, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 2015;162:1229–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Rizvi H, Sanchez-Vega F, La K, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti–programmed death-ligand 1 (PD-L1) blockade in patients with non–small-cell lung cancer profiled with targeted next-generation sequencing. J Clin Oncol 2018;36:633–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Miura Y, Sunaga N. Role of immunotherapy for oncogene-driven non-small cell lung cancer. Cancers (Basel) 2018;10:245–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Akbay EA, Koyama S, Carretero J, et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 2013;3:1355–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Ota K, Azuma K, Kawahara A, et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin Cancer Res 2015;21:4014–21. [DOI] [PubMed] [Google Scholar]
- 26. Sivan A, Corrales L, Hubert N, et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015;350:1084–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kordbacheh T, Honeychurch J, Blackhall F, et al. Radiotherapy and anti-PD-1/PD-L1 combinations in lung cancer: building better translational research platforms. Ann Oncol 2017;29:301–10. [DOI] [PubMed] [Google Scholar]
- 28. Golden EB, Pellicciotta I, Demaria S, et al. The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol 2012;2:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. van Gulijk M, Dammeijer F, Aerts JGJV, et al. Combination Strategies to Optimize Efficacy of Dendritic Cell-Based Immunotherapy. Front Immunol 2018;9:669–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Deng LF, Liang H, Xu M, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014;41:843–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Apetoh L, Ghiringehelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 2007;13:1050–9. [DOI] [PubMed] [Google Scholar]
- 32. Messmer D, Yang H, Telusma G, et al. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol 2004;173:307–13. [DOI] [PubMed] [Google Scholar]
- 33. Galluzzi L, Kepp O, Kroemer G. Immunogenic cell death in radiation therapy. Oncoimmunology 2013;2:e26536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Trembath, AP, Markiewicz MA. More than Decoration: Roles for Natural Killer Group 2 Member D Ligand Expression by Immune Cells. Front Immunol 2018;9:727–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Chakraborty M, Abrams SI, Camphausen K, et al. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 2003;170:6338–47. [DOI] [PubMed] [Google Scholar]
- 36. Xu J, Escamilla J, Mok S, et al. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res 2013;73:2782–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Demaria S, Golden EB, Formenti SC. Role of Local Radiation Therapy in Cancer Immunotherapy. JAMA Oncol 2015;1:1325–8. [DOI] [PubMed] [Google Scholar]
- 38. Matsumura S, Wang B, Kawashima N, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol 2008;181:3099–3107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Torihata H, Ishikawa F, Okada Y, et al. Irradiation up-regulates CD80 expression through two different mechanisms in spleen B cells, B lymphoma cells, and dendritic cells. Immunology 2004;112:219–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 2006;203:1259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014;124:687–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lugade AA, Sorensen EW, Gerber SA, et al. Radiation-induced IFN-gamma production within the tumor microenvironment influences antitumor immunity. J Immunol 2008;180:3132–9. [DOI] [PubMed] [Google Scholar]
- 43. Onishi H, Shirato H, Nagata Y, et al. Stereotactic body radiotherapy (SBRT) for operable stage I non-small-cell lung cancer: can SBRT be comparable to surgery? Int J Radiat Oncol Biol Phys 2011;81:1352–8. [DOI] [PubMed] [Google Scholar]
- 44.SBRT with immunotherapy in early stage non-small cell lung cancer: tolerability and lung effects. Available from: https://ClinicalTrials.gov/show/NCT03383302.
- 45.Clinical trials comparing immunotherapy plus stereotactic ablative radiotherapy (I-SABR) versus SABR alone for stage I, selected stage IIa or isolated lung parenchymal recurrent non-small cell lung cancer: I-SABR. Available from: https://ClinicalTrials.gov/show/NCT03110978.
- 46.Astra Zeneca (Immuno stereotactic ablative body radiotherapy) ISABR study: randomized phase I/II study of stereotactic body radiotherapy. Available from: https://ClinicalTrials.gov/show/NCT03148327.
- 47.Ablative stereotactic radiotherapy with durvalumab (MEDI4736). Available from: https://ClinicalTrials.gov/show/NCT03446547.
- 48.Atezolizumab and stereotactic body radiation therapy in treating patients with non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT02599454.
- 49.Stereotactic body radiation therapy (SBRT) combined with avelumab (anti-PD-L1) for management of early stage non-small cell lung cancer (NSCLC). Available from: https://ClinicalTrials.gov/show/NCT03050554.
- 50. Shaverdian N, Lisberg AE, Bornazyan K, et al. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol 2017;18:895–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Forde PM, Chaft JE, Smith KN, et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 2018;378:1976–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Pembrolizumab with and without radiotherapy for non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT03217071.
- 53.Neoadjuvant chemoradiation plus pembrolizumab followed by consolidation pembrolizumab in NSCLC. Available from: https://ClinicalTrials.gov/show/NCT02987998.
- 54.Neoadjuvant immunoradiation for resectable non-small cell lung Cancer. Available from: https://ClinicalTrials.gov/show/NCT03237377.
- 55.Adjuvant pembrolizumab in N2 positive non-small cell lung cancer patients. Available from: https://ClinicalTrials.gov/show/NCT03053856.
- 56. Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in Stage III NSCLC. N Engl J Med 2018;379:2342–50. [DOI] [PubMed] [Google Scholar]
- 57. Lin SH, Lin Y, Price J, et al. DETERRED: PD-L1 blockade to evaluate the safety of lung cancer therapy using carboplatin, paclitaxel, and radiation combined with MPDL3280A (atezolizumab). J Clin Oncol 2017;35:3064. [Google Scholar]
- 58. Durm GA, Althouse SK, Sadiq AA, et al. LUN 14-179: Phase II trial of concurrent chemoradiation followed by consolidation pembrolizumab in unresectable stage III NSCLC. J Clin Oncol 2018;36:8500. [Google Scholar]
- 59. Gerber DE, Urbanic JJ, Langer C, et al. Treatment design and rationale for a randomized trial of cisplatin and etoposide plus thoracic radiotherapy followed by nivolumab or placebo for locally advanced non–small-cell lung cancer (RTOG 3505). Clin Lung Cancer 2017;18:333–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Unresectable stage IIIA/IIIB Non-small Cell Lung Cancer (NSCLC). Available from: https://ClinicalTrials.gov/show/NCT03285321.
- 61. Solange P, Dirk DR, Urania D, et al. Safety evaluation of nivolumab added concurrently to radiotherapy in a standard first line chemo-RT regimen in unresectable locally advanced NSCLC: The ETOP NICOLAS phase II trial. J Clin Oncol 2018:36:8510. [Google Scholar]
- 62.Atezolizumab immunotherapy in patients with advanced NSCLC. Available from: https://ClinicalTrials.gov/show/NCT03102242.
- 63.Immunotherapy in combination with chemoradiation in patients with advanced solid tumors. Available from: https://ClinicalTrials.gov/show/NCT03509012.
- 64.Pembrolizumab, paclitaxel, carboplatin, and radiation therapy in treating patients with stage II-IIIB non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT02621398.
- 65. Tang C, Welsh JW, de Groot P, et al. Ipilimumab with stereotactic ablative radiation therapy: phase I results and immunologic correlates from peripheral T cells. Clin Cancer Res 2017;23:1388–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Study of combined ionizing radiation and ipilimumab in metastatic non-small cell lung cancer (NSCLC). Available from: https://ClinicalTrials.gov/show/NCT02221739.
- 67.Radiation therapy and MK-3475 for patients with recurrent/metastatic head and neck cancer, renal cell cancer, melanoma, and lung cancer. Available from: https://ClinicalTrials.gov/show/NCT02318771.
- 68.Pembrolizumab after SBRT versus pembrolizumab alone in advanced NSCLC. Available from: https://ClinicalTrials.gov/show/NCT02492568.
- 69.Study of PD1 blockade by pembrolizumab with stereotactic body radiotherapy in advanced solid tumors. Available from: https://ClinicalTrials.gov/show/NCT02608385.
- 70.SBRT and oncolytic virus therapy before pembrolizumab for metastatic TNBC and NSCLC. Available from: https://ClinicalTrials.gov/show/NCT03004183.
- 71.Sequencing of stereotactic ablative body radiotherapy in combination with PD-1 blockade using pembrolizumab in metastatic non-small cell lung carcinoma. Available from: https://ClinicalTrials.gov/show/NCT03307759.
- 72.Pembrolizumab in combination with radiotherapy in locally advanced non-small cell lung cancer (NSCLC). Available from: https://ClinicalTrials.gov/show/NCT03245177.
- 73.Pembrolizumab and SBRT in metastatic non-small-cell lung cancer patients. Available from: https://ClinicalTrials.gov/show/NCT03368222.
- 74.Pembrolizumab and stereotactic body radiotherapy in metastatic non-small-cell lung cancer patients. Available from: https://ClinicalTrials.gov/show/NCT03436056.
- 75. Tang C, de Groot P, Hess K, et al. Phase 1 study of pembrolizumab and stereotactic or hypofractionated radiation for metastatic non-small cell lung cancer. Int J Radiat Oncol Biol Phys 2017;99:S160–1. [Google Scholar]
- 76.Pembrolizumab and palliative radiotherapy in lung (PEAR). Available from: https://ClinicalTrials.gov/show/NCT02587455.
- 77.A randomized two arm phase II trial of pembrolizumab alone or sequentially following single fraction non-ablative radiation to one of the target lesions, in previously treated patients with stage IV NSCLC. Available from: https://ClinicalTrials.gov/show/NCT02658097.
- 78.Trial of radiation and gene therapy before nivolumab for metastatic non-small cell lung carcinoma and uveal melanoma. Available from: https://ClinicalTrials.gov/show/NCT02831933.
- 79.Concurrent or sequential immunotherapy and radiation therapy in patients with metastatic lung cancer. Available from: https://ClinicalTrials.gov/show/NCT03223155.
- 80.Checkpoint inhibition in combination with an immunoboost of external body radiotherapy in solid tumors. Available from: https://ClinicalTrials.gov/show/NCT03511391. [DOI] [PMC free article] [PubMed]
- 81.Fostering efficacy of anti-PD-1 treatment: nivolumab plus radiotherapy in advanced NSCLC. Available from: https://ClinicalTrials.gov/show/NCT03044626. [DOI] [PMC free article] [PubMed]
- 82.A pilot study of interlesional IL-2 and RT in patients with NSCLC. Available from: https://ClinicalTrials.gov/show/NCT03224871.
- 83.Radical-dose image guided radiation therapy in treating patients with metastatic non-small cell lung cancer undergoing immunotherapy. Available from: https://ClinicalTrials.gov/show/NCT03176173.
- 84.Hypofractionated radiation therapy to improve immunotherapy response in non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT03035890.
- 85.Priming immunotherapy in advanced disease with radiation. Available from: https://ClinicalTrials.gov/show/NCT03313804.
- 86.Trial of local consolidation therapy (LCT) after nivolumab and ipilimumab (LONESTAR). Available from: NCT03391869.
- 87.Radiation and immune checkpoints blockade in metastatic NSCLC (BMS # CA209-632). Available from: https://ClinicalTrials.gov/show/NCT03168464.
- 88.Stereotactic body radiotherapy (SBRT) plus immunotherapy for cancer (C4-MOSART). Available from: https://ClinicalTrials.gov/show/NCT03431948.
- 89.Phase I multicenter trial combining nivolumab, ipilimumab and hypo-fractionated radiotherapy for pretreated advanced stage non-small cell lung cancer patients. Available from: https://ClinicalTrials.gov/show/NCT03509584.
- 90.MPDL3280A and stereotactic ablative radiotherapy in patients with non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT02400814.
- 91.A pilot study of MPDL3280A and HIGRT in metastatic NSCLC. Available from: https://ClinicalTrials.gov/show/NCT02463994.
- 92.Image guided hypofractionated radiation therapy, nelfinavir mesylate, pembrolizumab, nivolumab and atezolizumab in treating patients with advanced melanoma, lung, or kidney cancer. Available from: https://ClinicalTrials.gov/show/NCT03050060.
- 93.Phase Ib study of stereotactic body radiotherapy (SBRT) in oligometastatic non-small lung cancer (NSCLC) with dual immune checkpoint inhibition. Available from: https://ClinicalTrials.gov/show/NCT03275597.
- 94.Durvalumab and tremelimumab with or without high or low-dose radiation therapy in treating patients with metastatic colorectal or non-small cell lung cancer. Available from: https://ClinicalTrials.gov/show/NCT02888743.
- 95.A trial of durvalumab and tremelimumab in combination with SBRT in patients with metastatic cancer. Available from: https://ClinicalTrials.gov/show/NCT03212469.
- 96.Trial of hypofractionated radiotherapy in combination with MEDI4736 and tremelimumab for patients with metastatic melanoma and lung, breast and pancreatic cancers. Available from: https://ClinicalTrials.gov/show/NCT02639026.
- 97. Withers HR. The four R’s of radiotherapy. Adv Radiat Biol 1975;5:241–71. [Google Scholar]
- 98. Xue J, Han C, Jackson A, et al. Doses of radiation to the pericardium, instead of heart, are significant for survival in patients with non-small cell lung cancer. Radiother Oncol 2018;1–7. 10.1016/j.radonc.2018.10.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 2009;15:5379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Aryankalayil MJ, Makinde AY, Gameiro SR, et al. Defining molecular signature of pro-immunogenic radiotherapy targets in human prostate cancer cells. Radiat Res 2014;182:139–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Lan J, Li R, Yin LM, et al. Targeting Myeloid-derived Suppressor Cells and Programmed Death Ligand 1 Confers Therapeutic Advantage of Ablative Hypofractionated Radiation Therapy Compared With Conventional Fractionated Radiation Therapy. Int J Radiat Oncol Biol Phys 2018;101:74–87. [DOI] [PubMed] [Google Scholar]
- 102. Nakamura N, Kusunoki Y, Akiyama M. Radiosensitivity of CD4 or CD8 positive human T-lymphocytes by an in vitro colony formation assay. Radiat Res 1990;123:224–7. [PubMed] [Google Scholar]
- 103. De Ruysscher D. Radiotherapy and PD-L1 inhibition in metastatic NSCLC. Lancet Oncol 2017;18:840–2. [DOI] [PubMed] [Google Scholar]
- 104. 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;24:5058–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Kwon ED, Drake CG, Scher HI, et al. 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;15:700–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Theelen W, Lalezari F, Heuvel M, et al. Randomized phase II study of pembrolizumab after SBRT versus pembrolizumab alone in patients with advanced NSCLC, preliminary results [PUB013]. J Thorac Oncol 2017;12:S1453. [Google Scholar]
- 107. Brooks ED, Chang JY. Time to abandon single-site irradiation for inducing abscopal effects. Nat Rev Clin Oncol 2019;16:123–35. [DOI] [PubMed] [Google Scholar]
- 108. Huang AC, Postow MA, Orlowski RJ, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res 2014;74: 5458–68. [DOI] [PubMed] [Google Scholar]
- 110. Young KH, Baird JR, Savage T, et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One 2016;11:e0157164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Samstein R, Rimner A, Barker CA, et al. Combined immune checkpoint blockade and radiation therapy: timing and dose fractionation associated with greatest survival duration among over 750 treated patients. Int J Radiat Oncol Biol Phys 2017;99:S129–30. [Google Scholar]
- 112. Wang Y, Deng W, Li N, et al. Combining immunotherapy and radiotherapy for cancer treatment: current challenges and future directions. Front Pharmacol 2018;9:5166–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Rosenberg SA. IL-2: The first effective immunotherapy for human cancer. J Immunol 2014;192:5451–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. 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;4:137ra174. [DOI] [PubMed] [Google Scholar]
- 115. Jiang T, Zhou C, Ren S. Role of IL-2 in cancer immunotherapy. Oncoimmunology 2016;5:e1163462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer 2016;4:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Vilalta M. The role of granulocyte macrophage colony stimulating factor (GM-CSF) in radiation-induced tumor cell migration. Clin Exp Metastasis 2018;35:247–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. 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;16:795–803. [DOI] [PubMed] [Google Scholar]
- 119.Trial of hypofractionated intensity modulated radiation therapy with temozolomide and granulocyte-macrophage colony-stimulating factor for patients with newly diagnosed glioblastoma multiforme. Available from: https://ClinicalTrials.gov/show/NCT02663440.
- 120.Sequential intranodal immunotherapy (SIIT) combined with anti-PD1 (Pembrolizumab) in follicular lymphoma. Available from: https://ClinicalTrials.gov/show/NCT02677155.
- 121.Study with CY, Pembrolizumab, GVAX, and SBRT in patients with locally advanced pancreatic cancer. Available from: https://ClinicalTrials.gov/show/NCT02648282.
- 122. Fujita T, Teh BS, Timme TL, et al. Sustained long-term immune responses after in situ gene therapy combined with radiotherapy and hormonal therapy in prostate cancer patients. Int J Radiat Oncol Biol Phys 2006;65:84–90. [DOI] [PubMed] [Google Scholar]
- 123. Aguilar LK, Shirley LA, Chung VM, et al. Gene-mediated cytotoxic immunotherapy as adjuvant to surgery or chemoradiation for pancreatic adenocarcinoma. Cancer Immunol Immunother 2015;64:727–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Chiocca EA, Aguilar LK, Bell SD, et al. Phase IB study of gene-mediated cytotoxic immunotherapy adjuvant to up-front surgery and intensive timing radiation for malignant glioma. J Clin Oncol 2011;29:3611–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Freeman-Keller M, Goldman J, Gray J. Vaccine immunotherapy in lung cancer: Clinical experience and future directions. Pharmacol Ther 2015;153:1–9. [DOI] [PubMed] [Google Scholar]
- 126. Ko EC, Formenti SC. Radiotherapy and checkpoint inhibitors: a winning new combination? Ther Adv Med Oncol 2018;10:1758835918768240. [DOI] [PMC free article] [PubMed] [Google Scholar]