1,976
Views
29
CrossRef citations to date
0
Altmetric
Original Research

Safety and efficacy of concurrent immune checkpoint inhibitors and hypofractionated body radiotherapy

, , , , , , , , , , , , , , , , , , & show all
Article: e1440168 | Received 01 Dec 2017, Accepted 07 Feb 2018, Published online: 15 Mar 2018

ABSTRACT

Integration of hypofractionated body radiotherapy (H-RT) into immune checkpoint inhibitor (ICI) therapy may be a promising strategy to improve the outcomes of ICIs, although sufficient data is lacking regarding the safety and efficacy of this regimen. We, hereby, reviewed the safety and efficacy of this combination in 59 patients treated with H-RT during or within 8 weeks of ICI infusion and compared results with historical reports of ICI treatment alone. Most patients had RCC or melanoma. Median follow-up was 11 months. Most patients received either Nivolumab alone or with Ipilimumab; 83% received stereotactic RT and 17% received conformal H-RT. Any grade adverse events (AEs) were reported in 46 patients, and grade 3–4 in 12 patients without any treatment-related grade 5 toxicity. The most common grade 3 AEs were fatigue and pneumonitis. Grade 3–4 toxicities were higher with ICI combination and with simultaneous ICIs. Overall, most any-grade or grade ≥3 AE rates did not differ significantly from historically reported rates with single-agent or multi-agent ICIs. Toxicity did not correlate with H-RT site, dose, fraction number, tumor type, or ICI and H-RT sequencing. Median progression-free survival was 6.5 months. Objective response rate (ORR) was 26%; 10% had complete response (CR). Median duration of response was 9.4 ± 4.6 months. H-RT of lung lesions was more likely to achieve CR than other sites. H-RT of bone lesions had a lower ORR than non-bone H-RT. In conclusion, combining body H-RT with ICIs is safe and promising. Prospective validation is warranted.

Introduction

Immune checkpoint inhibitors (ICIs) have improved outcomes for several metastatic cancers, including melanoma,Citation1-Citation7 renal cell carcinoma (RCC),Citation8-Citation11 non-small cell lung cancer (NSCLC),Citation12-Citation18 and bladder cancer,Citation19-Citation21 leading to their FDA approval. Despite promising outcomes in phase III clinical trials, complete responses are infrequent, and most patients who respond eventually progress. Thus, outcomes can be improved in these patients. Strategies under investigation to improve ICI response rates include simultaneous ICI therapy and combination with radiotherapy (RT).

Preclinical studies have shown that the combination of RT with ICIs is more effective than either treatment modality alone.Citation22-Citation27 Despite the hype, such treatment combinations are still far from standard clinical practice, because the optimal timing, dosing, and fractionation of RT to yield the best synergistic outcomes remain uncertain. Moreover, data on the safety of such combinations are scarce. ICIs are associated with serious and potentially fatal adverse events (AEs), with grade ≥3 AEs ranging between 5–15% in monotherapies and about 50% in simultaneous combinations.Citation1-Citation10,Citation12-Citation21,Citation28 The few studies that have evaluated the combination of ICIs with RT have shown that these combinations are probably safe for conventional RT and brain stereotactic radiosurgery (SRS).Citation29-Citation36

Evidence shows that hypofractionated RT (H-RT) is more immunogenic than conventional regimensCitation37-Citation39 and may synergize better with ICIs. However, only a few patients in the reported combination studies received stereotactic ablative body RT (SAbR). Multiple clinical trials are currently investigating the combination of RT and ICIs, with many combining body H-RT or stereotactic ablative RT (SAbR) with limited supporting data.Citation23 Our study analyzes toxicity and efficacy data for patients who received extra-cranial H-RT or SAbR concurrently or within 8 weeks of ICI infusion and compares the results with historically reported data of ICI alone treated patients.

Results

Patient characteristics

We identified 59 patients who received at least one ICI infusion and at least one H-RT course (SAbR or conformal RT; C-RT) concurrently or within 8 weeks of each other between 2012 and 2017. summarizes patient characteristics, including diagnoses, prior treatments, and characteristics of ICI and H-RT therapies. Most patients were male (64%), and most had RCC or melanoma (76%). Seventy-six percent of patients had widely metastatic disease, and 24% had locally advanced or oligo-metastatic disease. Median age was 64 (range 14–84) years at diagnosis of metastasis. Fifty patients (85%) had received prior systemic therapy. The remaining 9 patients (8 with melanoma, 1 with RCC) received ICIs in the first-line setting. Four patients received only a single infusion; their treatments were stopped due to toxicity (2), death (1), or patient preference for hospice enrollment (1). Most patients received Nivolumab alone (46%) or a combination of ICIs. Combination ICIs were sequential, simultaneous, or both. 14 patients (24%) received a simultaneous combination of ICIs, all of which consisted of Nivolumab and Ipilimumab.

Table 1. Patient characteristics and treatments.

All patients received at least one SAbR (83%) or hypofractionated C-RT (17%) treatment, either concurrently or within 8 weeks of the ICI (). Thirty-five patients (59%) received multiple RT treatments within the study's 8-week time constraint, making a total of 137 courses of radiation (56% SAbR) with a median of 2 RT courses per patient (range 1–8).

Toxicity

Forty-six patients (78%) experienced any treatment-related AE, mostly grade 1–2 (). The most common AEs were fatigue (37%), nausea (19%), transaminitis (17%), and rash/dermatitis (17%). Grade 3–4 treatment-related AEs occurred in 12 patients (20%). Only one patient, who did not receive liver RT, had a grade 4 AE (asymptomatic elevation of liver enzymes), but eventually recovered after the ICI was withheld. The most common grade 3 AEs were fatigue and pneumonitis (3 patients, 5% each). All patients with grade 3 pneumonitis had RCC, and only one received lung RT during or within 8 weeks of ICI. Four patients (7%) developed more than one grade ≥3 AE; two of whom received only Nivolumab, one had sequential Pembrolizumab and Ipilimumab, and one had simultaneous Nivolumab and Ipilimumab followed by Nivolumab only. No treatment-related grade 5 toxicities were observed. Median (average) times from treatment initiation to grade 1–2 or grade ≥3 toxicity were 32 days (65 days) or 98 days (107 days), respectively. Immune-related AEs were treated by cessation or holding of ICIs and usually steroids.

Table 2. Treatment-related adverse events.

Forty-five patients (76%) in our cohort received single or sequential ICI, and 14 (24%) received simultaneous ICIs. We compared treatment-related grade ≥3 AEs in each subgroup to historical controls of cutaneous melanoma patients receiving Nivolumab only,Citation40 RCC patients receiving Nivolumab only (CheckMate 025),Citation10 NSCLC patients receiving Nivolumab only (CheckMate 057),Citation17 cutaneous melanoma patients receiving Nivolumab + Ipilimumab,Citation40 and recurrent small cell lung cancer (SCLC) patients receiving Nivolumab + Ipilimumab in different doses (CheckMate 032)Citation13 (). Overall, there was no difference in most grade ≥3 toxicities compared to the historical controls. For patients receiving single-agent ICI, we observed a statistical increase in the risk of grade ≥3 fatigue (7% vs. 0.6%; p = 0.007) and grade ≥3 pneumonitis (4% vs. 0.2%; p = 0.01) between our study and the pooled analysis of cutaneous melanoma, and a statistical increase in the risk of grade ≥3 fatigue (7% vs. 1%; p = 0.03) between our study and CheckMate 057. For patients receiving simultaneous Nivolumab and Ipilimumab, we observed a statistical increase in the risk of grade ≥3 transaminitis (14% vs. 0%; p = 0.03) between our study and CheckMate 032 (Nivo 1 mg/kg and Ipi 3 mg/kg arm).

Table 3. Grade 3–4 toxicities in our cohort compared to cohorts from landmark studies.

We next analyzed correlations between different treatment parameters and any grade or grade ≥3 AEs (). We found more grade ≥3 AEs in patients who received multiple ICIs (whether sequential or simultaneous) than in those who only received a single ICI (36.8% vs 12.5%, p = 0.04) and more grade ≥3 AEs in patients who received simultaneous ICI combinations than in those who received single or multiple but sequential ICIs (42.9% vs. 13.3%, p = 0.03). Otherwise, there was no correlation between toxicity and diagnoses, treatment sequence, RT site, RT dose (BED, biologically equivalent dose), RT fraction number, receiving other concurrent RT (conventional fractionation or brain SRS) or number and duration of ICI infusions.

Table 4. Relationship of treatment parameters with occurrence of adverse events.

We grouped patients according to radiation site for every treatment-related AE (). Despite the small numbers, there was no clear correlation between particular toxicities and RT to or near the corresponding sites. For example, 5.4% of patients who received RT to the chest, including the lungs, chest wall, mediastinum, and T-spine, developed grade 3 pneumonitis, compared to 4.5% in those who did not receive RT to the chest (p = 1.00).

Table 5. Relationship of radiotherapy treatment site to occurrence of adverse events.

Evaluating patient outcomes

Median follow-up was 11.1 months (IQR: 6.5-15.8; mean 11.8±8.1). The most common reason for discontinuing ICI was disease progression (24 patients, 41%). Treatment-related AEs prompted treatment discontinuation in 6 patients (10%). At last follow-up, 22 patients (37%) continued to receive ICIs, 31 patients (53%) were alive, 22 patients (37%) had died, and 6 (10%) had been lost to follow-up. All deceased patients died of disease progression and/or its complications.

Among evaluable patients (n = 39), median follow-up was 13.5 months (IQR: 8.9-16.6; mean 13.5±6.6). Overall response rate (ORR) was 26% (10/39), including 10% (4/39) with complete response (CR); 6 patients had partial response (PR), 10 had stable disease (SD), and 19 had progressive disease (PD). All patients with CR (3 with melanoma and 1 with NSCLC) received SAbR to a lung lesion concurrently with Nivolumab only and are still alive receiving it at last follow-up. One-year overall survival (OS) was 69.8% (95% CI 55.8-80.1) (supplementary Table 1). Median OS was not reached. Median progression-free survival (PFS) was 6.5 months (3.4-12.3) for all evaluable patients (17.1 months for CR patients and 12.6 months for CR+PR+SD patients). Median duration of response was 9.4±4.6 months for patients with PR and CR; 5 of whom were ongoing responses at last follow up. All patients with CR had oligometastatic disease involving ≤5 lesions in 1–3 organ systems, who received SAbR to the lung concurrently with Nivolumab only and are still alive receiving it at last follow-up.

We analyzed the relationship between different treatment parameters and tumor responses ( and supplementary Table 2). Patients who received H-RT to bones tended to have less objective responses (CR+PR) (: 10.5% vs. 40%, p = 0.065). Otherwise, there was no correlation between tumor response and diagnoses, number of ICIs, treatment sequence, RT site, RT dose (BED), RT fraction number, receiving other concurrent RT (conventional fractionation or brain SRS) or number and duration of ICI infusions. Comparing CR with non-CR responses (PR+SD+PD), we found that RT of lung lesions was significantly more likely to induce CR than RT of any other site (: 26.7% vs. 0%; p = 0.0166).

Table 6. Relationship of treatment parameters with treatment response.

Discussion

This study has limitations associated with a retrospective study with a relatively small and heterogeneous sample of patients, which can be subject to bias. While the short median follow-up of 11.1 months is adequate to cover short-term toxicity, ORR, and PFS, longer follow-up is needed to cover long-term outcomes and late toxicities. We also lack a proper comparison group for comparing outcomes and toxicities. However, the focus of this study, looking exclusively at the toxicities and outcomes of concurrent (within 8 weeks) ICIs with extra-cranial H-RT, makes the results applicable and relevant to ongoing clinical trials.

ICIs are overall well tolerated, with grade ≥3 toxicities ranging from 5–15% in single ICIs and >50% in simultaneous ICIs.Citation41,Citation42 Our study's toxicity profile, with 78% any grade toxicity and 20% grade ≥3 toxicity, fits well within the reported values, given that 24% of our patients received simultaneous ICIs. H-RT, thus, did not increase treatment- or immune-related AEs of ICI therapy, indicating that H-RT does not augment immunity against self-antigens. This was independent of irradiation site, suggesting that this is an immune-related rather than an RT-induced toxicity. When compared to different historical controls from multiple landmark studies using single or simultaneous ICIs, we did not notice any significant increase in grade ≥3 toxicities, despite the acknowledged controversy of such a statistical comparison. Interestingly, we noticed a minimal but statistically significant increase in the rate of grade ≥3 pneumonitis, fatigue, and transaminitis in some of these comparisons. Admittedly, the historical control cohorts we chose are not ideal, but they have a large number of patients treated for different metastatic disease using single or simultaneous ICIs. While the population is small and heterogeneous and the comparison group is not ideal, vigilant monitoring of these patients is warranted.

We report an ORR of 26% (28% for single-agent ICIs), a median PFS of 6.5 months for all evaluable patients, and a duration of response of 9.4±4.6 months for patients who responded. It is difficult, however, to formulate clear conclusions given the heterogeneity in diagnoses and treatments. Interestingly, bone RT was the only site that trended towards a lower ORR compared to non-bone RT (). This may suggest that bones are the least immunogenic site, which agrees with the clinical understanding that bone lesions are often lytic and may contain fewer tumor cells available for antigen presentation when irradiated than soft tissue metastases. On the other hand, all patients with CR had a relatively small burden of disease and received SAbR to a lung lesion. This notion of organ-specific differences in immune-modulatory properties is well known in the field of transplant immunology, where kidney and liver transplants require far less immune-suppression than lungs and hearts to prevent rejection.Citation43 Therefore, it is feasible that the immune-modulatory response from RT is also organ-dependent, as our analysis suggests. Notably, three of the CRs had primary melanoma, yielding a 30% CR rate (3 of 10 evaluable patients) for melanoma patients (0-22% in prior reportsCitation3,Citation10,Citation35).

Only a few studies have evaluated combinations of ICIs and RT.Citation29-Citation31,Citation33,Citation35,Citation36,Citation44 While these combinations were deemed safe, these studies focused on brain SRS or conventional radiotherapy, and the number of patients who received H-RT or SAbR was minimal. Moreover, many patients received RT several months prior to ICIs, making them less relevant to the ongoing trials. For example, in a study of patients who had previous radiotherapy and received Pembrolizumab for NSCLC,Citation36 very few patients received H-RT, and the median time between RT and the first infusion of Pembrolizumab was 9.5 months. In a recently published phase I trial of patients (with multiple primary histologies) receiving SAbR to liver or lung lesions concurrently with Ipilimumab, grade 3 toxicities were observed in 34% of patients and no CRs were observed. The authors concluded the combination was safe and that immune activation, measured using assays of CD8+ and CD4+ Tcells, was greater after liver irradiation. This is contrasted to our study where liver RT did not increase response rate (Supplementary Table 2).Citation45 In another prospective study of patients with metastatic melanoma receiving Ipilimumab concurrently with RT (8/22 received SAbR), authors concluded that the combination was safe and promising. Interestingly, of the 3 CR observed, 2 had SAbR to lung lesions.Citation46 Overall, similar to these reports, our study adds to the evidence that combining RT with immunotherapy is probably safe and, at least, promising in terms of response.

While the exact mechanisms of the combination are not well understood, several studies indicate the cumulative benefits of combining RT with immune-related treatments to create an enhanced immunologic response.Citation24,Citation27,Citation47 The optimal fractionation required for tumor killing and immune response, however, remains debatable, though most studies suggest the benefit of hypofractionation and larger doses per fraction.Citation48-Citation52 We found no difference in toxicity or outcome between different numbers of H-RT fractions or biologically equivalent doses (BED). While this might be attributable to the heterogeneous patient population, dose regimens, and relatively small population size in this study, the lack of any trend towards significance suggests that there may be no universal optimal immune-modulatory radiation dose. Prospective and cancer site-specific clinical trials are required to adequately answer this question.

We have shown that combining immune checkpoint inhibitors with extra-cranial H-RT, such as SAbR, is safe and well tolerated with no significant increase in toxicity, no temporal or spatial relationships between toxicities and radiation timing or sites, and a possible association between lung SAbR and complete responses.

Patients and methods

Patient selection

With institutional review board approval (IRB # STU 042017-028), we retrospectively reviewed medical records of patients with pathologically-confirmed metastatic RCC, melanoma, NSCLC, bladder, pancreatic, and head and neck cancers. All patients were treated using ICIs combined with hypofractionated SAbR or conformal radiation therapy (C-RT) delivered to extra-cranial site(s) during or within 8 weeks of the start or end of ICI infusions at the University of Texas Southwestern Medical Center. Patients who only received brain SRS, conventional, superficial, or electron RT were excluded. The 8-week cut-off was selected to enrich our population with patients having a close temporal association between ICIs and RT. A multidisciplinary team of medical oncologists, radiation oncologists, and surgeons treated each patient.

Radiotherapy and immune checkpoint inhibitors

H-RT is defined as regimens of 1–5 fractions with ≥5 Gy per fraction and includes SAbR and C-RT. C-RT includes intensity modulated radiotherapy (IMRT) and 3-dimensional conformal radiation therapy (3D-CRT). Patients were included even if they received other concurrent RT (stereotactic brain RT or conventionally fractionated RT) as long as they received at least one course of SAbR or hypofractionated C-RT. Computed tomography (CT) simulation was performed per institutional guidelines. All SAbR treatments used a stereotactic body frame. The treating physician chose motion management and immobilization setups as needed. Image guidance was used where applicable. Planning target volume (PTV) expansion differed depending on RT technique. All RT plans were optimized to ensure adequate PTV coverage by the prescription dose. BED was calculated using the universal survival model.Citation53 Our institutional normal tissue constraints are published elsewhere.Citation54 Inhibitors of PD-1 (Nivolumab and Pembrolizumab), PD-L1 (Atezolizumab), and CTLA-4 (Ipilimumab) were administered according to FDA-approved guidelines.

Evaluating treatment toxicity and outcomes

We analyzed AEs based on CTCAE 4.0.Citation55 We defined time to toxicity from treatment initiation (ICI or H-RT, whichever came first) until a particular toxicity developed. We analyzed toxicities against H-RT site, dose, tumor type, or ICI and H-RT sequencing. We defined best overall response as best tumor response (complete response [CR], partial response [PR], stable disease [SD], and progressive disease [PD]) from ICI therapy initiation until disease progression or last follow-up, as evaluated by a board-certified radiologist, based on the immune-related (ir)-RECIST criteria.Citation56,Citation57 Radiated lesions were excluded from response evaluation. Objective response rate (ORR) included patients with CR or PR. We excluded from our outcome analysis patients who did not receive appropriate follow-up imaging (i.e. the two sets of imaging required for ir-RECIST) or did not have at least 1 non-radiated measurable lesions for response evaluation. We considered these patients non-evaluable for outcome analysis but evaluable for toxicity. We calculated overall survival (OS) from treatment initiation until death or last known date of survival. Patients lost to follow-up were censored at last follow-up. We calculated progression-free survival (PFS) from ICI therapy initiation until death or first dated progression, as defined by ir-RECIST criteria.

Statistical considerations

We used the Kaplan-Meier method to estimate OS and PFS. Fisher's exact test was used to compare data to historical controls. Correlation between treatment or patient variables and toxicity (or response to treatment) was assessed using Fisher's exact test for categorical parameters and Kruskal-Wallis test for continuous measures. All statistical tests were two-sided, and a p-value of ≤0.05 was considered statistically significant for all comparisons. We used SAS 9.4 (SAS Institute Inc., Cary, NC) for analysis.

Conflicts of interest

All authors report no conflicts of interest except 1) Dr. James Brugarolas who receives research funding from Genentech, is an advisory board member with Bethyl Laboratories and is a consultant with Nektar and 2) Dr. Hans Hammers who is an advisory board member with BMS and receives trial funding from BMS.

Supplemental material

2017ONCOIMM0989R-file005.docx

Download MS Word (18 KB)

Acknowledgment

The authors thank Dr. Jonathan S. Feinberg for scientific editing of the manuscript.

Funding

AC, KC and JB are supported by 1P50CA196516.

References

  • Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. doi:10.1056/NEJMoa1504030. PMID:26027431.
  • Hodi FS, Chesney J, Pavlick AC, Robert C, Grossmann KF, McDermott DF, Linette GP, Meyer N, Giguere JK, Agarwala SS, et al. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2016;17(11):1558–68. doi:10.1016/S1470-2045(16)30366-7. PMID:27622997.
  • Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N, Giguere JK, Agarwala SS, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17. doi:10.1056/NEJMoa1414428. PMID:25891304.
  • Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–30. doi:10.1056/NEJMoa1412082. PMID:25399552.
  • Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH Jr, Lao CD, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–84. doi:10.1016/S1470-2045(15)70076-8. PMID:25795410.
  • Weber JS, Hodi FS, Wolchok JD, Topalian SL, Schadendorf D, Larkin J, Sznol M, Long GV, Li H, Waxman IM, Jiang J, et al. Safety profile of nivolumab monotherapy: A pooled analysis of patients with advanced melanoma. J Clin Oncol. 2017;35(7):785–92. doi:10.1200/JCO.2015.66.1389. PMID:28068177.
  • Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL Jr, Lawrence DP, Logan TF, Schuchter LM, Nair S, Fecher L, et al. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol. 2016;17(7):943–55. doi:10.1016/S1470-2045(16)30126-7. PMID:27269740.
  • Escudier B, Sharma P, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al. CheckMate 025 randomized phase 3 study: Outcomes by key baseline factors and prior therapy for nivolumab versus everolimus in advanced Renal Cell Carcinoma. Eur Urol. 2017;72(6):962–71. doi:10.1016/j.eururo.2017.02.010.
  • Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, Vaishampayan UN, Drabkin HA, George S, Logan TF, et al. Nivolumab for metastatic renal cell carcinoma: Results of a randomized phase II trial. J Clin Oncol. 2015;33(13):1430–7. doi:10.1200/JCO.2014.59.0703. PMID:25452452.
  • Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER,et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13. doi:10.1056/NEJMoa1510665. PMID:26406148.
  • Hammers HJ, Plimack ER, Infante JR, Rini BI, McDermott DF, Lewis LD, Voss MH, Sharma P, Pal SK, Razak ARA, et al. Safety and efficacy of nivolumab in combination with ipilimumab in metastatic Renal Cell Carcinoma: The CheckMate 016 Study. J Clin Oncol. 2017;35(34):3851–8. JCO2016721985. doi:10.1200/JCO.2016.72.1985. PMID:28678668
  • Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, Horn L, Lena H, Minenza E, Mennecier B, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16(3):257–65. doi:10.1016/S1470-2045(15)70054-9. PMID:25704439.
  • Antonia SJ, Lopez-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17(7):883–95. doi:10.1016/S1470-2045(16)30098-5. PMID:27269741.
  • Rizvi NA, Hellmann MD, Brahmer JR, Juergens RA, Borghaei H, Gettinger S, Chow LQ, Gerber DE, Laurie SA, Goldman JW, et al. Nivolumab in combination with platinum-based doublet chemotherapy for first-line treatment of advanced Non-Small-Cell Lung Cancer. J Clin Oncol. 2016;34(25):2969–79. doi:10.1200/JCO.2016.66.9861. PMID:27354481.
  • Borghaei H, Brahmer J. Nivolumab in nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med. 2016;374(5):493–4. PMID:26840144.
  • Gettinger S, Rizvi NA, Chow LQ, Borghaei H, Brahmer J, Ready N, Gerber DE, Shepherd FA, Antonia S, Goldman JW, et al. Nivolumab monotherapy for first-line treatment of advanced Non-Small-Cell Lung Cancer. J Clin Oncol. 2016;34(25):2980–7. doi:10.1200/JCO.2016.66.9929. PMID:27354485.
  • Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al. Nivolumab versus docetaxel in advanced nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med. 2015;373(17):1627–39. doi:10.1056/NEJMoa1507643. PMID:26412456.
  • Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al. Nivolumab versus docetaxel in advanced squamous-cell Non-Small-Cell Lung Cancer. N Engl J Med. 2015;373(2):123–35. doi:10.1056/NEJMoa1504627. PMID:26028407.
  • Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, Hoffman-Censits J, Perez-Gracia JL, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67–76. doi:10.1016/S0140-6736(16)32455-2. PMID:27939400.
  • Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O'Donnell PH, Balmanoukian A, Loriot Y, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909–20. doi:10.1016/S0140-6736(16)00561-4. PMID:26952546.
  • Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, Plimack ER, Vaena D, Grimm MO, Bracarda S, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18(3):312–22. doi:10.1016/S1470-2045(17)30065-7. PMID:28131785.
  • Ishihara D, Pop L, Takeshima T, Iyengar P, Hannan R. Rationale and evidence to combine radiation therapy and immunotherapy for cancer treatment. Cancer Immunology, Immunotherapy: CII. 2017;66(3):281–98. doi:10.1007/s00262-016-1914-6. PMID:27743027.
  • Crittenden M, Kohrt H, Levy R, Jones J, Camphausen K, Dicker A, Demaria S, Formenti S. Current clinical trials testing combinations of immunotherapy and radiation. Semin Radiat Oncol. 2015;25(1):54–64. doi:10.1016/j.semradonc.2014.07.003. PMID:25481267.
  • Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, Stratford IJ, Poon E, Morrow M, Stewart R, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74(19):5458–68. doi:10.1158/0008-5472.CAN-14-1258. PMID:25274032.
  • Park SS, Dong H, Liu X, Harrington SM, Krco CJ, Grams MP, Mansfield AS, Furutani KM, Olivier KR, Kwon ED. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol Res. 2015;3(6):610–9. doi:10.1158/2326-6066.CIR-14-0138. PMID:25701325.
  • Twyman-Saint\sVictor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–7. doi:10.1038/nature14292. PMID:25754329.
  • Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687–95. doi:10.1172/JCI67313. PMID:24382348.
  • George S, Motzer RJ, Hammers HJ, Redman BG, Kuzel TM, Tykodi SS, Plimack ER, Jiang J, Waxman IM, Rini BI. Safety and efficacy of nivolumab in patients with metastatic renal cell carcinoma treated beyond progression: A subgroup analysis of a randomized clinical trial. JAMA Oncol. 2016;2(9):1179–86. doi:10.1001/jamaoncol.2016.0775. PMID:27243803.
  • Bang A, Wilhite TJ, Pike LRG, Cagney DN, Aizer AA, Taylor A, Spektor A, Krishnan M, Ott PA, Balboni TA,et al. Multicenter evaluation of the tolerability of combined treatment with PD-1 and CTLA-4 immune checkpoint inhibitors and palliative radiation therapy. Int J Radiat Oncol Biol Phys. 2017;98(2):344–51. doi:10.1016/j.ijrobp.2017.02.003. PMID:28463153.
  • Ahmed KA, Stallworth DG, Kim Y, Johnstone PA, Harrison LB, Caudell JJ, Yu HH, Etame AB, Weber JS, Gibney GT. Clinical outcomes of melanoma brain metastases treated with stereotactic radiation and anti-PD-1 therapy. Ann Oncol. 2016;27(3):434–41. doi:10.1093/annonc/mdv622. PMID:26712903.
  • Williams NL, Wuthrick EJ, Kim H, Palmer JD, Garg S, Eldredge-Hindy H, Daskalakis C, Feeney KJ, Mastrangelo MJ, Kim LJ, et al. Phase 1 study of ipilimumab combined with whole brain radiation therapy or radiosurgery for melanoma patients with brain metastases. IJROBP. 2017;99(1):22–30.
  • Gao J, He Q, Subudhi S, Aparicio A, Zurita-Saavedra A, Lee DH, Jimenez C, Suarez-Almazor M, Sharma P. Review of immune-related adverse events in prostate cancer patients treated with ipilimumab: MD Anderson experience. Oncogene. 2015;34(43):5411–7. doi:10.1038/onc.2015.5. PMID:25659583.
  • Qin R, Olson A, Singh B, Thomas S, Wolf S, Bhavsar NA, Hanks BA, Salama JK, Salama AK. 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:10.1016/j.ijrobp.2016.04.017. PMID:27375168.
  • Boyer MJ, Gu L, Wang X, Kelsey CR, Yoo DS, Onaitis MW, Dunphy FR, Crawford J, Ready NE, Salama JK. Toxicity of definitive and post-operative radiation following ipilimumab in non-small cell lung cancer. Lung Cancer. 2016;98:76–78. doi:10.1016/j.lungcan.2016.05.014. PMID:27393510.
  • Koller KM, Mackley HB, Liu J, Wagner H, Talamo G, Schell TD, Pameijer C, Neves RI, Anderson B, Kokolus KM, et al. Improved survival and complete response rates in patients with advanced melanoma treated with concurrent ipilimumab and radiotherapy versus ipilimumab alone. Cancer Biol Ther. 2017;18(1):36–42. doi:10.1080/15384047.2016.1264543. PMID:27905824.
  • Shaverdian N, Lisberg AE, Bornazyan K, Veruttipong D, Goldman JW, Formenti SC, Garon EB, Lee P. 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(7):895–903. doi:10.1016/S1470-2045(17)30380-7. PMID:28551359.
  • Camphausen K, Moses MA, Menard C, Sproull M, Beecken WD, Folkman J, O'Reilly MS. Radiation abscopal antitumor effect is mediated through p53. Cancer Res. 2003;63(8):1990–3. PMID:12702593.
  • Filatenkov A, Baker J, Mueller AM, Kenkel J, Ahn GO, Dutt S, Zhang N, Kohrt H, Jensen K, Dejbakhsh-Jones S, et al. Ablative tumor radiation can change the tumor immune cell microenvironment to induce durable complete remissions. Clin Cancer Res. 2015;21(16):3727–39. doi:10.1158/1078-0432.CCR-14-2824. PMID:25869387.
  • Wattenberg MM, Fahim A, Ahmed MM, Hodge JW. Unlocking the combination: potentiation of radiation-induced antitumor responses with immunotherapy. Radiat Res. 2014;182(2):126–38. doi:10.1667/RR13374.1. PMID:24960415.
  • D'Angelo SP, Larkin J, Sosman JA, Lebbé C, Brady B, Neyns B, Schmidt H, Hassel JC, Hodi FS, Lorigan P, et al. Efficacy and safety of nivolumab alone or in combination with ipilimumab in patients with mucosal melanoma: A pooled analysis. J Clin Oncol. 2017;35(2):226–35. doi:10.1200/JCO.2016.67.9258. PMID:28056206.
  • Chen TW, Razak AR, Bedard PL, Siu LL, Hansen AR. A systematic review of immune-related adverse event reporting in clinical trials of immune checkpoint inhibitors. Ann Oncol. 2015;26(9):1824–9. doi:10.1093/annonc/mdv182. PMID:25888611.
  • Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors: A review. JAMA Oncol. 2016;2(10):1346–53. doi:10.1001/jamaoncol.2016.1051. PMID:27367787.
  • Madariaga ML, Kreisel D, Madsen JC. Organ-specific differences in achieving tolerance. Curr Opin Organ Transplant. 2015;20(4):392–9. doi:10.1097/MOT.0000000000000206. PMID:26147678.
  • Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, Krainer M, Houede N, Santos R, Mahammedi H, 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(7):700–12. doi:10.1016/S1470-2045(14)70189-5. PMID:24831977.
  • Tang C, Welsh JW, de Groot P, Massarelli E, Chang JY, Hess KR, Basu S, Curran MA, Cabanillas ME, Subbiah V, et al. Ipilimumab with stereotactic ablative radiation therapy: Phase I results and immunologic correlates from peripheral T cells. Clin Cancer Res. 2017;23(6):1388–96. doi:10.1158/1078-0432.CCR-16-1432. PMID:27649551.
  • Hiniker SM, Reddy SA, Maecker HT, Subrahmanyam PB, Rosenberg-Hasson Y, Swetter SM, Saha S, Shura L, Knox SJ. A prospective clinical trial combining radiation therapy with systemic immunotherapy in metastatic melanoma. Int J Radiat Oncol Biol Phys. 2016;96(3):578–88. doi:10.1016/j.ijrobp.2016.07.005. PMID:27681753.
  • Verbrugge I, Hagekyriakou J, Sharp LL, Galli M, West A, McLaughlin NM, Duret H, Yagita H, Johnstone RW, Smyth MJ, et al. Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies. Cancer research. 2012;72(13):3163–74. doi:10.1158/0008-5472.CAN-12-0210. PMID:22570253.
  • Mason KA, Ariga H, Neal R, Valdecanas D, Hunter N, Krieg AM, Whisnant JK, Milas L. Targeting toll-like receptor 9 with CpG oligodeoxynucleotides enhances tumor response to fractionated radiotherapy. Clin Cancer Res: J Am Assoc Cancer Res. 2005;11(1):361–9.
  • Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, Demaria S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res: J Am Assoc Cancer Res. 2009;15(17):5379–88. doi:10.1158/1078-0432.CCR-09-0265..
  • Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, Beckett M, Sharma R, Chin R, Tu T,et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114(3):589–95. doi:10.1182/blood-2009-02-206870. PMID:19349616.
  • Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. 2012;83(4):1306–10. doi:10.1016/j.ijrobp.2011.09.049. PMID:22208977.
  • Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, Inghirami G, Coleman CN, Formenti SC, Demaria S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. doi:10.1038/ncomms15618. PMID:28598415.
  • Park C, Papiez L, Zhang S, Story M, Timmerman RD. Universal survival curve and single fraction equivalent dose: useful tools in understanding potency of ablative radiotherapy. Int J Radiat Oncol Biol Phys. 2008;70(3):847–52. doi:10.1016/j.ijrobp.2007.10.059. PMID:18262098.
  • Kim DWN, Medin PM, Timmerman RD. Emphasis on repair, not just avoidance of injury, facilitates prudent stereotactic ablative radiotherapy. Semin Radiat Oncol. 2017;27(4):378–92. doi:10.1016/j.semradonc.2017.04.007. PMID:28865521.
  • NIH. Common Terminology Criteria for Adverse Events (CTCAE), Version 4.0. 2009; https://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_8.5x11.pdf. Accessed August 2017.
  • Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47. doi:10.1016/j.ejca.2008.10.026. PMID:19097774.
  • Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, Maio M, Binder M, Bohnsack O, Nichol G, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–20. doi:10.1158/1078-0432.CCR-09-1624. PMID:19934295.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.