Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2021 Oct;11(10):a037713. doi: 10.1101/cshperspect.a037713

Radiation Therapy in Non-Small-Cell Lung Cancer

Michael Dohopolski 1,3, Sujana Gottumukkala 1,3, Daniel Gomez 2, Puneeth Iyengar 1
PMCID: PMC8485739  PMID: 34127511

Abstract

The management of non-small-cell lung cancer (NSCLC) varies according to stage. Surgical resection is reserved for operable patients with early-stage NSCLC, while high-dose target radiation—stereotactic body radiation therapy (SBRT)—is reserved for patients whose comorbidities prohibit them from a major surgical procedure. The treatment of locally advanced NSCLC (LA-NSCLC) is stratified according to resectability. Those with resectable disease may require additional treatments such as chemotherapy and radiation, while patients with unresectable disease will require definitive chemoradiation therapy with adjuvant durvalumab. Patients with limited metastatic disease benefit from the combination of SBRT and systemic therapy.


The treatment and prognosis of patients with non-small-cell lung cancer (NSCLC) vary substantially depending on the burden of disease; therefore, it is of utmost importance that patients undergo proper staging procedures. The typical treatment paradigm for patients with early-stage NSCLC (ES-NSCLC) is resection for those willing and able to undergo surgery, while stereotactic body radiation therapy (SBRT) is reserved for inoperable patients. Patients with resectable locally advanced NSCLC (LA-NSCLC) will require adjuvant therapies following resection, potentially including postoperative radiation. Patients with unresectable LA-NSCLC now receive definitive chemoradiation followed by adjuvant durvalumab as standard of care. Unfortunately, despite aggressive treatment, many of these patients, especially those with LA-NSCLC, may develop metastatic disease. Historically, the standard of care focused on chemotherapy, with radiation used mostly for palliative intent; however, there are those with limited metastatic disease burden who may derive long-term benefit from aggressive multimodal therapy including SBRT and immunotherapy. In this article, we discuss the management of patients with NSCLC, the data supporting these recommendations, and the new and upcoming studies seeking to advance the field.

EARLY-STAGE NON-SMALL-CELL LUNG CANCER (ES-NSCLC)

Currently, surgical resection remains the standard of care for patients with operable ES-NSCLC. Hence, initial workup of patients with newly diagnosed ES-NSCLC should include an assessment of a patient's eligibility for operative treatment. Preoperative evaluation should include pulmonary function testing with calculation of predicted postoperative (PPO) FEV1 and diffusing capacity for carbon monoxide (DLCO); cardiac workup may also be necessary depending on patient comorbidities (Brunelli et al. 2013). Patients with both PPO FEV1 and PPO DLCO >60% are considered low risk and may proceed to surgery without further testing. If patients have either PPO FEV1 or PPO DLCO <60%, further testing with an exercise test is recommended. Per National Comprehensive Cancer Network (NCCN) guidelines, initial workup should also include positron emission tomography/computed tomography (PET/CT) imaging and bronchoscopy; in peripheral tumors without suspicion for mediastinal lymph node involvement, bronchoscopy may be performed intraoperatively but PET/CT should still be performed prior to surgery.

In patients with ES-NSCLC who undergo complete resection of their tumors with negative margins and without findings of nodal involvement, there is no indication for postoperative radiotherapy (PORT). The PORT Meta-analysis Trialists Group analyzed individual patient data from nine prospective randomized trials with a total of 2128 patients randomized to PORT to 30–60 Gy versus surgery alone (PORT Meta-analysis Trialists Group 1998). The study demonstrated a statistically significant decrease in 2-yr overall survival (OS) in the PORT cohort (48% vs. 55%), with subgroup analysis finding the greatest detriment to be in patients with stage I/II NSCLC and in patients with N0-N1 disease. An update of the meta-analysis, which included data from an Italian trial, did not substantially change the results (Burdett and Stewart 2005). A retrospective analysis of 7465 patients with resected stage II or III NSCLC in the Surveillance, Epidemiology, and End Results (SEER) database also investigated the association between PORT and survival (Lally et al. 2006). On subset analysis based on nodal stage, the authors found a statistically significant decrease in survival in patients with N0 disease who underwent PORT (hazard ratio [HR] = 1.176; P = 0.0435). Margin status was unavailable within the database and was not included in the analysis.

For patients who are not surgical candidates due to underlying comorbidities, extent of disease, or patient preference, the preferred treatment is SBRT. Historically, patients with inoperable ES-NSCLC were treated with definitive intent, conventionally fractionated radiotherapy with limited success. In a review examining outcomes after treating ES-NSCLC with conventional radiotherapy alone, Qiao et al. (2003) analyzed 18 studies published between 1988 and 2000. The majority of the included studies were retrospective and local failure rates varied widely, ranging from 6.4% to 70% with the use of various conventional regimens. The median local recurrence rate among the reported studies was 40% with local failure listed as the most common cause for treatment failure. In an effort to improve local failure rates, Timmerman et al. (2003) initially explored the impact of dose escalation and hypofractionation using SBRT in a single institution phase I trial of 37 patients with T1-T2 (tumor size ≤7 cm), N0, M0 disease. Patients with T1 versus T2 disease were dose escalated separately from a starting dose of 24 Gy in three fractions to a maximum dose of 60 Gy in three fractions for T1 tumors and 66 Gy in three fractions for T2 tumors with neither cohort ultimately reaching the maximum tolerated dose (MTD) in tumors <5 cm. In patients with tumors measuring 5–7 cm, 72 Gy in three fractions was considered to be the dose-limiting regimen. In a subsequent phase II trial from the same group, 70 patients with T1-T2 (tumor size ≤7 cm), N0, M0 disease underwent SBRT to a total dose of 60 Gy in three fractions for T1 tumors and 66 Gy in three fractions for T2 tumors with excellent local control (LC) (Timmerman et al. 2006; Fakiris et al. 2009). The trial reported 2-yr and 3-yr LC rates of 95% and 88.1%, respectively. Median survival (MS), 3-yr OS, and 3-yr cancer-specific survival were 32.4 mo, 42.7%, and 81.7%, respectively. Protocol-specified grade 3–5 toxicities were reported in 11 patients with an additional six patients experiencing non-protocol-specified toxicities. Although not statistically significant, patients with central tumors, defined as tumors within the hilar/perihilar region, tended to have higher rates of grade 3–5 toxicities (10.4% in central tumors vs. 27.3% in peripheral tumors). In a large multi-institutional Japanese retrospective study of 257 patients with T1-T2 disease treated to a total of 18–75 Gy in 1–22 fractions and median follow-up of 38 mo, Onishi et al. (2004, 2007) reported a similarly low local progression rate of 14.0. The median calculated biological equivalent dose (BED) administered was 108 Gy. Patients receiving higher BED doses were found to have statistically significantly better LC and OS rates; local progression and 5-yr OS were 8.4% and 53.9%, respectively, for patients with BED ≥100 Gy versus 42.9% and 19.7%, respectively, for those with BED <100 Gy.

Following promising results from both retrospective studies and prospective single institution studies, the Radiation Therapy Oncology Group (RTOG) initiated RTOG 0236, a North American phase II multicenter trial investigating the use of SBRT in medically inoperable patients with T1-T2 (tumor size ≤5 cm), N0, M0 disease (Timmerman et al. 2010, 2018a). A total of 55 evaluable patients were treated to a total dose of 60 Gy in three fractions; dose was later corrected to 54 Gy in three fractions when accounting for tissue density heterogeneity. Given the prior concerns of increased toxicities in patients with central tumors, the study excluded patients with planning target volumes extending within 2 cm of the proximal bronchial tree, defined as the area including the distal trachea, carina, and proximal lobar bronchi to their first bifurcation. With median follow-up of 48 mo, 3-yr and 5-yr primary tumor control rates were 97.6% and 92.7%, respectively. LC, defined as no recurrence in the primary tumor or involved lobe, was 90.6% at 3 yr and 80.0% at 5 yr. Disease-free survival (DFS) and OS at 3 yr was 48.3% and 55.8%, respectively; at 5 yr, DFS and OS was 25.5% and 40.0%, respectively. Protocol-specified grade 3–4 treatment-related toxicities occurred in 17 patients (30.9%). No grade 5 toxicities were reported. As RTOG 0236 was limited to peripheral tumors, RTOG 0813 sought to establish MTD and assess toxicity and efficacy for SBRT in centrally located tumors (Bezjak et al. 2019). In this phase I/II dose escalation study, 100 patients with medically inoperable T1-T2 (tumor size ≤5 cm), N0, M0 centrally located tumors were accrued into five dose levels beginning at 50 Gy in five fractions and escalated by 0.5 Gy per fraction to a maximum dose of 60 Gy in five fractions. The majority of patients (n = 71) were accrued into the two highest dose levels. The study found the MTD to be 60 Gy in five fractions with a dose-limiting toxicity (DLT) rate of 7.2% at this dose. Although not considered DLT, four patients in the two highest dose levels experienced grade 5 toxicities more than a year after completing treatment. Median follow-up was 37.9 mo with 2-yr LC rates in the 11.5 Gy per fraction group and the 12 Gy per fraction group of 89.4% and 87.9%, respectively. Two-year OS and progression-free survival (PFS) was 67.9% and 52.2%, respectively, in the 11.5 Gy per fraction group and 72.7% and 54.5%, respectively, in the 12 Gy per fraction group. RTOG 0915, a randomized phase II trial of 84 evaluable patients with peripheral T1-T2, N0, M0 tumors, compared the impact of different SBRT schedules on toxicity and tumor control (Videtic et al. 2015, 2019). Patients were randomized to receive either 34 Gy in one fraction (Arm 1, n = 39) or 48 Gy in four fractions (Arm 2, n = 45). With a median follow-up of 4.0 yr, protocol-specified grade 3–5 toxicity rates were 2.6% in Arm 1 and 11.1% in Arm 2. Primary tumor failure rate at 5 yr was 10.6% in Arm 1 and 6.8% in Arm 2. There was no statistically significant difference in PFS or OS at 5 yr; 5-yr PFS and OS was19.1% and 29.6%, respectively, in Arm 1 and 33.3% and 41.1%, respectively, in Arm 2. However, the study was not powered to detect survival differences.

Recent prospective multicenter studies comparing outcomes from SBRT versus conventional radiotherapy have confirmed SBRT to be the standard of care in inoperable ES-NSCLC. SPACE, a Scandinavian multicenter randomized phase II trial, compared SBRT to 66 Gy in three fractions with conventional radiotherapy to 70 Gy in 35 fractions in 102 patients with inoperable peripheral T1-T2 (tumor size ≤6 cm), N0, M0 tumors (Nyman et al. 2016). Median follow-up was 37 mo with no statistically significant difference in PFS, OS, or LC between the groups. Of note, the study was underpowered to detect differences in PFS, and there was a trend toward better disease control with SBRT (70% with SBRT vs. 59% with conventional radiotherapy). Despite the SBRT cohort having significantly more T2 disease, patients treated with SBRT exhibited significantly less toxicity and reported improved quality of life. CHISEL, a multicenter phase III trial from Australia and New Zealand, randomized 101 patients with inoperable peripheral T1-T2, N0, M0 tumors in a 2:1 ratio to either SBRT or conventional radiotherapy (Ball et al. 2019). Patients in the SBRT arm received 54 Gy in three fractions or 48 Gy in four fractions (if the tumor was <2 cm from the chest wall) while those in the conventional radiotherapy arm were treated with 66 Gy in 33 fractions or 50 Gy in 20 fractions depending on institutional preference. With a median follow-up of 2.6 yr for the SBRT arm and 2.1 yr for the conventional radiotherapy arm, the authors reported a statistically significant improvement in both LC (HR 0.32) and OS (HR 0.53) with SBRT.

In recent years, a number of studies have also addressed the use of SBRT in operable patients. In a Japanese retrospective multi-institutional study of 87 patients with operable T1-T2, N0, M0 disease treated with SBRT to a total dose of 45–72.5 Gy in 3–10 fractions, Onishi et al. (2011) reported outcomes potentially comparable with surgical resection. With a median follow-up of 55 mo, 5-yr LC for the total cohort was 86.7%; 5-yr LC for T1 and T2 tumors was 92% and 73%, respectively. OS at 5 yr was 69.5% for the total cohort with 5-yr OS of 72% and 62% for patients with T1 and T2 tumors, respectively. In a meta-analysis of 40 SBRT studies (n = 4850) and 23 surgery studies (n = 7071) examining treatment outcomes for T1-T2, N0, M0 disease, Zheng et al. (2014) also reported similar LC, OS, and DFS for SBRT and surgery after adjusting for the proportion of operable patients and patient age. RTOG 0618 was a North American multicenter single-arm phase II study, which prospectively investigated the use of SBRT in patients with operable ES-NSCLC (Timmerman et al. 2018b). A total of 26 evaluable patients with operable biopsy-proven peripheral T1-T2 (tumor size ≤5 cm), N0, M0 tumors were treated with SBRT to a total dose of 54 Gy in three fractions. With a median follow-up of 48.1 mo, the study reported excellent rates of primary tumor control. Both 4-yr primary tumor control rate and LC rate were 96% with a 4-yr local-regional control (LRC) rate of 88%. MS was 55.2 mo with 4-yr DFS and OS of 57% and 56%, respectively. Protocol-specified grade 3 toxicities were reported in two patients (8%) with an additional two patients experiencing non-protocol-specified toxicities. No grade 4 or 5 toxicities were reported. A pooled analysis of STARS and ROSEL, two randomized phase III trials comparing SBRT and surgery that closed early due to poor accrual, has reported significantly better OS and lower rates of treatment-related toxicities with SBRT (Chang et al. 2015). In these trials, a total of 58 patients with T1-2a, N0, M0 tumors were randomized to either SBRT (n = 31) or lobectomy with mediastinal lymph node dissection or sampling (n = 27). Patients on the SBRT arm were treated to a total dose of 54 Gy in three fractions or 60 Gy in five fractions for peripheral tumors and to a dose of 50 Gy in four fractions for central tumors. With a median follow-up of 40.2 mo for the SBRT arm and 35.4 mo for the surgery arm, the authors reported a 3-yr OS of 95% for the SBRT arm versus 79% for the surgery arm. The rate of treatment-related grade 3–5 toxicities was 10% in the SBRT arm compared to 48% in the surgery arm; of note, no grade 4–5 toxicities were reported in the SBRT arm. There was no significant difference in local, regional, or distant failure between the two arms. Although promising, these results are met with skepticism given the small sample size and high rate of treatment-related toxicities in the surgical arm (compared to historical controls).

Several ongoing prospective randomized trials are also directly comparing SBRT to surgery. In the United States, the Joint Lung Cancer Trialist's Coalition (JoLT-Ca) STABLEMATES trial, a randomized phase III trial comparing SBRT to sublobar resection in high-risk patients with operable stage I NSCLC, is currently accruing patients. The Veterans Affairs Lung Cancer Surgery or Stereotactic Radiotherapy (VALOR) trial, another randomized trial comparing SBRT to surgery with lobectomy or segmentectomy in patients with stage I NSCLC, is also underway but is currently on temporary recruitment hold. In China, the Radical Resection vs. Ablative Stereotactic Radiotherapy in Patients with Operable Stage I NSCLC (POSTLIV) trial is a randomized phase II trial comparing SBRT to radical resection in patients with T1, N0, M0 peripheral tumors and is also currently accruing patients.

LOCALLY ADVANCED NON-SMALL-CELL LUNG CANCER (LA-NSCLC)

Resectable

Primary disease extension and nodal involvement determines the resectability of LA-NSCLC. The initial workup typically includes PET/CT followed by endoscopic ultrasound/endobronchial ultrasound as needed to rule out potential false negative findings from PET/CT imaging (Dong et al. 2013). Surgical candidates will likely have T3-4, N0-1 disease; patients with N2 disease are considered surgical candidates in select cases (single nodal station location with disease <3 cm). The PORT meta-analysis, retrospective analysis of the adjuvant Navelbine International Trialist Association (ANITA) randomized trial data, and several large database analyses have sought to establish the role PORT in resectable LA-NSCLC (PORT Meta-analysis Trialists Group 1998; Burdett and Stewart 2005; Lally et al. 2006; Douillard et al. 2008; Robinson et al. 2015). While the PORT meta-analysis demonstrated a detriment to OS in ES-NSCLC, the updated analysis showed that OS in stage III patients with N2 disease was unaffected. These data included an era where patients received much higher doses of radiation to the heart and lungs. In contrast, an OS benefit was shown in the ANITA, SEER, and NCBD analyses for patients with N2 disease who received PORT. ANITA was a phase III trial that randomized patients with stage IB-IIIA NSCLC who underwent surgical resection to receive adjuvant cisplatin and vinorelbine versus observation; PORT was recommended in patients with pathologically confirmed nodal disease. Approximately 33% and 22% of patients in the experimental and observation arms, respectively, received PORT; PORT improved MS for patients with pN2 disease in both the experimental and observation arms (47.4 vs. 23.8 mo and 22.7 vs. 12.7 mo). The Lung Adjuvant Radiotherapy Trial (ART) (NCT00410683) is a current phase III, randomized trial comparing PORT to no PORT in patients with completely resected NSCLC with N2 disease; results were presented at the European Society for Medical Oncology 2020 Annual Meeting (Le Pechoux et al. 2020) and showed no survival advantage to PORT for pN2 patients, so its use is being reconsidered. American Society for Radiation Oncology (ASTRO) guidelines still recommend PORT following chemotherapy in patients incidentally found to have R1 resections or N2 disease. The ANITA data demonstrates that patients with N1 disease who are not receiving adjuvant chemotherapy should be considered candidates for PORT as well. The dose of radiation should be escalated in the presence of residual disease (50–54 Gy for R0 resection, 54–60 Gy for R1 or extracapsular extension, 60+ Gy for gross disease) (Douillard et al. 2008; Rodrigues et al. 2015). Recommended clinical target volumes can be found in the Lung ART protocol (NCT00410683).

Patients with known T1-3, pN2 disease who are otherwise healthy are candidates for trimodality therapy (TMT), which includes neoadjuvant cisplatin/etoposide and radiation followed by resection and adjuvant cisplatin/etoposide. This regimen is based on the Intergroup 0139 trial, which compared definitive chemoradiation therapy to TMT. TMT remains controversial as results showed improved PFS but did not affect OS (mortality was higher in patients who underwent pneumonectomies) (Albain et al. 2009).

Unresectable

The gold standard for patients with unresectable LA-NSCLC is definitive chemoradiation. RTOG 7301 established 60 Gy in 30 fractions as a minimum dose for the management of LA-NSCLC in the 1970s (Perez et al. 1980). CALGB 8433 was one of the first phase III randomized trials that showed the addition of sequential chemotherapy (cisplatin and vinblastine) to radiation improved PFS and OS in patients with stage III NSCLC (Dillman et al. 1996). RTOG 9410 reported improved OS in patients receiving concurrent chemoradiation therapy compared to sequential chemoradiation therapy (MS 17 mo vs. 14.6 mo, respectively); however, there were higher incidences of acute esophagitis in the concurrent chemoradiation arm (Curran et al. 2011). Improved OS (4.5% absolute survival benefit at 5 yr) at the expense of increased esophageal toxicity (18% vs. 4%) was similarly reported in a large meta-analysis (Aupérin et al. 2010).

There have been several trials that have evaluated the impact of dose escalation (Bradley et al. 2005, 2015; Rosenzweig et al. 2005). RTOG 0617 was a phase III trial with a 2 × 2 design that randomized patients to 60 Gy in 30 fractions or 74 Gy in 37 fractions with concurrent carboplatin and paclitaxel followed by adjuvant carboplatin and paclitaxel (± cetuximab). Patients in the dose-escalated arm had worse OS, higher incidence of grade 3 toxicities, and worse quality of life as measured by the Functional Assessment of Cancer Therapy-Lung Cancer Subscale (FACT-LCS) (Bradley et al. 2015; Movsas et al. 2016). These results were surprising as prior studies had shown higher doses were safely tolerated. Concerns were raised that practitioners underdosed targets to meet protocol constraints and this may have led to worse oncologic outcomes. Subanalyses from this trial have provided important data such as the benefit of using intensity-modulated radiation therapy (vs. 3D conformal) to reduce the heart V40, a marker that was associated with increased mortality (Chun et al. 2017).

Consolidative durvalumab has recently been added to the standard of care regimen given the positive results reported from the PACIFIC trial published in 2017. This trial showed consolidative durvalumab (vs. placebo) improved median PFS (18.1 mo vs. 5.6 mo) and OS (2-yr 83.1% vs. 74.6%, respectively; 3-yr 66.3% vs. 55.6%, respectively) in patients with unresectable LA-NSCLC previously treated with concurrent chemoradiation (Antonia et al. 2017, 2018; Gray et al. 2020). A subset analysis showed no benefit in patients whose tumors were programmed death-ligand 1 negative, so its use in that subset of patients is controversial. Similar rates of grade 3 or 4 toxicities were reported between the two groups, and therapy discontinuation occurred in 15.4% of patients in the experimental arm versus 9.8% of patients in the placebo arm. Separately, other institutions have seen as many as 30% of patients not initiating durvalumab because of rapid disease progression or toxicities due to chemoradiation (Sakaguchi et al. 2019; Shaverdian et al. 2020). Pneumonitis in particular can lead to many patients either not starting durvalumab or discontinuing durvalumab therapy.

Groups are exploring the use of SBRT and proton therapy for treating patients with LA-NSCLC. Lee et al. published their tolerability outcomes from their phase II prospective trial evaluating 28 patients treated with hypofractionated chemoradiation therapy (40 Gy in 10 fractions) followed by a dose-escalated SBRT boost (5 Gy × 5, 6 Gy × 5, or 7 Gy × 5); the 70 Gy in 15 fractions regimen was determined to be the MTD, and OS was similar to historical controls (Lee et al. 2019). Phase I and II studies have investigated the use of protons in definitive chemoradiation therapy for patients with LA-NSCLC; both trials were closed early due to poor accrual. There were minimal treatment-related events (Hoppe et al. 2016, 2020).

STAGE IV AND OLIGOMETASTATIC NSCLC

Metastatic NSCLC (mNSCLC) has a poor prognosis with only 5% of patients living past 5 yr. Systemic therapies, including immunotherapies such as pembrolizumab, are the standard of care with radiation therapy historically held in reserve for palliative treatments (Reck et al. 2019). Over the last several decades, SBRT has been used for patients with limited metastatic disease known as oligometastatic disease. For these patients, aggressive local therapies could provide long-term disease control or even cure (Hellman and Weichselbaum 1995; Pastorino et al. 1997). Many clinical trials have used various definitions to clinically outline the patient characteristics that may reflect oligometastatic disease; the reader can review the studies cited, but, in summary, patients typically have fewer than six targetable lesions in fewer than four sites (Gomez et al. 2016; Iyengar et al. 2018; Dingemans et al. 2019).

Early evidence suggesting the benefit of aggressive local therapy for patients with oligometastatic disease stems from surgical literature (Patchell et al. 1990; Patrini et al. 2018). As not all patients are candidates for surgery or are willing to undergo surgery, SBRT is an effective alternative modality that significantly reduces and even eliminates recovery time; this is particularly beneficial for those needing to return to systemic therapy quickly. Gomez et al. (2016, 2019) and Iyengar et al. (2018) were some of the first to publish randomized data evaluating the use of SBRT in oligometastatic NSCLC. Gomez et al. reported on 49 patients with ≤3 metastatic lesions after first-line systemic therapy (four or more cycles of platinum doublet therapy or three or more months of epidermal growth factor receptor or anaplastic lymphoma kinase inhibitors) who received maintenance treatment ± local therapy (SBRT, chemoradiation, or surgery). The study closed early after the local therapy arm was noted to have superior PFS (11.9 mo vs. 3.9 mo). These results were later supported and augmented when longer follow-up showed both a PFS and OS benefit (median OS 41.2 mo vs. 17.0 mo; median PFS 14.2 mo vs. 4.4 mo at a median follow-up of 38.8 mo). Iyengar et al. explored a similar cohort of 29 patients that had nonprogressive oligometastatic disease (primary disease plus ≤5 metastatic sites with ≤3 sites in the liver or lung) who had already received 4–6 cycles of first-line platinum-based chemotherapy randomized to maintenance therapy ± SBRT. This trial was also closed early when interim analysis showed superior PFS in the SBRT arm (9.7 mo vs. 3.5 mo). These data are encouraging; however, extrapolation to a more generalized population is limited since both study cohorts were small (47 and 29 patients, respectively) and patients did not receive immunotherapy.

The results from SABR-COMET, a phase II randomized trial, were recently published. Ninety-nine patients with oligometastatic cancer of various histologies (18 patients with NSCLC) were randomized to receive standard palliative treatment ± SBRT to all sites of metastatic disease; results were notable for improved PFS and OS in patients receiving SBRT along with standard palliative treatment (Palma et al. 2019). We currently await the results of NRG-LU-002 (NCT03137771) and SARON (NCT02417662); both are randomized, multicenter, phase III trials properly powered to detect an OS difference between patients receiving maintenance therapy ± SBRT.

Distinct from oligometastatic disease, some individuals may have more stable widespread metastatic disease with only a few progressive foci. This is referred to as the oligoprogressive state; currently, there is limited data in this patient subset. One single arm phase II trial evaluated 24 patients with less than seven sites of progressive disease following platinum-based systemic therapy who received erlotinib and SBRT. The addition of SBRT allowed patients to continue systemic therapy for an additional 6–9 mo; moreover, median PFS and OS were 14.7 mo and 20.4 mo, respectively, improved compared to historical controls (Iyengar et al. 2014).

Immunotherapies, such as pembrolizumab, are currently incorporated into the standard of care for patients with mNSCLC after the publication of multiple encouraging trials such as KEYNOTE-24. KEYNOTE-24 was a randomized phase III trial that compared the use of platinum-based chemotherapy versus pembrolizumab for patients with metastatic disease with improved survival seen in patients receiving pembrolizumab (30.0 mo vs. 14.2 mo) (Reck et al. 2019). More recently, there have been results published that evaluated the use of immunotherapies in addition to SBRT. One phase II, single-arm study published by Bauml et al. evaluated 45 patients treated with pembrolizumab after undergoing localized therapy (resection, chemoradiation, radiofrequency ablation). Median PFS was 19.1 mo, which was higher than historical controls and OS was 41.6 mo (Bauml et al. 2019). A phase II trial randomized patients with mNSCLC to pembrolizumab or SBRT then pembrolizumab and showed patients in the SBRT arm had improved OS and PFS (15.9 mo vs. 7.6 mo and 6.6 mo vs. 1.9 mo, respectively) (Theelen et al. 2019). There are currently no published phase III randomized data evaluating immunotherapy with SBRT. However, there was a phase III trial comparing patients with ≤5 sites of oligometastatic NSCLC treated with tyrosine kinase inhibitors (TKIs) to those receiving TKIs + SBRT was recently presented as an abstract at ASCO 2020. Patients in the SBRT arm had improved PFS (20.2 mo vs. 12.5 mo, median follow-up 19.6 mo) (Wang et al. 2020).

CONCLUSION

The management of NSCLC has evolved throughout the years. Initially, patients with ES-NSCLC were managed with surgical resection alone. Decades of evidence now support the use of SBRT in inoperable patients. The LC rates after SBRT for ES-NSCLC are comparable to surgical results; however, data comparing the two are sparse. We eagerly await the results from trials such as STABLEMATES to address this further. The treatment of LA-NSCLC is stratified according to resectability. Patients with resectable disease may require PORT if the patient underwent an R1 resection or had N2 disease. There is even evidence to support the use of PORT in patients with N1 disease who will not be receiving chemotherapy. Patients with LA-NSCLC but still resectable disease may also be managed with TMT; however, this is controversial. There are several phase II trials reporting the utility of SBRT and systemic therapy for treating patients with oligometastatic disease. Currently, we await the results from the phase III trials, NRG-LU-002 (NCT03137771) and SARON (NCT02417662), to see whether there is an OS benefit with the combination of systemic therapy and SBRT in this patient subset.

Footnotes

Editors: Christine M. Lovly, David P. Carbone, and John D. Minna

Additional Perspectives on Lung Cancer: Disease Biology and Its Potential for Clinical Translation available at www.perspectivesinmedicine.org

REFERENCES

  1. Alboin KS, Swann RS, Rusch VW, Turrisi AT III, Shepherd FA, Smith C, Chen Y, Livingston RB, Feins RH, Gandara DR, et al. 2009. Radiotherapy plus chemotherapy with or without surgical resection for stage III non-small-cell lung cancer: a phase III randomised controlled trial. Lancet 374: 379–386. 10.1016/S0140-6736(09)60737-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, et al. 2017. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 377: 1919–1929. 10.1056/NEJMoa1709937 [DOI] [PubMed] [Google Scholar]
  3. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Kurata T, Chiappori A, Lee KH, de Wit M, et al. 2018. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med 379: 2342–2350. 10.1056/NEJMoa1809697 [DOI] [PubMed] [Google Scholar]
  4. Aupérin A, Le Péchoux C, Rolland E, Curran WJ, Furuse K, Fournel P, Belderbos J, Clamon G, Ulutin HC, Paulus R, et al. 2010. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol 28: 2181–2190. 10.1200/JCO.2009.26.2543 [DOI] [PubMed] [Google Scholar]
  5. Ball D, Mai GT, Vinod S, Babington S, Ruben J, Kron T, Chesson B, Herschtal A, Vanevski M, Rezo A, et al. 2019. Stereotactic ablative radiotherapy versus standard radiotherapy in stage 1 non-small-cell lung cancer (TROG 09.02 CHISEL): a phase 3, open-label, randomised controlled trial. Lancet Oncol 20: 494–503. 10.1016/S1470-2045(18)30896-9 [DOI] [PubMed] [Google Scholar]
  6. Bauml JM, Mick R, Ciunci C, Aggarwal C, Davis C, Evans T, Deshpande C, Miller L, Patel P, Alley E, et al. 2019. Pembrolizumab after completion of locally ablative therapy for oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol 5: 1283–1290. 10.1001/jamaoncol.2019.1449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bezjak A, Paulus R, Gaspar LE, Timmerman RD, Straube WL, Ryan WF, Garces YI, Pu AT, Singh AK, Videtic GM, et al. 2019. Safety and efficacy of a five-fraction stereotactic body radiotherapy schedule for centrally located non-small-cell lung cancer: NRG Oncology/RTOG 0813 Trial. J Clin Oncol 37: 1316–1325. 10.1200/JCO.18.00622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa WH, Ryu JK, Bosch W, Emami B. 2005. Toxicity and outcome results of RTOG 9311: a phase I-II dose-escalation study using three-dimensional conformal radiotherapy in patients with inoperable non-small-cell lung carcinoma. Int J Radiat Oncol Biol Phys 61: 318–328. 10.1016/j.ijrobp.2004.06.260 [DOI] [PubMed] [Google Scholar]
  9. Bradley JD, Paulus R, Komaki R, Masters G, Blumenschein G, Schild S, Bogart J, Hu C, Forster K, Magliocco A, et al. 2015. 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 16: 187–199. 10.1016/S1470-2045(14)71207-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brunelli A, Kim AW, Berger KI, Addrizzo-Harris DJ. 2013. Physiologic evaluation of the patient with lung cancer being considered for resectional surgery: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 143: e166S–e190S. 10.1378/chest.12-2395 [DOI] [PubMed] [Google Scholar]
  11. Burdett S, Stewart L. 2005. Postoperative radiotherapy in non-small-cell lung cancer: update of an individual patient data meta-analysis. Lung Cancer 47: 81–83. 10.1016/j.lungcan.2004.09.010 [DOI] [PubMed] [Google Scholar]
  12. Chang JY, Senan S, Paul MA, Mehran RJ, Louie AV, Balter P, Groen HJ, McRae SE, Widder J, Feng L, et al. 2015. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol 16: 630–637. 10.1016/S1470-2045(15)70168-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chun SG, Hu C, Choy H, Komaki RU, Timmerman RD, Schild SE, Bogart JA, Dobelbower MC, Bosch W, Galvin JM, et al. 2017. Impact of intensity-modulated radiation therapy technique for locally advanced non-small-cell lung cancer: a secondary analysis of the NRG oncology RTOG 0617 randomized clinical trial. J Clin Oncol 35: 56–62. 10.1200/JCO.2016.69.1378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Curran WJ, Paulus R, Langer CJ, Komaki R, Lee JS, Hauser S, Movsas B, Wasserman T, Rosenthal SA, Gore E, et al. 2011. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst 103: 1452–1460. 10.1093/jnci/djr325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR. 1996. Improved survival in stage III non-small-cell lung cancer: seven-year follow-up of cancer and leukemia group B (CALGB) 8433 trial. J Natl Cancer Inst 88: 1210–1215. 10.1093/jnci/88.17.1210 [DOI] [PubMed] [Google Scholar]
  16. Dingemans AC, Hendriks LEL, Berghmans T, Levy A, Hasan B, Faivre-Finn C, Giaj-Levra M, Giaj-Levra N, Girard N, Greillier L, et al. 2019. Definition of synchronous oligometastatic non-small cell lung cancer—a consensus report. J Thorac Oncol 14: 2109–2119. 10.1016/j.jtho.2019.07.025 [DOI] [PubMed] [Google Scholar]
  17. Dong X, Qiu X, Liu Q, Jia J. 2013. Endobronchial ultrasound-guided transbronchial needle aspiration in the mediastinal staging of non-small cell lung cancer: a meta-analysis. Ann Thorac Surg 96: 1502–1507. 10.1016/j.athoracsur.2013.05.016 [DOI] [PubMed] [Google Scholar]
  18. Douillard JY, Rosell R, De Lena M, Riggi M, Hurteloup P, Mahe MA. 2008. Impact of postoperative radiation therapy on survival in patients with complete resection and stage I, II, or IIIA non-small-cell lung cancer treated with adjuvant chemotherapy: the adjuvant Navelbine International Trialist Association (ANITA) Randomized Trial. Int J Radiat Oncol Biol Phys 72: 695–701. 10.1016/j.ijrobp.2008.01.044 [DOI] [PubMed] [Google Scholar]
  19. Fakiris AJ, McGarry RC, Yiannoutsos CT, Papiez L, Williams M, Henderson MA, Timmerman R. 2009. Stereotactic body radiation therapy for early-stage non-small-cell lung carcinoma: four-year results of a prospective phase II study. Int J Radiat Oncol Biol Phys 75: 677–682. 10.1016/j.ijrobp.2008.11.042 [DOI] [PubMed] [Google Scholar]
  20. Gomez DR, Blumenschein GR, Lee JJ, Hernandez M, Ye R, Camidge DR, Doebele RC, Skoulidis F, Gaspar LE, Gibbons DL, et al. 2016. Local consolidative therapy versus maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer without progression after first-line systemic therapy: a multicentre, randomised, controlled, phase 2 study. Lancet Oncol 17: 1672–1682. 10.1016/S1470-2045(16)30532-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Gomez DR, Tang C, Zhang J, Blumenschein GR, Hernandez M, Lee JJ, Ye R, Palma DA, Louie AV, Camidge DR, et al. 2019. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol 37: 1558–1565. 10.1200/JCO.19.00201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Gray JE, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Kurata T, Chiappori A, Lee KH, Cho BC, et al. 2020. Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC—update from PACIFIC. J Thorac Oncol 15: 288–293. 10.1016/j.jtho.2019.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Hellman S, Weichselbaum RR. 1995. Oligometastases. J Clin Oncol 13: 8–10. 10.1200/JCO.1995.13.1.8 [DOI] [PubMed] [Google Scholar]
  24. Hoppe BS, Henderson R, Pham D, Cury JD, Bajwa A, Morris CG, D'Agostino H, Flampouri S, Huh S, Li Z, et al. 2016. A phase 2 trial of concurrent chemotherapy and proton therapy for stage III non-small cell lung cancer: results and reflections following early closure of a single-institution study. Int J Radiat Oncol Biol Phys 95: 517–522. 10.1016/j.ijrobp.2015.11.004 [DOI] [PubMed] [Google Scholar]
  25. Hoppe BS, Nichols RC, Flampouri S, Li Z, Morris CG, Pham DC, Mohindra P, Hartsell W, Mohammed N, Chon BH, et al. 2020. Hypofractionated proton therapy with concurrent chemotherapy for locally advanced non-small cell lung cancer: a Phase 1 Trial from the University of Florida and Proton Collaborative Group. Int J Radiat Oncol Biol Phys 107: 455–461. 10.1016/j.ijrobp.2020.03.015 [DOI] [PubMed] [Google Scholar]
  26. Iyengar P, Kavanagh BD, Wardak Z, Smith I, Ahn C, Gerber DE, Dowell J, Hughes R, Abdulrahman R, Camidge DR, et al. 2014. Phase II trial of stereotactic body radiation therapy combined with erlotinib for patients with limited but progressive metastatic non-small-cell lung cancer. J Clin Oncol 32: 3824–3830. 10.1200/JCO.2014.56.7412 [DOI] [PubMed] [Google Scholar]
  27. Iyengar P, Wardak Z, Gerber DE, Tumati V, Ahn C, Hughes RS, Dowell JE, Cheedella N, Nedzi L, Westover KD, et al. 2018. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol 4: e173501. 10.1001/jamaoncol.2017.3501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Lally BE, Zelterman D, Colasanto JM, Haffty BG, Detterbeck FC, Wilson LD. 2006. Postoperative radiotherapy for stage II or III non-small-cell lung cancer using the surveillance, epidemiology, and end results database. J Clin Oncol 24: 2998–3006. 10.1200/JCO.2005.04.6110 [DOI] [PubMed] [Google Scholar]
  29. Lee P, Luterstein E, Goldman J, et al. 2019. Accelerated hypofractionated CRT followed by SABR boost (HyCRT-SABR) for locally advanced unresectable NSCLC: a prospective phase II radiation dose-escalation study. Int J Radiat Oncol 105: S44. 10.1016/j.ijrobp.2019.06.469 [DOI] [Google Scholar]
  30. Le Pechoux C, Pourel N, Barlesi F, Faivre-Finn C, Lerouge D, Zalcman G, Antoni D, Lamezec B, Nestle U, Boisselier P, et al. 2020. LBA3_PR an international randomized trial, comparing post-operative conformal radiotherapy (PORT) to no PORT, in patients with completely resected non-small cell lung cancer (NSCLC) and mediastinal N2 involvement: primary end-point analysis of LungART (IFCT-0503, UK NCRI, SAKK) NCT00410683. Ann Oncol 31: S1178–S1215. 10.1016/j.annonc.2020.08.2280 [DOI] [Google Scholar]
  31. Movsas B, Hu C, Sloan J, Bradley J, Komaki R, Masters G, Kavadi V, Narayan S, Michalski J, Johnson DW, et al. 2016. Quality of life analysis of a radiation dose-escalation study of patients with non-small-cell lung cancer: a secondary analysis of the radiation therapy oncology group 0617 randomized clinical trial. JAMA Oncol 2: 359–367. 10.1001/jamaoncol.2015.3969 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Nyman J, Hallqvist A, Lund J, Brustugun OT, Bergman B, Bergström P, Friesland S, Lewensohn R, Holmberg E, Lax I. 2016. SPACE—a randomized study of SBRT vs conventional fractionated radiotherapy in medically inoperable stage I NSCLC. Radiother Oncol 121: 1–8. 10.1016/j.radonc.2016.08.015 [DOI] [PubMed] [Google Scholar]
  33. Onishi H, Araki T, Shirato H, Nagata Y, Hiraoka M, Gomi K, Yamashita T, Niibe Y, Karasawa K, Hayakawa K, et al. 2004. Stereotactic hypofractionated high-dose irradiation for stage I nonsmall cell lung carcinoma: clinical outcomes in 245 subjects in a Japanese multiinstitutional study. Cancer 101: 1623–1631. 10.1002/cncr.20539 [DOI] [PubMed] [Google Scholar]
  34. Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, Niibe Y, Karasawa K, Hayakawa K, Takai Y, et al. 2007. Hypofractionated stereotactic radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study. J Thorac Oncol 2: S94–S100. 10.1097/JTO.0b013e318074de34 [DOI] [PubMed] [Google Scholar]
  35. Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, Karasawa K, Hayakawa K, Niibe Y, Takai Y, et al. 2011. 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 81: 1352–1358. 10.1016/j.ijrobp.2009.07.1751 [DOI] [PubMed] [Google Scholar]
  36. Palma DA, Olson R, Harrow S, Gaede S, Louie AV, Haasbeek C, Mulroy L, Lock M, Rodrigues GB, Yaremko BP, et al. 2019. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): a randomised, phase 2, open-label trial. Lancet 393: 2051–2058. 10.1016/S0140-6736(18)32487-5 [DOI] [PubMed] [Google Scholar]
  37. Pastorino U, Buyse M, Friedel G, Ginsberg RJ, Girard P, Goldstraw P, Johnston M, McCormack P, Pass H, Putnam JB Jr. 1997. Long-term results of lung metastasectomy: prognostic analyses based on 5206 cases. J Thorac Cardiovasc Surg 113: 37–49. 10.1016/S0022-5223(97)70397-0 [DOI] [PubMed] [Google Scholar]
  38. Patchell RA, Tibbs PA, Walsh JW, Dempsey RJ, Maruyama Y, Kryscio RJ, Markesbery WR, Macdonald JS, Young B. 1990. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 322: 494–500. 10.1056/NEJM199002223220802 [DOI] [PubMed] [Google Scholar]
  39. Patrini D, Panagiotopoulos N, Bedetti B, Mitsos S, Crisci R, Solli P, Bertolaccini L, Scarci M. 2018. Surgical approach in oligometastatic non-small cell lung cancer. Ann Transl Med 6: 93. 10.21037/atm.2018.02.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Perez CA, Stanley K, Rubin P, Kramer S, Brady L, Perez-Tamayo R, Brown GS, Concannon J, Rotman M, Seydel HG. 1980. A prospective randomized study of various irradiation doses and fractionation schedules in the treatment of inoperable non-oat-cell carcinoma of the lung. Preliminary Report by the Radiation Therapy Oncology Group. Cancer 45: 2744–2753. [DOI] [PubMed] [Google Scholar]
  41. PORT Meta-analysis Trialists Group. 1998. Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. PORT meta-analysis trialists group. Lancet 352: 257–263. 10.1016/S0140-6736(98)06341-7 [DOI] [PubMed] [Google Scholar]
  42. Qiao X, Tullgren O, Lax I, Sirzén F, Lewensohn R. 2003. The role of radiotherapy in treatment of stage I non-small cell lung cancer. Lung Cancer 41: 1–11. 10.1016/S0169-5002(03)00152-1 [DOI] [PubMed] [Google Scholar]
  43. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al. 2019. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol 37: 537–546. 10.1200/JCO.18.00149 [DOI] [PubMed] [Google Scholar]
  44. Robinson CG, Patel AP, Bradley JD, DeWees T, Waqar SN, Morgensztern D, Baggstrom MQ, Govindan R, Bell JM, Guthrie TJ, et al. 2015. Postoperative radiotherapy for pathologic N2 non-small-cell lung cancer treated with adjuvant chemotherapy: a review of the National Cancer Data Base. J Clin Oncol 33: 870–876. 10.1200/JCO.2014.58.5380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Rodrigues G, Choy H, Bradley J, Rosenzweig KE, Bogart J, Curran WJ, Gore E, Langer C, Louie AV, Lutz S, et al. 2015. Adjuvant radiation therapy in locally advanced non-small cell lung cancer: executive summary of an American Society for Radiation Oncology (ASTRO) evidence-based clinical practice guideline. Pract Radiat Oncol 5: 149–155. 10.1016/j.prro.2015.02.013 [DOI] [PubMed] [Google Scholar]
  46. Rosenzweig KE, Fox JL, Yorke E, Amols H, Jackson A, Rusch V, Kris MG, Ling CC, Leibel SA. 2005. Results of a phase I dose-escalation study using three-dimensional conformal radiotherapy in the treatment of inoperable nonsmall cell lung carcinoma. Cancer 103: 2118–2127. 10.1002/cncr.21007 [DOI] [PubMed] [Google Scholar]
  47. Sakaguchi T, Ito K, Furuhashi K, Nakamura Y, Suzuki Y, Nishii Y, Taguchi O, Hataji O. 2019. Patients with unresectable stage III non-small cell lung cancer eligible to receive consolidation therapy with durvalumab in clinical practice based on PACIFIC study criteria. Respir Investig 57: 466–471. 10.1016/j.resinv.2019.03.011 [DOI] [PubMed] [Google Scholar]
  48. Shaverdian N, Offin MD, Rimner A, Shepherd AF, Wu AJ, Rudin CM, Hellmann MD, Chaft JE, Gomez DR. 2020. Utilization and factors precluding the initiation of consolidative durvalumab in unresectable stage III non-small cell lung cancer. Radiother Oncol 144: 101–104. 10.1016/j.radonc.2019.11.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Theelen W, Peulen HMU, Lalezari F, van der Noort V, de Vries JF, Aerts J, Dumoulin DW, Bahce I, Niemeijer AN, de Langen AJ, et al. 2019. Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol 5: 1276–1282. 10.1001/jamaoncol.2019.1478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Timmerman R, Papiez L, McGarry R, Likes L, DesRosiers C, Frost S, Williams M. 2003. Extracranial stereotactic radioablation: results of a phase I study in medically inoperable stage I non-small cell lung cancer. Chest 124: 1946–1955. 10.1378/chest.124.5.1946 [DOI] [PubMed] [Google Scholar]
  51. Timmerman R, McGarry R, Yiannoutsos C, Papiez L, Tudor K, DeLuca J, Ewing M, Abdulrahman R, DesRosiers C, Williams M, et al. 2006. Excessive toxicity when treating central tumors in a phase II study of stereotactic body radiation therapy for medically inoperable early-stage lung cancer. J Clin Oncol 24: 4833–4839. 10.1200/JCO.2006.07.5937 [DOI] [PubMed] [Google Scholar]
  52. Timmerman R, Paulus R, Galvin J, Michalski J, Straube W, Bradley J, Fakiris A, Bezjak A, Videtic G, Johnstone D, et al. 2010. Stereotactic body radiation therapy for inoperable early stage lung cancer. JAMA 303: 1070–1076. 10.1001/jama.2010.261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Timmerman RD, Hu C, Michalski JM, Bradley JC, Galvin J, Johnstone DW, Choy H. 2018a. Long-term results of stereotactic body radiation therapy in medically inoperable stage I non-small cell lung cancer. JAMA Oncol 4: 1287–1288. 10.1001/jamaoncol.2018.1258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Timmerman RD, Paulus R, Pass HI, Gore EM, Edelman MJ, Galvin J, Straube WL, Nedzi LA, McGarry RC, Robinson CG, et al. 2018b. Stereotactic body radiation therapy for operable early-stage lung cancer: findings from the NRG oncology RTOG 0618 trial. JAMA Oncol 4: 1263–1266. 10.1001/jamaoncol.2018.1251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Videtic GM, Hu C, Singh AK, Chang JY, Parker W, Olivier KR, Schild SE, Komaki R, Urbanic JJ, Timmerman RD, et al. 2015. A randomized phase 2 study comparing 2 stereotactic body radiation therapy schedules for medically inoperable patients with stage I peripheral non-small cell lung cancer: NRG oncology RTOG 0915 (NCCTG N0927). Int J Radiat Oncol Biol Phys 93: 757–764. 10.1016/j.ijrobp.2015.07.2260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Videtic GM, Paulus R, Singh AK, Chang JY, Parker W, Olivier KR, Timmerman RD, Komaki RR, Urbanic JJ, Stephans KL, et al. 2019. Long-term follow-up on NRG oncology RTOG 0915 (NCCTG N0927): a randomized phase 2 study comparing 2 stereotactic body radiation therapy schedules for medically inoperable patients with stage I peripheral non-small cell lung cancer. Int J Radiat Oncol Biol Phys 103: 1077–1084. 10.1016/j.ijrobp.2018.11.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Wang X, Bai YF, Zeng M. 2020. First-line tyrosine kinase inhibitor with or without aggressive upfront local radiation therapy in patients with EGFRm oligometastatic non-small-cell lung cancer: interim results of a randomized phase III, open-label clinical trial (SINDAS) (NCT02893332). Int J Radiat Oncol Biol Phys 108 (Suppl): E81. 10.1016/j.ijrobp.2020.07.1169 [DOI] [Google Scholar]
  58. Zheng X, Schipper M, Kidwell K, Lin J, Reddy R, Ren Y, Chang A, Lv F, Orringer M, Spring Kong FM. 2014. Survival outcome after stereotactic body radiation therapy and surgery for stage I non-small cell lung cancer: a meta-analysis. Int J Radiat Oncol Biol Phys 90: 603–611. 10.1016/j.ijrobp.2014.05.055 [DOI] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES