| Home | E-Submission | Sitemap | Contact Us |  
top_img
Radiation Oncology Journal > Volume 41(3); 2023 > Article
Kim, Kim, and Kim: Exploring the past, present, and future of postoperative radiotherapy for N2 stage non-small cell lung cancer

Abstract

Despite conventionally applied postoperative radiotherapy (PORT) in pathological N2 (pN2) stage non-small cell lung cancer (NSCLC) considering high locoregional recurrence, its survival benefit has been a continuous topic of debate. Although several randomized clinical trials have been conducted, many of them have been withdrawn or analyzed without statistical significance due to slow accrual, making it difficult to determine the efficacy of PORT. Recently, the results of large-scale randomized clinical trials have been published, which showed some improvement in disease-free survival with PORT, but finally had no impact on overall survival. Based on these results, it was expected that the debate over PORT in pN2 patients with NSCLC would come to an end. However, since pN2 patients have different clinicopathologic features, it has become more important to carefully select the patient population who will benefit from PORT. In addition, given the development of systemic treatments such as molecular-targeted therapy and immunotherapy, it is crucial to evaluate whether there is any benefit to PORT in the midst of these recent changes. Therefore, determining the optimal treatment approach for NSCLC pN2 patients remains a complex issue that requires further research and evaluation.

Introduction

Cancer is the first leading cause of death in Korea and lung cancer accounts for 22% of cancer deaths with approximately 30,000 new cases diagnosed in 2019 [1]. In terms of histological type, 80%-90% of lung cancers are classified as non-small cell lung cancer (NSCLC) [2]. Stage I NSCLC has recently increased, but more advanced stages are still 70%, which need multimodal therapy [2].
Among NSCLC patients with positive nodes, especially pathological N2 (pN2) disease, 35%–55% experience treatment failures even after complete resection followed by adjuvant chemotherapy and are associated with inferior overall survival (OS) [3,4]. According to the American Joint Committee on Cancer 8th edition, N2 is defined when metastasis is found in ipsilateral mediastinal and/or subcarinal lymph nodes [5]. With regard to the location and number of involved lymph nodes, N2 consists of a highly heterogeneous group. Besides, stage III pN2 possesses a wide variety of clinicopathologic features such as the size and characteristics of the primary tumor, making it more difficult to select patients who will benefit from postoperative treatments.
There has been a long debate regarding the appropriate use of postoperative radiotherapy (PORT) for patients with completely resected NSCLC, although PORT is often employed for patients with pN2 NSCLC. Especially, the Lung ART trial presented in 2020 seemed to put an end to the controversy over PORT: no more PORT for stage IIIA pN2 after complete resection [6]. However, in 2021, the updated results of Lung ART reported that PORT significantly improved mediastinal relapse-free survival, despite no difference in metastasis-free survival [7]. Therefore, the authors concluded that a personalized prescription of PORT might be allowed based on prognostic factors.
In this review, we will not discuss PORT after incomplete resection (R1 or R2), which is a well-established PORT indication, and briefly summarize previous studies for PORT after R0 resection. We then highlight the major breakthroughs derived from recent randomized clinical trials (RCTs), updating current lung cancer treatment guidelines. Ultimately, we want to give answers about how to select and manage patients with completely resected pN2 NSCLC patients.

Historical Perspective on PORT for N2 Stage NSCLC

Given the assumption that high local recurrence in pN2 patients than pN0-1 was thought to be due to the challenges of achieving complete surgical removal of microscopic lymph node metastasis in the mediastinum, a retrospective analysis was conducted at Mayo Clinic on a cohort of 224 patients with pN2 status between 1987 and 1993 to evaluate the potential benefit of PORT in these patients [8]. The 4-year freedom from local recurrence (83% vs. 40%, p < 0.0001) and survival rate (43% vs. 22%, p = 0.0005) were higher in the PORT group. This study published in 1997 was the largest evaluating PORT in pN2 patients in that era, suggesting that PORT can enhance both local control and OS. Large-scale RCTs have been initiated since that.
The landmark PORT meta-analysis in 1998, which initially included nine old RCTs (PORT vs. observation), showed unfavorable survival outcomes after PORT—OS, hazard ratio (HR) = 1.21, p = 0.001; disease-free survival (DFS), HR = 1.13, p=0.007; locoregional relapse-free survival (LRRFS), HR = 1.16, p = 0.005 [9]. In subgroup analysis, PORT was detrimental with stages I–II, whereas, for stage III, there was no clear evidence of a detriment. After the publication of this meta-analysis, there was a sustained decline in PORT use. Consequently, few papers were published in early 2000. However, considering patients were recruited in the mid to late 1900s, this meta-analysis had several criticized points, despite the importance of this analysis. Participants might have been evaluated with inadequate staging and they received no longer standard treatment including old radiotherapy (RT) techniques such as cobalt-60 equipment, large fraction size, and large treated volume with two-dimensional RT planning. In some trials included in the meta-analysis, the total dose was quite lower than the currently used, and adjuvant chemotherapy was not done.
Meanwhile, two studies published in 2006 made clinicians reconsider PORT for N2 NSCLC. The first one, the Surveillance, Epidemiology, and End Results (SEER) database analysis using 7,465 patients with resected NSCLC between 1998 and 2002 showed that although PORT was associated with a significant decrease in survival for patients with N0 and N1 disease, PORT was associated with more prolonged cancer-specific (5-year: 36% vs. 27%, p = 0.0298; HR = 0.850, p = 0.0133) and overall survival (5-year: 27% vs. 20%, p = 0.0036; HR = 0.855, p = 0.0077) for patients with N2 disease [10]. In addition, there was a post-hoc analysis of the Adjuvant Navelbine International Trialist Association (ANITA) trial, which was designed to compare the effect of adjuvant vinorelbine plus cisplatin with observation in completely resected NSCLC [11]. As PORT was not mandatory in this study, a non-randomized sub-analysis was conducted to compare OS in patients who did or did not receive PORT. The results showed that PORT led to longer OS, both in the chemotherapy arm (5-year: 47% vs. 34%) and in the observation arm in N2 disease (5-year: 21% vs. 17%).
Subsequent several retrospective studies have been published and they also suggested that a significant increase in DFS and 5%–15% of overall survival benefit was observed with PORT use in unselected N2 [1214]. China multicenter retrospective study showed that the 5-year OS was 30.5% in the postoperative chemoradiotherapy group and 22.2% in the postoperative chemotherapy group (p = 0.007) [12]. For 5-year DFS, the postoperative chemoradiotherapy and postoperative chemotherapy group had a rate of 14.4% and 9.3%, respectively (p = 0.003). Another single institutional retrospective study showed that PORT had a significantly longer OS time (p = 0.046), DFS interval (p = 0.009), as well as significantly higher LRRFS (p = 0.025), distant metastasis-free survival rate (p = 0.001) [13]. Based on this, they conducted a PORT-C trial which will be discussed in a later section. The largest retrospective one was the US National Cancer Data Base (NCDB) study published in 2015. It included 4,483 patients with pN2 NSCLC who underwent complete resection and adjuvant chemotherapy from 2006 to 2010. On multivariable analysis, it demonstrated a similar gain of additional PORT (HR = 0.886) compared to the previous results of the SEER database: use of PORT was associated with an increase in median and 5-year OS compared with no PORT (median: 45.2 vs. 40.7 months, 5-year: 39.3% vs. 34.8%; p = 0.014) [14].
The steady advancement of RT delivery methods has raised expectations that modern PORT will contribute to improved survival. In the updated meta-analysis of 11 trials, a total of 2,387 patients were included for survival analysis, and previous RCTs were stratified according to the use or non-use of linear accelerators (LINAC) [15]. In terms of local tumor failure, local recurrence significantly decreased with PORT, and the most significant decrease was observed in the group treated with LINAC only, with a relative risk (RR) of 0.31. With regard to OS, for the whole group, there was no improvement with PORT use. However, an increase in OS was observed in the LINAC-only group, with a RR of 0.76. PORT with LINACs was estimated to reduce local recurrence rates from 30% to 10% and PORT was estimated to increase the absolute 5-year OS by 13% (approximately 20% × 2/3 by generating a hypothesis model). These results reinforced further investigations of PORT using modern LINAC.
Lastly, the Cochran database review was published in 2016, updating the above-mentioned meta-analysis in 1998 with two trials conducted in Italy and Korea [16]. In this analysis, patients with T3N0M0 were reclassified from stage IIIA to stage IIB according to the TNM system change, and treatment bias was evaluated using the Fisher test. The results of 2,343 patients from 11 trials demonstrated that PORT showed a detrimental effect on patients with completely resected NSCLC. Besides, in contrast to the above meta-analysis, LINAC only did not affect OS (HR = 1.02, 95% confidence interval [CI] = 0.80-1.31, p = 0.85).
The aforementioned studies are summarized in Table 1.

Recent Results of Representative RCTs on PORT for N2 Stage NSCLC

Several RCTs have been conducted on patients recruited in the recent 2000s, during which the advancement of imaging technology and treatment methods enabled more precise cancer staging, as well as, more sophisticated surgery and RT. In the following section, we will discuss four RCTs with a particular focus on the PORT-C and Lung ART trials, which were published in 2021 and 2022, respectively (Table 2).
The first RCT using modern PORT was conducted in China to compare adjuvant chemotherapy with adjuvant concurrent chemoradiotherapy (CCRT) in pN2 NSCLC [17]. Although this study has a small number of patients due to slow accrual, three-dimensional conformal radiotherapy (3D-CRT) with optimal RT dose of 50.4 Gy in 28 fractions and modern chemotherapy regimen—paclitaxel (175 mg/m2) and cisplatin (60 mg/m2)—was applied. As a result, the adjuvant CCRT group showed decreased local (18/66 vs. 34/69, p = 0.01) and distant failures (32/66 vs. 45/69, p = 0.05) and improved DFS rate (5-year: 30.3% vs. 18.8%, p = 0.04) of these patients, compared with adjuvant chemotherapy. Despite the marginal significance of the difference, a superior OS was observed in the adjuvant CCRT group (5-year: 37.9% vs. 27.5%, p = 0.07), and in subgroup analysis, adjuvant CCRT increased the OS rate of patients with multiple N2 (p = 0.02).
A similar randomized phase II trial for only unsuspected or minimal N2 NSCLC was conducted in South Korea, reporting no significant differences in DFS and OS between adjuvant CCRT and chemotherapy alone [18]. The exploratory analysis of subgroups demonstrated that OS might be improved when patients with non-adenocarcinoma (HR = 0.359, 95% CI = 0.093–1.390) or with fewer than 15 lymph nodes dissected (HR = 0.575, 95% CI = 0.135–2.446) were treated with adjuvant CCRT. A cautious interpretation of this result needs because of a very small number of patients. Compared to the study by Shen et al. [17], the Korean study showed a higher OS, and the authors attributed this to the thorough staging workup and invasive mediastinal staging procedures. This difference in study populations might reduce the role of adjuvant RT in the Korean trial. Furthermore, there was a contrast between the two groups that multistation pN2 patients favored chemotherapy alone in the Korean trial and CCRT in the Chinese study. Distant metastasis frequently occurred in multistation pN2 disease and the insufficient dose of chemotherapy in CCRT did not control distant metastasis properly, leading to decreased OS. Further validation is required. The number of patients experiencing locoregional failure only was 5 (9.8%) in CCRT and 7 (14.0%) in the chemotherapy alone.
The phase III PORT-C trial enrolled patients with completely resected N2 NSCLC and randomized 1:1 to PORT 50 Gy and observation after four cycles of adjuvant platinum-based chemotherapy [19]. Intensity-modulated radiotherapy (IMRT) was applied in 89% of patients. The primary endpoint was DFS. The clinical features were comparable between the two groups: 80% of patients had adenocarcinoma, and nearly 60% were not cN2 which underlies the importance of pretreatment mediastinal staging. The 3-year DFS rates in PORT and observation were 40.5% versus 32.7%, respectively, in the modified intent-to-treat analysis (HR = 0.84, 95% CI = 0.65–1.09, p = 0.20). In the per-protocol analysis, PORT significantly improved DFS (42.8% vs. 30.6%; HR = 0.75, 95% CI = 0.57–1.00, p = 0.05) but not OS (82.6% vs. 83.1%; HR = 0.83, 95% CI = 0.53–1.30, p = 0.41). The local recurrence-only was less observed in the PORT group compared to the non-PORT (3-year: 9.5% vs. 18.3%; Fine-Gray HR = 0.55, 95% CI = 0.31–0.97, Gray test p = 0.04). The authors described that no RT-related mortality was observed, and most deaths occurred due to cancer progression. The low toxicity rate can be attributed to the use of IMRT and the implementation of stricter dose constraints for organs at risk. We need to take a closer look at which 44 of 184 patients (23.9%) in the PORT arm refused PORT, and 10 of 180 patients (5.6%) in the observation arm actually received PORT. Besides, although all patients completed four cycles of postoperative chemotherapy, 61.9% of patients still experienced distant metastasis. This may have offset the benefits of locoregional control from PORT, resulting in no improvement in DFS and OS.
Another phase III trial, the Lung ART trial, also randomized 1:1 to PORT of 54 Gy (3D-CRT of 89% and IMRT of 11%) and observation [20]. This trial did not meet the primary accrual goal due to slow enrollment. Baseline patient characteristics were comparable between the two groups. Major tumor histology was adenocarcinoma over 70%. Although over 90% of patients were staged with positron emission tomography-computed tomography scan, of enrolled cases, 40% had microscopic, unforeseen N2. And about 34% of the patients had single N2 station involvement. Adjuvant chemotherapy was not mandatory at that time and unlike the previous three studies, neoadjuvant chemotherapy was allowed. Nevertheless, the majority of patients (96%) were treated with chemotherapy (preoperative, postoperative, or both). In primary endpoint analysis, non-significant improvement of DFS was shown: 3-year DFS rate of 43.8% in control, higher than expected, and 47.1% with PORT; median DFS of 8 months increased by PORT (30.5 vs. 22.8 months; HR = 0.86, 95% CI = 0.68–1.08, p = 0.18). PORT did not increase OS. The control arm was much more likely to suffer mediastinal relapse (46.1% vs. 25%), which indicated approximately 50% risk reduction of locoregional failure by PORT, but intercurrent death was more common in the PORT arm (14.6% vs. 5.3%). Notably, 11 patients among 21 deaths in the PORT group died with cardiopulmonary toxicity and these may result from the most common application of 3D-CRT (89%). But the mean dose of heart was not different between patients with 3D-CRT and IMRT—median with interquartile range, 13.5 Gy (8–19) vs. 13.7 Gy (6–18), requiring more detailed analysis. Multivariable analysis identified the significant prognostic factors for DFS as follows: gender, histology, N1 involvement, number of involved mediastinal nodes, and quality of resection.
Overall, up-to-date PORT trials showed that it can reduce locoregional recurrence approximately by half after the standard treatment approach, but this locoregional control benefit may not be translated into DFS or OS benefit because of potential cardiopulmonary toxicity by 3D-CRT or unsatisfactory systemic control with platinum-doublet chemotherapy agents. Given that a subgroup analysis has demonstrated the benefits of PORT in certain patient populations, further research is necessary to define and characterize these patients.

Current Guidelines and Recommendations on PORT for N2 Stage NSCLC

As discussed in the previous parts, robust evidence to support the use of PORT in the treatment of N2 stage NSCLC is still lacking. Currently, most international guidelines mention the use of PORT as follows: in the case of R1 or R2 resection, there is generally no big difference in the use of PORT, but for pN2 disease after R0 resection, the recommendation level is not high or it is suggested to apply selectively [2124] (Table 3).
In the National Comprehensive Cancer Network guidelines, postoperative chemotherapy can be administered followed by PORT or concurrently depending on the margin status [21]. PORT is recommended with a category 2A for N2 after R0 resection. Although the American Society for Radiation Oncology (ASTRO) does not publish very up-to-date official guidelines after the publication of recent RCTs, in the previous guidelines they described that PORT addition after R0 resection in N2 disease showed no definitive proof of a positive or negative impact on OS but showed better local control than observation strategies [22].
However, the guidelines of the medical society including the European Society for Medical Oncology (ESMO) and the American Society of Clinical Oncology (ASCO) more conservatively state that PORT use in R0 resection can be considered “only in selected cases” following individual risk assessment [23,24]. The ESMO recommends no routine use of PORT in single N2 status and only permits PORT after careful evaluation of locoregional recurrence risks [23]. In multistation N2, definitive CCRT is preferred but multidisciplinary approaches are required. According to the ASCO guidelines published in 2017, postoperative multimodal assessment is recommended to determine the advantages and disadvantages of receiving PORT [24]. Recently updated ASCO guideline mentions that PORT should not be routinely offered for patients receiving platinum-based chemotherapy either before or after surgery and without extracapsular extension (ECE) [25]. But, the strength of its recommendation is weak. ASTRO has released an updated article agreeing with this ASCO guideline [26].
Conclusively, in the current status, PORT is not recommended routinely for all patients with completely resected pN2 NSCLC. The decision on whether to use PORT should be made on an individualized basis, taking into account the risk factors presented in the following section.

Factors Affecting the Choice of PORT for N2 Stage NSCLC: Implications for Patient Selection

There is still no robust answer in PORT patient selection, and various data exist for each study. Here are some of the most important factors to consider:

1. Multistation/extensive mediastinal involvement after mediastinal dissection

In the Lung ART trial, multiple mediastinal node stations were involved in about 30% of patients [20]. DFS was affected by ≥2 stations involved, compared with a single station (HR = 1.46, 95% CI = 1.1–1.9, p = 0.01). A previous study suggested a potential DFS benefit of PORT in patients with multiple station mediastinal lymph node metastases (5-year: 43.2% vs. 16.6%, p = 0.037) [27]. It also demonstrated in another retrospective study that PORT improved DFS in patients with multiple N2 stations metastases compared with single N2 station metastasis (5-year: 41.7% vs 5.9%, p = 0.0220) [28]. In regard to OS, patients with multiple pN2 favored postoperative CCRT over chemotherapy alone [17]. On the contrary, some investigators also claimed that single N2 station involvement was a predictor of benefit from PORT [29]. Although the number of patients was too small, the Korean trial reported that chemotherapy alone (n=8) was more effective than CCRT (n = 5) for patients with N2 multistation (OS: HR = 5.572, 95% CI = 1.01–30.754) [18].

2. The number of metastatic lymph nodes or lymph node ratio

In the SEER data analysis including 3,373 patients from 2004 to 2013, multivariable analysis showed that the number of positive lymph nodes (≤3) was independently associated with better OS and lung cancer-specific survival, then the use of PORT demonstrated better OS compared to no-PORT for patients with positive lymph nodes (>3), but not for patients with less number of positive lymph nodes (≤3) [30]. In terms of DFS, it also showed consistent results in the PORT-C (HR = 0.75, 95% CI = 0.58–0.98, p = 0.04) [19]. Other studies using the SEER data or NCDB showed that PORT can be indicated in patients with a specific range of lymph node-positive ratios [3134]. The proposed cutoff for lymph node ratio varied among studies, with some suggesting a threshold of >15% [34], ≥30% [33], ≥50% [31], or 60%–80% [32].

3. ECE

Importantly, ECE has reflected the aggressive biological behavior in several types of cancers, including head and neck, breast, and colorectal cancers [3537]. In patients with completely resected stage IIA–IIIA NSCLC, ECE also represents a powerful prognostic factor [38]. PORT led to a significant improvement in OS among negative ECE patients (HR = 0.518, 95% CI = 0.276–0.971, p = 0.04) but did not have a similar effect on positive ECE patients [39]. Although recently conducted RCTs did not evaluate the association between ECE and PORT [1720], the status of ECE can potentially aid in selecting pN2 patients who will benefit from PORT. On the other hand, some argue that in cases where there is ECE or lymph node capsular rupture, the resection should be considered incomplete [40], and this may necessitate the use of PORT.

4. Histology (squamous cell carcinoma vs. adenocarcinoma)

There is a discrepancy regarding the NSCLC histology that shows the benefits of PORT. Several studies proposed that PORT may confer a greater advantage in treatment outcomes for squamous cell carcinoma [27,41]. They reported increases in the 5-year OS (from 37.1% to 63.2%, p = 0.026) and DFS (from 23.3% to 70.1%, p = 0.011) by PORT. But others found that the presence of the papillary predominant adenocarcinoma subtype was a reliable indicator of the potential benefit from PORT [29].

5. ypN2 in case of receiving neoadjuvant chemotherapy

Neoadjuvant chemotherapy is an alternative treatment option for resectable NSCLC [21]. Patients with ypN2 status may have a greater extent of regional tumor involvement and increased chemotherapy resistance when compared to those with pN2 status. Thus, the use of PORT can potentially enhance locoregional management and ultimately improve overall survival [42]. Large-scale retrospective studies using SEER or NCDB suggested that PORT benefited patients with persistent N2 disease after neoadjuvant chemotherapy [34,42,43]. In the Lung ART trial, subgroup analysis revealed that PORT significantly improved DFS in ypN2 patients (3-year: 49.4% vs. 20.8%; HR = 0.52, 95% CI = 0.28–0.98) [20].
Several risk factors above can predict the benefit of PORT and can be considered when selecting patients. Several nomograms or scoring systems have been developed to help physicians screen and counsel patients with resected N2 [4446]. As a simplified model of them, one article insisted that patients who meet three or more of the following criteria are strongly advised to undergo PORT: smoking index (number of cigarettes smoked per day × number of cigarette-years) ≤400, cN2, pT3, squamous cell carcinoma, and ≥4 positive nodes [44].
Although definite high-risk factors are still not consistently identified, PORT should not be completely disregarded in these patients as a potentially useful treatment option. Rather, it can be summarized that steady research efforts are needed in consideration of the future perspectives introduced in the next section.

Ongoing Trials, Future Directions, and Conclusions

As summarized above, it is thought that more clinical evidence is still needed for the application of the PORT for pN2 patients, but the protracted length of adjuvant trials for the resectable stage of NSCLC has resulted in slow progress and high expenses. It is highly likely that the high-level evidence for PORT we currently had will not be updated for years. Japan Clinical Oncology Group 1916 (J-PORT study, UMIN000042905) is in progress using a scheme similar to the PORT-C trial, but we still have to wait several years to get another prospective randomized evidence [47].
From a different perspective, the clinical trial using stereotactic body radiotherapy (SBRT) in the adjuvant setting was launched in 2019 for patients with close/involved surgical margins or pN2 (NCT04073745). Considering SBRT has an extremely short treatment duration and fewer side effects compared with conventional RT, the positive results of this trial would broaden the PORT indications in NSCLC.
Another big change to consider is the development and change of systemic chemotherapy for resectable stage NSCLC. More recently, the promising progress in the treatment of metastatic NSCLC has increased interest in using immune checkpoint blockades or targeted agents at resectable stages. IMpower010 made adjuvant atezolizumab maintenance one of the options after routine adjuvant chemotherapy [48]. Of course, the usual PORT that can be considered subsequently after adjuvant chemotherapy was not allowed under the IMpower010 research protocol. However, a considerable portion of enrolled patients subsequently required post-relapse mediastinal RT. These results suggest the possibility that there is still a role for PORT to further increase the therapeutic outcome.
In contrast, PORT may not be beneficial in patients with actionable mutations. The ADAURA trial found that the use of osimertinib after surgery significantly improved DFS in patients with epidermal growth factor receptor (EGFR) mutation-positive completely resected stage IB to III NSCLC (compared with placebo), therefore the additional benefit of PORT may be limited considering the high efficacy of osimertinib in EGFR mutation-positive stage III NSCLC [49].
Another important recent challenge to applying PORT is the increased use of the neoadjuvant approach led by the CheckMate 816 regimen [50]. Since many neoadjuvant trials did not include PORT in the protocol, there is very little evidence on how to apply PORT to patients who received these treatments. Hopefully, adopting perioperative immune checkpoint blockades can improve distant metastasis control and may re-consider the role of robust locoregional control by PORT.
Along with these, other ongoing trials of novel systemic therapy with targeted agents or immunotherapy may also change PORT indications. The contribution of improved locoregional control to survival depends on the effectiveness of systemic treatment. If better systemic control were achieved and patterns of failure were changed, the role of PORT may be revisited. So far, in surgically resected NSCLC, the efficacy of systemic treatment varies from patient to patient due to various factors, and it is not possible to accurately determine whether PORT can improve survival under certain levels of systemic control. Further evidence is needed in the midst of these recent changes and adequately powered trials to establish clinically meaningful benefits are awaited.
In conclusion, PORT for completely resected pN2 NSCLC has been an area of ongoing debate. The first choice for pN2 NSCLC patients after complete resection had been PORT since the late 1990s; however, the Lung ART and PORT-C trials have challenged this notion. Despite demonstrating a noteworthy decrease in the locoregional recurrence rate, PORT has yet to produce any survival advantage. Nevertheless, the potential benefits of PORT for patients with high-risk features remain uncertain. Nevertheless, we cautiously recommend the use of PORT for N2 stage NSCLC patients with high-risk features, such as high and/or persistent mediastinal tumor burdens. Future research should focus on identifying optimal candidates for PORT and determining its effectiveness in combination with novel systemic therapies. Ultimately, the goal is to establish clinically meaningful benefits for patients with completely resected pN2 NSCLC.

Notes

Statement of Ethics

As this study did not involve any human subjects, Institutional Review Board approval and informed consent were not required.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Funding

None.

Author Contributions

Conceptualization, Kim BH, Kim HJ. Supervision, Kim HJ. Writing of the original draft, Kim BH, Kim JS. Writing of the review and editing, Kim BH, Kim JS, Kim HJ.

Data Availability Statement

Not applicable.

Table 1.
Overview of historical studies investigating postoperative radiotherapy for non-small cell lung cancer
Trial Study design Study period n Stage Study arm DFS OS Subgroup
Sawyer et al. [8] Retrospective 1987–1993 224 pN2 Observation vs. PORT (30–66.4 Gy) 4-yr: 19% vs. 51% (p < 0.001) 4-yr: 22% vs. 43% (p < 0.001) OS: >1 involved pN2, PORT favored
PORT Meta-analysis Trialists Group [9] Meta-analysis 1966–1995 2128 I-III Observation vs. PORT (30–60 Gy) HR: 1 vs. 1.13 (p = 0.018) HR: 1 vs. 1.21 (p = 0.001) OS: Stage I, pN0, Observation favored
Lally et al. [10] Retrospective 1998–2002 7465 II-III Observation vs. PORT - 3-yr: 47% vs. 41% (p < 0.001) DFS & OS: pN2, PORT favored
DFS 5-yr: 27% vs. 36% (p = 0.030)
OS 5-yr: 20% vs. 27% (p = 0.004)
Douillard et al. [11] Post-hoc analysis 1994–2000 840 IB-IIIA (Adjuvant chemotherapy was randomized) - POCT group (5-yr: 46% vs. 45%) OS: pN2, PORT favored
Observation vs. PORT (45–60 Gy) Observation group (5-yr: 27% vs. 32%) POCT group (5-yr: 34% vs. 47%)
Observation group (5-yr: 17% vs. 21%)
Zou et al. [12] Retrospective 1998–2005 183 III-pN2 POCT vs. POCRT (48–54 Gy) 5-yr: 9.3% vs. 14.4% (p = 0.003) 5-yr: 22.2% vs. 30.5% (p = 0.007) -
Dai et al. [13] Retrospective 2003–2005 221 IIIA-pN2 (Adjuvant chemotherapy not mandatory) 5-yr: 16.5% vs. 32.1% (p = 0.009) Median: 31.8 vs. 43.9 mo -
Observation vs. PORT (60 Gy) 5-yr: 30.6% vs. 36.6% (p = 0.046)
Robinson et al. [14] Retrospective 2006–2010 4483 IIIA-pN2 (After adjuvant chemotherapy) - Median: 40.7 vs. 45.2 mo -
Observation vs. PORT (45–82.8 Gy) 5-yr: 34.8% vs. 39.3% (p = 0.014)

DFS, disease-free survival; OS, overall survival; pN2, pathological N2; PORT, postoperative radiotherapy; HR, hazard ratio; POCT, postoperative chemotherapy; POCRT, postoperative concurrent chemoradiotherapy.

Table 2.
Summary characteristics of recent randomized clinical trials for postoperative radiotherapy in completely resected N2 stage non-small cell lung cancer
Trial Study period n Stage Randomization DFS OS Subgroup
Shen et al. [17] 2004–2009 135 IIIA-pN2 POCT vs. POCRT (50.4 Gy/28 Fx) Median: 18 vs. 28 mo Median: 28 vs. 40 mo OS: Multiple pN2, POCRT favored
5-yr: 18.8% vs. 30.3% (p = 0.04) 5-yr: 27.5% vs. 37.9% (p = 0.07)
Sun et al. [18] 2009–2014 101 IIIA-pN2 POCT vs. POCRT (50 Gy/25 Fx) Median: 21.9 vs. 24.7 mo (p = 0.40) Median: 83.5 vs. 74.3 mo (p = 0.38) OS: Never smoker, multiple pN2, POCT favored
PORT-C [19] 2009–2017 364 IIIA-pN2 (After adjuvant chemotherapy) mITT mITT DFS: Lymph nodes ≥4, PORT favored
Observation vs. PORT (50 Gy/25 Fx)  Median: 18.6 vs. 22.1 mo  Median: 81.5 mo vs. not reached
 3-yr: 32.7% vs. 40.5% (p = 0.20)  3-yr: 82.8% vs. 78.3% (p = 0.93)
PP PP
 3-yr: 30.6% vs. 42.8% (p = 0.05)  3-yr: 83.1% vs. 82.6% (p = 0.41)
Lung ART [20] 2007–2018 501 IIIA-pN2 (Adjuvant chemotherapy not mandatory) Median: 22.8 vs. 30.5 mo 3-yr: 69% vs. 67% (NS) DFS: Preoperative chemotherapy alone, PORT favored
Observation vs. PORT (54 Gy/27 Fx) 3-yr: 44% vs. 47% (p = 0.18)

DFS, disease-free survival; OS, overall survival; pN2, pathological N2; POCT, postoperative chemotherapy; POCRT, postoperative concurrent chemoradiotherapy; Fx, fraction; pN2, pathological N2; PORT, postoperative radiotherapy; mITT, modified intent-to-treat; PP, per-protocol; NS, not significant.

Table 3.
The latest recommendation in various guidelines for postoperative radiotherapy in completely resected N2 stage non-small cell lung cancera)
Year Guideline Recommendation levelb) Description
2015 ASTRO Strong May improve local control
2017 ESMO C (optional) May be an option following individual risk assessment
2017 ASCO Moderate Recommend a postoperative multimodality evaluation
2022 ASCO Weak Not routine use for patients without ECE who received platinum-based chemotherapy
2023 NCCN Category 2Ac) Consider RT

ASTRO, American Society for Radiation Oncology; ESMO, European Society for Medical Oncology; ASCO, American Society of Clinical Oncology; ECE, extracapsular extension; NCCN, National Comprehensive Cancer Network; RT, radiotherapy.

a) At present, there are no official recommendations from the European Society for Radiotherapy and Oncology.

b) This indicates the level of recommendation in the original guideline.

c) Category 1: chemotherapy followed by atezolizumab or pembrolizumab or osimertinib.

References

1. Kang MJ, Won YJ, Lee JJ, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2019. Cancer Res Treat 2022;54:330–44.
crossref pmid pmc pdf
2. Lee JG, Kim HC, Choi CM. Recent trends of lung cancer in Korea. Tuberc Respir Dis (Seoul) 2021;84:89–95.
crossref pmid pmc pdf
3. Arriagada R, Bergman B, Dunant A, et al. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med 2004;350:351–60.
crossref pmid
4. Winton T, Livingston R, Johnson D, et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. N Engl J Med 2005;352:2589–97.
crossref pmid
5. Amin MB, Edge SB, Greene FL, et al. AJCC cancer staging manual. 8th ed. New York, NY: Springer International Publishing; 2017.

6. Le Pechoux C, Pourel N, Barlesi F, et al. 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 2020;31(suppl_4):S1178.
crossref
7. Le Pechoux C, Barlesi F, Pourel N, et al. 1170O 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: characterisation of PORT efficacy in lung ART (IFCT-0503, UK NCRI, SAKK). Ann Oncol 2021;32(suppl_5):S939.
crossref
8. Sawyer TE, Bonner JA, Gould PM, et al. The impact of surgical adjuvant thoracic radiation therapy for patients with nonsmall cell lung carcinoma with ipsilateral mediastinal lymph node involvement. Cancer 1997;80:1399–408.
crossref pmid
9. PORT Meta-analysis Trialists Group. Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. Lancet 1998;352:257–63.
crossref pmid
10. Lally BE, Zelterman D, Colasanto JM, Haffty BG, Detterbeck FC, Wilson LD. Postoperative radiotherapy for stage II or III non-small-cell lung cancer using the surveillance, epidemiology, and end results database. J Clin Oncol 2006;24:2998–3006.
crossref pmid
11. Douillard JY, Rosell R, De Lena M, et al. Adjuvant vinorelbine plus cisplatin versus observation in patients with completely resected stage IB-IIIA non-small-cell lung cancer (Adjuvant Navelbine International Trialist Association [ANITA]): a randomised controlled trial. Lancet Oncol 2006;7:719–27.
crossref pmid
12. Zou B, Xu Y, Li T, et al. A multicenter retrospective analysis of survival outcome following postoperative chemoradiotherapy in non-small-cell lung cancer patients with N2 nodal disease. Int J Radiat Oncol Biol Phys 2010;77:321–8.
crossref pmid
13. Dai H, Hui Z, Ji W, et al. Postoperative radiotherapy for resected pathological stage IIIA-N2 non-small cell lung cancer: a retrospective study of 221 cases from a single institution. Oncologist 2011;16:641–50.
crossref pmid pmc pdf
14. Robinson CG, Patel AP, Bradley JD, et al. 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 2015;33:870–6.
crossref pmid pmc
15. Billiet C, Decaluwe H, Peeters S, et al. Modern post-operative radiotherapy for stage III non-small cell lung cancer may improve local control and survival: a meta-analysis. Radiother Oncol 2014;110:3–8.
crossref pmid
16. Burdett S, Rydzewska L, Tierney J, et al. Postoperative radiotherapy for non-small cell lung cancer. Cochrane Database Syst Rev 2016;10:CD002142.
crossref pmid pmc
17. Shen WY, Ji J, Zuo YS, et al. Comparison of efficacy for postoperative chemotherapy and concurrent radiochemotherapy in patients with IIIA-pN2 non-small cell lung cancer: an early closed randomized controlled trial. Radiother Oncol 2014;110:120–5.
crossref pmid
18. Sun JM, Noh JM, Oh D, et al. Randomized phase II trial comparing chemoradiotherapy with chemotherapy for completely resected unsuspected N2-positive non-small cell lung cancer. J Thorac Oncol 2017;12:1806–13.
crossref pmid
19. Hui Z, Men Y, Hu C, et al. Effect of postoperative radiotherapy for patients with pIIIA-N2 non-small cell lung cancer after complete resection and adjuvant chemotherapy: the phase 3 PORT-C randomized clinical trial. JAMA Oncol 2021;7:1178–85.
crossref pmid pmc
20. Le Pechoux C, Pourel N, Barlesi F, et al. Postoperative radiotherapy versus no postoperative radiotherapy in patients with completely resected non-small-cell lung cancer and proven mediastinal N2 involvement (Lung ART): an open-label, randomised, phase 3 trial. Lancet Oncol 2022;23:104–14.
crossref pmid
21. National Comprehensive Cancer Network. Non-small cell lung cancer version 3.2023 [Internet]. Plymouth Meeting, PA: National Comprehensive Cancer Network; 2023 [cited 2023 Jun 12]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf.

22. Rodrigues G, Choy H, Bradley J, et al. 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 2015;5:149–55.
crossref pmid
23. Postmus PE, Kerr KM, Oudkerk M, et al. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017;28(suppl_4):iv1–21.
crossref
24. Kris MG, Gaspar LE, Chaft JE, et al. Adjuvant systemic therapy and adjuvant radiation therapy for stage I to IIIA completely resected non-small-cell lung cancers: American Society of Clinical Oncology/Cancer Care Ontario Clinical Practice Guideline update. J Clin Oncol 2017;35:2960–74.
crossref pmid
25. Daly ME, Singh N, Ismaila N, et al. Management of stage III non-small-cell lung cancer: ASCO guideline. J Clin Oncol 2022;40:1356–84.
crossref pmid
26. Simone CB 2nd, Bradley J, Chen AB, et al. ASTRO radiation therapy summary of the ASCO guideline on management of stage III non-small cell lung cancer. Pract Radiat Oncol 2023;13:195–202.
crossref pmid
27. Kim BH, Kim HJ, Wu HG, et al. Role of postoperative radiotherapy after curative resection and adjuvant chemotherapy for patients with pathological stage N2 non-small-cell lung cancer: a propensity score matching analysis. Clin Lung Cancer 2014;15:356–64.
crossref pmid
28. Matsuguma H, Nakahara R, Ishikawa Y, et al. Postoperative radiotherapy for patients with completely resected pathological stage IIIA-N2 non-small cell lung cancer: focusing on an effect of the number of mediastinal lymph node stations involved. Interact Cardiovasc Thorac Surg 2008;7:573–7.
crossref pmid
29. Yuan C, Tao X, Zheng D, et al. The lymph node status and histologic subtypes influenced the effect of postoperative radiotherapy on patients with N2 positive IIIA non-small cell lung cancer. J Surg Oncol 2019;119:379–87.
crossref pmid pdf
30. Wang S, Ma Z, Yang X, et al. Choice of postoperative radiation for stage IIIA pathologic N2 non-small cell lung cancer: impact of metastatic lymph node number. Radiat Oncol 2017;12:207.
crossref pmid pmc pdf
31. Mankuzhy NP, Almahariq MF, Stevens CW, Quinn TJ. High lymph node positive to sampled ratio as a potential indication for postoperative radiation therapy in patients with pN2 non-small-cell lung cancer. J Cancer Immunol 2020;2:207–10.
crossref
32. Lee S, Noh OK. Optimal positive lymph node ratio showing the benefit of postoperative radiotherapy in pathologic N2 non-small cell lung cancer: an exploratory study using the Surveillance, Epidemiology, and End Results data. Radiat Oncol J 2022;40:37–44.
crossref pmid pmc pdf
33. Zhu F, Wang H, Ashamalla H. The significance of lymph node ratio and total lymph nodes examined in determining the indications of adjuvant radiation in pN2 non-small cell lung cancer. Clin Lung Cancer 2022;23:e384–93.
crossref pmid
34. Shinde A, Horne ZD, Li R, et al. Optimal adjuvant therapy in clinically N2 non-small cell lung cancer patients undergoing neoadjuvant chemotherapy and surgery: the importance of pathological response and lymph node ratio. Lung Cancer 2019;133:136–43.
crossref pmid
35. Kelder W, Ebrahimi A, Forest VI, Gao K, Murali R, Clark JR. Cutaneous head and neck squamous cell carcinoma with regional metastases: the prognostic importance of soft tissue metastases and extranodal spread. Ann Surg Oncol 2012;19:274–9.
crossref pmid
36. Kanyılmaz G, Fındık S, Yavuz BB, Aktan M. The significance of extent of extracapsular extension in patients with T1-2 and N1 breast cancer. Eur J Breast Health 2018;14:218–24.
crossref pmid pmc
37. Veronese N, Nottegar A, Pea A, et al. Prognostic impact and implications of extracapsular lymph node involvement in colorectal cancer: a systematic review with meta-analysis. Ann Oncol 2016;27:42–8.
crossref pmid
38. Liu W, Shao Y, Guan B, et al. Extracapsular extension is a powerful prognostic factor in stage IIA-IIIA non-small cell lung cancer patients with completely resection. Int J Clin Exp Pathol 2015;8:11268–77.
pmid pmc
39. Moretti L, Yu DS, Chen H, et al. Prognostic factors for resected non-small cell lung cancer with pN2 status: implications for use of postoperative radiotherapy. Oncologist 2009;14:1106–15.
crossref pmid pmc pdf
40. Rami-Porta R, Wittekind C, Goldstraw P; International Association for the Study of Lung Cancer (IASLC) Staging Committee. Complete resection in lung cancer surgery: proposed definition. Lung Cancer 2005;49:25–33.
crossref pmid
41. Tian C, Liu G, Xu Y, et al. Postoperative radiotherapy for patients with completely resected pathological stage IIIA-N2 non-small cell lung cancer: a preferential benefit for squamous cell carcinoma. Radiol Oncol 2020;55:66–76.
crossref pmid pmc
42. Bao Y, Yang X, Men Y, et al. Postoperative radiotherapy improves survival of patients with ypN2 non-small cell lung cancer after neoadjuvant chemotherapy followed by surgery: a propensity score matching study of the Surveillance, Epidemiology, and End Results database. Thorac Cancer 2022;13:404–11.
crossref pmid pmc
43. Komiya T, Takamori S, Wilding G. Adjuvant radiation therapy improves survival in stage IIIA (N2) non-small cell lung cancer with persistent N2 disease after neoadjuvant chemotherapy. Radiother Oncol 2022;176:234–8.
crossref pmid
44. Hui Z, Dai H, Liang J, et al. Selection of proper candidates with resected pathological stage IIIA-N2 non-small cell lung cancer for postoperative radiotherapy. Thorac Cancer 2015;6:346–53.
crossref pmid pmc
45. Zhang B, Yuan Z, Zhao L, Pang Q, Wang P. Nomograms for predicting progression and efficacy of post-operation radiotherapy in IIIA-pN2 non-small cell lung cancer patients. Oncotarget 2017;8:37208–16.
crossref pmid pmc
46. Zeng Y, Liu J, Wan M, et al. The association of postoperative radiotherapy with survival in resected N2 non-small cell lung cancer. J Thorac Dis 2023;15:42–53.
crossref pmid pmc
47. Shimoyama R, Nakagawa K, Ishikura S, et al. A multi-institutional randomized phase III trial comparing postoperative radiotherapy to observation after adjuvant chemotherapy in patients with pathological N2 Stage III non-small cell lung cancer: Japan Clinical Oncology Group Study JCOG1916 (J-PORT study). Jpn J Clin Oncol 2021;51:999–1003.
crossref pmid pdf
48. Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 2021;398:1344–57.
crossref pmid
49. Wu YL, Tsuboi M, He J, et al. Osimertinib in resected EGFR-mutated non-small-cell lung cancer. N Engl J Med 2020;383:1711–23.
crossref pmid
50. Forde PM, Spicer J, Lu S, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med 2022;386:1973–85.
crossref pmid pmc
Editorial Office
Department of Radiation Oncology, Samsung Medical Center,
Proton Therapy Center, B2, 81, Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
Tel : +82-2-3410-3617
E-mail: rojeditor@gmail.com, roj@kosro.or.kr
Copyright © The Korean Society for Radiation Oncology.                      Developed in M2PI
Close layer
prev next