Cancer Research and Treatment

Search

Close

Jang, Kim, Song, Shin, Lee, Ji, Choi, and Choi: Clinical Outcome of Stereotactic Body Radiotherapy in Patients with Early-Stage Lung Cancer with Ground-Glass Opacity Predominant Lesions: A Single Institution Experience

Clinical Outcome of Stereotactic Body Radiotherapy in Patients with Early-Stage Lung Cancer with Ground-Glass Opacity Predominant Lesions: A Single Institution Experience

Jeong Yun Jang1, Su Ssan Kim1, Si Yeol Song1, Young Seob Shin1, Sei Won Lee2, Wonjun Ji2, Chang-Min Choi2, Eun Kyung Choi1
Received December 26, 2022;       Accepted March 20, 2023;
ABSTRACT
Purpose
The detection rate of early-stage lung cancer with ground-glass opacity (GGO) has increased, and stereotactic body radiotherapy (SBRT) has been suggested as an alternative to surgery in inoperable patients. However, reports on treatment results are limited. Therefore, we performed a retrospective study to investigate the clinical outcome after SBRT in patients with early-stage lung cancer with GGO-predominant tumor lesions at a single institution.
Materials and Methods
This study included 89 patients with 99 lesions who were treated with SBRT for lung cancer with GGO-predominant lesions that had a consolidation-to-tumor ratio of ≤ 0.5 at Asan Medical Center between July 2016 and July 2021. A median total dose of 56.0 Gy (range, 48.0 to 60.0) was delivered using 10.0–15.0 Gy per fraction.
Results
The overall follow-up period for the study was median 33.0 months (range, 9.9 to 65.9 months). There was 100% local control with no recurrences in any of the 99 treated lesions. Three patients had regional recurrences outside of the radiation field, and three had distant metastasis. The 1-year, 3-year, and 5-year overall survival rates were 100.0%, 91.6%, and 82.8%, respectively. Univariate analysis revealed that advanced age and a low level of diffusing capacity of the lungs for carbon monoxide were significantly associated with overall survival. There were no patients with grade ≥ 3 toxicity.
Conclusion
SBRT is a safe and effective treatment for patients with GGO-predominant lung cancer lesions and is likely to be considered as an alternative to surgery.
Introduction
Introduction
Based on the National Lung Screening Trial data, the adoption of low-dose computed tomography (CT) as a screening tool for lung cancer has increased worldwide [1]. As a result, the detection of early-stage lung cancer with ground-glass opacity (GGO) has increased significantly, up to 63% [2]. The TALENT study also demonstrated the effectiveness of low-dose CT screening in a predefined high-risk group of non-smokers and reported a detection rate of T0 lung cancer of up to 2.6% [3].
Most ground-glass nodules (GGN) or subsolid nodules found on chest CT are transient or have benign findings such as inflammation, fibrosis, or hemorrhage, but their malignant potential increases if they persist or increase in size. To identify tumors and adopt treatment at the optimal time, several academic societies have recently suggested guidelines for GGN or subsolid nodules, but no established statement has been published yet [4,5]. Most guidelines suggest determining the timing of routine follow-up according to the nodule size and recommend surgery if the total size and consolidation increase above certain criteria, but the underlying level of evidence is weak. Therefore, in the real world, when determining surgical indications, the experience of the surgeon or institution is often relied upon.
Meanwhile, promising results of stereotactic body radiotherapy (SBRT) have been revealed through pooled analyses of the STARS and ROSEL trials and the revised STARS trial for patients with early-stage lung cancer [6,7]. As a result, SBRT is being used as an alternative option for patients with early-stage lung cancer who are difficult to treat surgically [8]. However, limited reports have been published with GGO-predominant lesions.
Therefore, we performed a single-institution retrospective study to investigate the clinical outcome after SBRT in patients with early-stage lung cancer with GGO-predominant lesions.
Materials and Methods
Materials and Methods
1. Patients
1. Patients
From July 2016 to July 2021, the medical records of patients who were pathologically or clinically diagnosed with lung cancer and who underwent SBRT at Asan Medical Center were investigated, and a total of 636 patients with 649 lesions were identified. We defined a GGO-predominant lesion as a lesion with a consolidation-to-tumor ratio (CTR) of ≤ 0.5 using the long-axis dimension of the tumor identified on the CT image, and 110 patients (120 lesions) met the criteria. Other major inclusion criteria were as follows: patients with (1) no regional lymph node (LN) involvement and no distant metastasis (DM); (2) no previous irradiation history to the ipsilateral side of the thorax; (3) no uncontrolled double primary tumor; and (4) sufficient follow-up images or medical records > 3 months after SBRT. Finally, 89 patients with 99 treated lesions were included in the study (Fig. 1). The clinical stage of the patients was evaluated on the basis of the eighth edition of the American Joint Committee on Cancer staging system.
2. Treatments
2. Treatments
Stereotactic body frames for linear accelerators (Elekta Oncology, Stockholm, Sweden) were used, and immobilization was achieved through vacuum-fitted frames. The simulation CT images of all patients were acquired four-dimensionally with a 2.5-mm slice thickness. The internal target volume was delineated in consideration of the movement of the primary tumor, and the planning target volume was set by expanding the median of 5 mm (range, 3 to 8 mm) from the internal target volume. Respiratory-gated radiotherapy (RT) was used with the Varian RPM respiratory gating system (Varian, Palo Alto, CA) and GE Lightspeed 4D CT (GE Healthcare, Waukesha, WI) scanner if the tumor movement was > 5 mm. In all cases, we planned to use either intensity-modulated RT or volumetric modulated arc therapy techniques, with a median total dose of 56.0 Gy (range, 48 to 60 Gy) delivered for 4 consecutive days or twice weekly for 2 weeks. Treatment verification was performed daily with cone-beam CT, fluoroscopy, or both.
3. Follow-up and outcomes
3. Follow-up and outcomes
Chest CT was performed 1 month after SBRT and thereafter every 3 months for 2 years and once every 6 months up to 5 years. If necessary, bronchoscopy, positron emission tomography–CT (PET-CT), and a bone scan were additionally performed, and a biopsy was also conducted for lesions suspected of recurrence. One radiation oncologist was responsible for evaluating tumor response with reference to official readings approved by thoracic specialist radiologists. The tumor response was evaluated on the basis of the Response Evaluation Criteria for Solid Tumors ver. 1.1 as follows: local control (LC) was defined as a response of stable disease or better, and a regional recurrence was defined as disease recurrence in the ipsilateral hemithorax or regional LN outside the radiation field. In addition, since there are limitations in evaluating CT images with Response Evaluation Criteria in Solid Tumor alone for response evaluation after SBRT to lung, the following high-risk features for recurrent disease were simultaneously considered: (1) infiltration into adjacent organs/structures, (2) sustained growth over serial scans, (3) bulging margins, (4) mass-like growth, (5) predominantly spherical growth, (6) craniocaudal growth, (7) air space obliteration/loss of air bronchograms [9]. DM was defined as a disease recurrence in the contralateral lung, pleura, or any distant organ, and DM-free survival (DMFS) was calculated from the termination of SBRT to the development of a distant recurrence or death. Disease-free survival (DFS) and overall survival (OS) were defined as the time between the end of SBRT and the first recurrence of the disease or death and the time between the end of SBRT and death from any cause, respectively. The National Cancer Institute’s Common Terminology Criteria for Adverse Events ver. 5.0 was used to assess all toxicities.
4. Statistical analyses
4. Statistical analyses
Statistical analyses were performed using IBM SPSS Statistics for Windows ver. 21.0 (IBM Corp., Armonk, NY). The Kaplan-Meier method was used to calculate the LC, freedom from regional recurrence (FFRR), DMFS, DFS, and OS. A probability level of < 0.05 was considered statistically significant. Univariate analysis was performed through Cox regression analysis to examine the clinical risk factors that influence DFS and OS.
Results
Results
Eighty-nine patients were found to be eligible for the analysis, and their characteristics are shown in Table 1. The median age was 72 years (range, 45 to 90 years), and 55% of the patients were men. Approximately 15.7% of patients had an Eastern Cooperative Oncology Group performance status score of 2 or 3, and 14.6% had an underlying lung disease such as chronic obstructive pulmonary disease, asthma, or interstitial lung disease. Fifty-one patients (57.3%) were previously diagnosed with lung cancer, and most of these patients underwent surgery, although some patients were treated with SBRT or a combination of SBRT and surgery. Seventy-two patients (80.9%) had single primary lung cancer, whereas 17 patients (19.1%) had multiple primary lung cancer, with up to four tumors identified in one patient. In the case of multifocal lesions, some were treated with multiple sessions of SBRT, and some were treated with a combination of surgery and SBRT; a few small, slow-growing lesions were observed. The characteristics of each lesion are shown in Table 2. In all cases, the necessity and possibility of biopsy and surgery were discussed in a multidisciplinary team meeting. Among them, 44 lesions (44.4%) were pathologically diagnosable, and 55 (55.6%) were clinically diagnosed as lung cancer through imaging tests such as serial chest CT and PET-CT. All lesions identified using histological examination were adenocarcinomas. Pure GGN was observed in 43 cases (43.4%), and lesions with subsolid nodule was 56 cases (56.6%). Median value of CTR was 0.3, ranging from 0.0 to 0.5.
The overall follow-up period for the study was median 33.0 months (range, 9.9 to 65.9 months). The follow-up period for tumor response was 23.2 months (range, 1.8 to 65.1 months), during which there was no local progression, and 100% LC was achieved. Any type of recurrence occurred in a total of five patients, and the characteristics of each patient are shown in Table 3. As for the initial pattern of failure, three patients (3.4%) had a regional recurrence, and two patients (2.2%) had a distant recurrence as the first event. It took an average of 12.6 months for the first recurrence to occur and be confirmed on CT and PET-CT images in all cases, and no pathologic confirmation was made. A distant recurrence of abdominal LNs occurred in one of the five patients after a regional recurrence, and all but one patient received systemic therapy as a salvage treatment. A total of eight patients expired, of whom five died of an unknown cause. Of the three patients whose causes of death could be determined, one died of pneumonia and sepsis, and the other two died of cardiac problems unrelated to cancer.
Three-year FFRR, DMFS, DFS, and OS rates were 96.3%, 93.0%, 92.6%, and 91.6%, respectively (Fig. 2A–D). A univariate analysis was performed, and the results are shown in Table 4. None of the risk factors associated with DFS were identified. Patients with a younger age or a higher level of diffusing capacity of the lung for carbon monoxide (DLCO) showed significantly improved OS. Female patients, patients with good performance status, and patients with high levels of forced vital capacity also seemed to have better survival outcomes, but there was no statistical significance. The results of the log-rank test with the Kaplan-Meier method were similar to those of the univariate analysis, showing low 3-year OS rates in the older population (age ≥ 75 years vs. < 75 years, 84.3% vs. 96.8%; p=0.007) and in those with low DLCO (DLCO ≥ 60% vs. < 60%, 96.4% vs. 62.5%; p=0.001) (Fig. 3A and B). Patients with subsolid nodules and pathologically confirmed nodules seemed to have worse survival rates, but this was not significant (Fig. 3C and D).
The toxicity results are shown in Table 5. No patients experienced grade ≥ 3 serious adverse effects for both acute and late toxicities. Five patients with grade 2 radiation pneumonitis (RP) were identified, but they recovered with oral steroid administration.
Discussion
Discussion
The results of the limited studies on the clinical outcomes of SBRT for GGO-predominant lung cancer lesions are shown in S1 Table [1012]. Except for the study by Nagata et al. [11], all showed LC of 100%. In studies using photon beams, various doses were administered depending on the location and size of the tumor, but generally, approximately 50 Gy was prescribed. In the study by Onishi et al. [12], patients were recruited by defining GGO-predominant lung tumors with a CTR of ≤ 0.5 using the same criteria as ours, reporting an LC of 100% and a 3-year OS of 94.6%. Although not all reported rates of regional or distant recurrences are the same, in our case, the recurrence rate appears to be slightly higher than others. This may be because the proportion of patients with pathologically confirmed cancer was quite high at 44.4%, and patients with a history of lung cancer accounted for more than half at 57.3%. According to the subgroup analysis of the study by Onishi et al. [12], the 2-year distant failure rate for patients with a history of lung cancer was 25.0%. Additionally, in a surgical study published in Taiwan, only patients with stage I lung cancer were included, and patients with a higher pathologic T category had a higher chance of any recurrence, DM, and lower OS rates, and patients with a pathology of non-squamous cell carcinoma had a more distant recurrence [13]. Even though recurrences occurred in some patients in our study, all survived well with appropriate salvage therapy and are well managed. The PACIFIC-4 trial, which is currently in progress, is a study on the administration of durvalumab after definitive SBRT in patients with clinical stage I or II node-negative non–small cell lung cancer (NSCLC) [14]. If the results of the PACIFIC-4 trial are promising, they will provide a solid foundation for administering immunotherapeutic agents to patients such as those included in our study. Usually, after SBRT in early-stage lung cancer, RP grade ≥ 2 is known to occur in < 10% of cases, but as many as 29% cases have been reported [15,16]. Symptomatic RP was reported in 5% of participants in the present study, which may be due to a small target volume and advances in planning and targeting techniques.
The 5-year survival rate of patients with clinical stage I NSCLC is 68%–92% [17]. Regarding the long-term outcome for patients with GGO-predominant lung cancer lesions reported by Samsung Medical Center 2015, the 5-year survival rate after wedge resection was 98.6%, and recurrences occurred in 5.1% of patients, which was superior to the prognosis of typical patients with early-stage lung cancer [18]. Tomita et al. [19] published a study comparing the outcomes of SBRT and surgery in patients with stage I NSCLC, including some patients with GGN. There was no significant difference between the two groups in any of the oncologic outcomes, including local recurrence-free survival, DFS, cause-specific survival, or OS. Based on the results of a study of early-stage NSCLC, SBRT and surgery seem to yield comparable results for patients with GGN [6].
The greatest advantage of SBRT is that it can be a useful alternative as a non-invasive local therapy. Patients with insufficient remaining lung volume after multiple lung surgeries or with poor underlying lung function may benefit the most. However, SBRT also has some limitations. First, lobectomy with mediastinal LN sampling or dissection is still suggested as the standard of care. It has the advantage of being able to search for occult LNs surgically. However, occult LN metastasis is rare in GGO-predominant lung cancer lesions and almost always absent in pure nodules [20,21]. Second, many patients receive RT without pathologic confirmation. If salvage treatment is required to recurrence later, it may be difficult to use appropriate drugs without histological information. However, on the other hand, it takes quite a while for recurrence to occur, and since molecular or immunological status changes may occur during that time, it may be better to perform a biopsy again if necessary [22]. Third, evaluating the response of the treated lesion with certainty is often difficult. Owing to various presumed mechanisms, the density and size often increase in the post-SBRT CT image, and the treated nodule may be obscured because of fibrosis [23]. Similar results were obtained in a study of CT findings after SBRT for GGN, and a careful evaluation of the initial response was recommended [10].
The strength of this study is that we recruited a sufficient number of participants with GGO-predominant lung cancer lesions and had a consistent treatment policy as a single institution study. Furthermore, all cases were discussed at the tumor board, and decisions were made on the basis of multi-departmental consensus. As for the radiation dose, a sufficient dose was administered unless the tumor was adjacent to a major organ, such as a central tumor, and 88.9% of patients received an ablative biologically effective dose > 130 Gy10. Several recent studies have shown that survival is improved with an ablative dose for lung cancer [24,25]. Therefore, the results of our institution’s study have a unique advantage in that they better reflect the latest opinion on total dose than those of other institutions (S1 Table).
There are some limitations to our study. First, despite the recommendation of some guidelines to postpone treatment for small lesions, even these lesions were irradiated [4,5]. However, studies have shown that lesions with increased size or density on serial chest CT have a high possibility of malignancy; considering this, we attempted to start treatment as soon as possible [26,27]. Nevertheless, even if some of them are classified as a high-risk group in imaging tests, they may have an indolent characteristic with slow growth and are routinely examined mainly by a department of pulmonary medicine. Unfortunately, when designing this study, we collected the data of patients who had received radiotherapy, so there was no pool of patients who had regular follow-up only without treatment, so direct comparison was not possible. Therefore, it would be useful to carry out comparative analysis with these patients in the future. The second limitation, as mentioned above, is the ambiguity in response evaluations due to changes in the lung parenchyma, such as fibrosis. Accordingly, there are efforts to assess the response using various other imaging techniques other than chest CT [9,28]. Finally, considering that lung cancer with an adenocarcinoma has a longer latency period to recurrence than squamous cell carcinoma, our follow-up period may seem short. This observation period can be explained by the fact that we included only patients treated within the last five years and that there were many elderlies who expired of causes unrelated to lung cancer. However, the RTOG 0618 study also had a follow-up period of about 48 months (range, 15.4 to 73.7 months) for patients with early-stage lung cancer who were operable, and compared to this, it was thought that our study period was not that insufficient [29]. If the next study is conducted over a longer period of time with more patients, it will be of great help in the future.
In conclusion, for patients with GGO-predominant lung cancer lesions, the application of SBRT has been shown to yield high LC and appears to be a safe treatment with low toxicity. SBRT is likely to be one of the alternatives to surgery in patients who have multiple lesions, a surgical history, or who are medically inoperable owing to age or comorbidities.

Electronic Supplementary Material

Electronic Supplementary Material

Supplementary materials are available at Cancer Research and Treatment website (https://www.e-crt.org).
Notes
Notes

Ethical Statement

This study was conducted in accordance with the 1964 Declaration of Helsinki. This study was approved by the Institutional Review Board of Asan Medical Center (#2022-1153), and written informed consent was waived due to the retrospective nature of this study.

Notes
Notes

Author Contributions

Conceived and designed the analysis: Jang JY, Kim SS.

Collected the data: Jang JY, Kim SS.

Contributed data or analysis tools: Jang JY, Kim SS, Song SY, Shin YS, Lee SW, Ji W, Choi CM, Choi EK.

Performed the analysis: Jang JY, Kim SS.

Wrote the paper: Jang JY.

Conflict of interest
Conflict of interest

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

Fig. 1
Flow diagram of patient selection.
crt-2022-1656f1.gif
Fig. 2
(A) Freedom from regional recurrence after stereotactic body radiotherapy for ground-glass nodules. (B) Distant metastasis-free survival after stereotactic body radiotherapy for ground-glass nodules. (C) Disease-free survival after stereotactic body radiotherapy for ground-glass nodules. (D) Overall survival after stereotactic body radiotherapy for ground-glass nodules.
crt-2022-1656f2.gif
Fig. 3
Overall survival after stereotactic body radiotherapy for ground-glass nodules (GGNs) according to (A) age, (B) the level of diffusing capacity of the lung for carbon monoxide (DLCO), (C) the type of GGN, (D) pathologic confirmation.
crt-2022-1656f3.gif
Table 1
Patient characteristics
Variable No. (%) (n=89)
Age (yr), median (range) 72 (45–90)
Sex
 Male 49 (55.1)
 Female 40 (44.9)
ECOG PS
 0 14 (15.7)
 1 61 (68.5)
 2 13 (14.6)
 3 1 (1.1)
Underlying lung diseasea)
 Yes 13 (14.6)
 No 76 (85.4)
Smoking status
 Never-smoker 53 (59.6)
 (Ex-) Smoker 36 (40.5)
Pulmonary function test, median (range)
 FVC (%) 83 (50–129)
 FEV1 (L) 2.04 (0.51–3.70)
 FEV1 (%) 79 (33–121)
 DLCO (%) 76 (34–113)
Past history of lung cancer
 Yes 51 (57.3)
  Surgery for previous lung cancer 47 (52.8)
  SBRT for previous lung cancer 2 (2.2)
  Both surgery and SBRT for previous lung cancer 2 (2.2)
 No 38 (42.7)
No. of tumors, median (range) 1 (1–4)
 Single primary lung cancer 72 (80.9)
 Multiple primary lung cancer 17 (19.1)
Treatment for multiple primary lung cancer
 SBRT only 13 (76.5)
 SBRT+observation 2 (11.8)
 SBRT+operation 2 (11.8)

DLCO, diffusing capacity of the lung for carbon monoxide; ECOG PS, Eastern Cooperative Oncology Group performance status; FEV1, forced expiratory volume in 1 second; FVC, forced vital capacity; SBRT, stereotactic body radiotherapy.

a) The underlying lung diseases include chronic obstructive pulmonary disease, asthma, and interstitial lung disease. The sum of percentages may not be 100, rounded to one decimal place.

Table 2
Characteristics of lesions after SBRT
Variable No. (%) (n=99a))
Biopsy for diagnosis
 Yes 44 (44.4)
 No 55 (55.6)
Reason for unproven pathology
 Lesion close to blood vessels 19 (34.5)
 Deeply located lesions that are difficult to access 10 (18.2)
 Lesions that are too small to obtain a sufficient sample 9 (16.4)
 Clinically diagnosable lesions through follow-up images 17 (30.9)
Ground-glass opacity nodules
 Pure ground-glass opacity nodules 43 (43.4)
 Subsolid nodules 56 (56.6)
Location of tumorb)
 Central tumor 5 (5.1)
 Peripheral tumor 94 (94.9)
Tumor size
 Maximum tumor diameter (mm), median (range) 15.7 (5.7–45.0)
 Maximum consolidation diameter (mm), median (range) 4.5 (0.0–16.0)
 Consolidation-to-tumor ratio, median (range) 0.3 (0.0–0.5)
T category (AJCC eighth edition)
 cTis 43 (43.4)
 cT1mi 8 (8.1)
 cT1a 33 (33.3)
 cT1b 14 (14.1)
 cT1c 1 (1.0)
SBRT dose, median (range)
 Total dose (Gy) 56.0 (48.0–60.0)
 Fraction size (Gy) 14 (10–15)
 BED (Gy10)c) 134.4 (105.6–150.0)
PTV size (cm3) 10.9 (4.1–56.7)
PTV margin from ITV (mm) 5.0 (3.0–8.0)

AJCC, American Joint Committee on Cancer; BED, biologically effective dose; ITV, internal target volume; PTV, planning target volume; SBRT, stereotactic body radiotherapy.

a) Seventy-two patients with a single lesion were treated for each lesion corresponding to 72 cases. Of the 17 patients with multiple lesions, 10 patients treated with SBRT for two ground glass nodules simultaneously, three patients with SBRT for all the multiple lesions (but with only one lesion included in the analysis due to inclusion criteria regarding the consolidation to tumor ratio), and four patients with SBRT to only one lesions, corresponding to total 27 cases totally - all together making 99 cases,

b) As defined by RTOG 0236, lesions within 2 cm of the proximal bronchial tree were classified as central tumors and lesions outside of them were classified as peripheral tumors,

c) Biologically effective doses are calculated using an α/β ratio of 10 Gy. The sum of percentages may not be 100, rounded to one decimal place.

Table 3
Patients with regional recurrence or distant metastasis
Patient No.
1 2 3 4 5
Sex/Age (yr) M/59 M/77 M/74 M/75 F/76
ECOG PS 0 1 1 2 1
Previous lung cancer stage pT2aN0M0 pT3N0M0 pT2aN0M0 pT1bN0M0 pT2aN0M0
TNM stage cTisN0M0 cT1aN0M0 cTisN0M0 cT1aN0M0 cT1aN0M0
No. of nodules 1 1 1 1 1
GGN type Pure GGN Subsolid nodule Pure GGN Subsolid nodule Subsolid nodule
Pathologic confirmation No No No Yes Yes
Regional recurrence Same lobe lung mass and both hilar LN Same lobe lung mass and ipsilateral hilar LN Ipsilateral lung mass - -
Distant metastasis Abdominal LN - - Pleural seeding Pleural seeding
Salvage treatment Systemic therapy Systemic therapy Surveillance Systemic therapy Systemic therapy

ECOG PS, Eastern Cooperative Oncology Group performance status; GGN, ground-glass opacity nodule; LN, lymph node; TNM, tumor–node–metastasis.

Table 4
Factors influencing disease-free survival and overall survival rates after stereotactic body radiotherapy
Variable Univariate analysis Comparison group

Disease-free survival Overall survival


HR (95% CI) p-value HR (95% CI) p-value
Male sex 3.398 (0.379–30.471) 0.274 6.831 (0.837–55.736) 0.073 Female

Age 1.028 (0.926–1.421) 0.602 1.214 (1.086–1.356) 0.001 (Continuous)

ECOG PS, 2–3 1.842 (0.204–16.634) 0.587 4.079 (0.967–17.204) 0.056 0–1

Underlying lung disease, yes 1.392 (0.155–12.466) 0.768 2.032 (0.410–10.078) 0.385 No

Smoking, (Ex-)smoker 2.246 (0.375–13.459) 0.376 2.621 (0.624–11.015) 0.188 Never-smoker

FVC (%) 1.001 (0.947–1.059) 0.961 0.952 (0.907–1.000) 0.050 (Continuous)

FEV1 (L) 1.027 (0.287–3.678) 0.968 0.679 (0.230–2.008) 0.484 (Continuous)

FEV1 (%) 0.992 (0.948–1.037) 0.716 0.745 (0.957–1.032) 0.745 (Continuous)

DLCO (%) 1.016 (0.965–1.070) 0.544 0.933 (0.892–0.976) 0.003 (Continuous)

Metachronous lung cancer, yes 45.429 (0.027–76,854.241) 0.314 0.494 (0.117–2.080) 0.336 No

Multiple primary lung cancer, yes 28.060 (0.002–403,571.066) 0.495 0.561 (0.069–4.563) 0.589 No

Biopsy-proven lung cancer 1.073 (0.179–6.438) 0.939 0.146 (0.660–16.329) 0.146 Unproven

Pure ground-glass nodule 0.732 (0.122–4.383) 0.732 2.682 (0.541–13.310) 0.227 Subsolid nodule

CI, confidence interval; DLCO, diffusing capacity of the lung for carbon monoxide; ECOG PS, Eastern Cooperative Oncology Group performance status; FEV1, forced expiratory volume in 1 second; FVC, forced vital capacity; HR, hazard ratio.

Table 5
Acute and late toxicities after stereotactic body radiotherapy
Event No. of patients (%)
Grade 1 Grade 2 Grade 3 Grade 4
Acute toxicities
 Fatigue 8 (9.0) 0 0 0
 Cough 0 1 (1.1) 0 0
 Sputum 0 1 (1.1) 0 0
 Nausea 1 (1.1) 0 0 0
Late toxicities
 Radiation pneumonitis 73 (82.0) 5 (5.6) 0 0
 Rib fracture 1 (1.1) 3 (3.4) 0 0
 Sputum 1 (1.1) 0 0 0
REFERENCES
REFERENCES

References

1. National Lung Screening Trial Research Team, Aberle DR, Adams AM, Berg CD, Black WC, Clapp JD, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365:395–409.
[Article] [PubMed] [PMC]
2. Migliore M, Fornito M, Palazzolo M, Criscione A, Gangemi M, Borrata F, et al. Ground glass opacities management in the lung cancer screening era. Ann Transl Med. 2018;6:90.
[Article] [PubMed] [PMC]
3. Novello S, Monica V, Serke M, Grohe C, Meyer A, Geissler M, et al. PS01.04 International tailored chemotherapy adjuvant trial: ITACA trial. Final results. J Thorac Oncol. 2021;16(3 Suppl):S58–9.
[Article]
4. Callister ME, Baldwin DR, Akram AR, Barnard S, Cane P, Draffan J, et al. British Thoracic Society guidelines for the investigation and management of pulmonary nodules. Thorax. 2015;70:Suppl 2ii1–54.
[Article] [PubMed]
5. Aokage K, Saji H, Suzuki K, Mizutani T, Katayama H, Shibata T, et al. A non-randomized confirmatory trial of segmentectomy for clinical T1N0 lung cancer with dominant ground glass opacity based on thin-section computed tomography (JCOG1211). Gen Thorac Cardiovasc Surg. 2017;65:267–72.
[Article] [PubMed]
6. Chang JY, Senan S, Paul MA, Mehran RJ, Louie AV, Balter P, et al. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol. 2015;16:630–7.
[Article] [PubMed] [PMC]
7. Chang JY, Mehran RJ, Feng L, Verma V, Liao Z, Welsh JW, et al. Stereotactic ablative radiotherapy for operable stage I non-small-cell lung cancer (revised STARS): long-term results of a single-arm, prospective trial with prespecified comparison to surgery. Lancet Oncol. 2021;22:1448–57.
[Article] [PubMed] [PMC]
8. Rosenzweig K. Stereotactic body radiation therapy as an alter-native to surgery in early-stage non-small-cell lung cancer. Oncology (Williston Park). 2017;31:492–8.
[PubMed]
9. Alcantara P, Martinez BC, Garcia-Esquinas MG, Belaustegui LG, Bustos A. Evaluation of tumor response after stereotactic body radiation therapy for lung cancer: role of 18F-fluorodeoxyglucose positron emission tomography/computed tomography. J Clin Transl Res. 2020;6:155–67.
[PubMed] [PMC]
10. Mikami N, Takeda A, Hashimoto A, Takeda T, Kimura Y, Oku Y, et al. CT findings and treatment outcomes of ground-glass opacity predominant lung cancer after stereotactic body radiotherapy. Clin Lung Cancer. 2022;23:428–37.
[Article] [PubMed]
11. Nagata I, Ogino T, Arimura T, Yoshiura T. Clinical outcomes of proton beam therapy for ground-glass opacity-type lung cancer. Lung Cancer (Auckl). 2020;11:105–11.
[PubMed] [PMC]
12. Onishi H, Shioyama Y, Matsumoto Y, Shibamoto Y, Miyakawa A, Suzuki G, et al. Stereotactic body radiotherapy in patients with lung tumors composed of mainly ground-glass opacity. J Radiat Res. 2020;61:426–30.
[Article] [PubMed] [PMC]
13. Hung JJ, Jeng WJ, Hsu WH, Chou TY, Huang BS, Wu YC. Predictors of death, local recurrence, and distant metastasis in completely resected pathological stage-I non-small-cell lung cancer. J Thorac Oncol. 2012;7:1115–23.
[Article] [PubMed]
14. Robinson C, Hu C, Machtay M, Newton M, Wu K, Barrett K, et al. P1.18-12 PACIFIC-4/RTOG 3515: phase III study of durvalumab following SBRT for unresected stage I/II, lymph-node negative NSCLC. J Thorac Oncol. 2019;14(10 Suppl):S630–1.
[Article]
15. Yamashita H, Nakagawa K, Nakamura N, Koyanagi H, Tago M, Igaki H, et al. Exceptionally high incidence of symptomatic grade 2–5 radiation pneumonitis after stereotactic radiation therapy for lung tumors. Radiat Oncol. 2007;2:21.
[Article] [PubMed] [PMC]
16. Marks LB, Bentzen SM, Deasy JO, Kong FM, Bradley JD, Vogelius IS, et al. Radiation dose-volume effects in the lung. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S70–6.
[Article] [PubMed] [PMC]
17. Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389:299–311.
[Article] [PubMed]
18. Cho JH, Choi YS, Kim J, Kim HK, Zo JI, Shim YM. Long-term outcomes of wedge resection for pulmonary ground-glass opacity nodules. Ann Thorac Surg. 2015;99:218–22.
[Article] [PubMed]
19. Tomita N, Okuda K, Osaga S, Miyakawa A, Nakanishi R, Shibamoto Y. Surgery versus stereotactic body radiotherapy for clinical stage I non-small-cell lung cancer: propensity score-matching analysis including the ratio of ground glass nodules. Clin Transl Oncol. 2021;23:638–47.
[Article] [PubMed]
20. Ye B, Cheng M, Li W, Ge XX, Geng JF, Feng J, et al. Predictive factors for lymph node metastasis in clinical stage IA lung adenocarcinoma. Ann Thorac Surg. 2014;98:217–23.
[Article] [PubMed]
21. Moon Y, Sung SW, Namkoong M, Park JK. The effectiveness of mediastinal lymph node evaluation in a patient with ground glass opacity tumor. J Thorac Dis. 2016;8:2617–25.
[Article] [PubMed] [PMC]
22. Woo CG, Son SM, Lee HC, Han HS, Lee KH, Kim D, et al. Histologic changes in non-small cell lung cancer under various treatments: a comparison of histology and mutation status in serial samples. Cancer Res Treat. 2022;54:737–43.
[Article] [PubMed] [PMC]
23. Dahele M, Palma D, Lagerwaard F, Slotman B, Senan S. Radiological changes after stereotactic radiotherapy for stage I lung cancer. J Thorac Oncol. 2011;6:1221–8.
[Article] [PubMed]
24. Koshy M, Malik R, Weichselbaum RR, Sher DJ. Increasing radiation therapy dose is associated with improved survival in patients undergoing stereotactic body radiation therapy for stage I non-small-cell lung cancer. Int J Radiat Oncol Biol Phys. 2015;91:344–50.
[Article] [PubMed]
25. Moreno AC, Fellman B, Hobbs BP, Liao Z, Gomez DR, Chen A, et al. Biologically effective dose in stereotactic body radiotherapy and survival for patients with early-stage NSCLC. J Thorac Oncol. 2020;15:101–9.
[Article] [PubMed]
26. Hammer MM, Palazzo LL, Kong CY, Hunsaker AR. Cancer risk in subsolid nodules in the National Lung Screening Trial. Radiology. 2019;293:441–8.
[Article] [PubMed] [PMC]
27. Lin YH, Hsu HS. Ground glass opacity on chest CT scans from screening to treatment: a literature review. J Chin Med Assoc. 2020;83:887–90.
[Article] [PubMed] [PMC]
28. Pierson C, Grinchak T, Sokolovic C, Holland B, Parent T, Bowling M, et al. Response criteria in solid tumors (PERCIST/RECIST) and SUV(max) in early-stage non-small cell lung cancer patients treated with stereotactic body radiotherapy. Radiat Oncol. 2018;13:34.
[Article] [PubMed] [PMC]
29. Timmerman RD, Paulus R, Pass HI, Gore EM, Edelman MJ, Galvin J, et al. Stereotactic body radiation therapy for operable early-stage lung cancer: findings from the NRG Oncology RTOG 0618 Trial. JAMA Oncol. 2018;4:1263–6.
[Article] [PubMed] [PMC]

Go to Top