Key Points español 中文 (chinese) Question
Does thoracic twice-daily radiotherapy increase the risk for brain metastases after prophylactic cranial irradiation in patients with small cell lung cancer?
Findings
This multicenter cohort study, involving 778 patients during the period from 2003 to 2016, revealed a significantly higher rate of brain metastasis among patients who underwent twice-daily radiotherapy than among those who underwent once-daily radiotherapy, which was further confirmed in subsequent propensity score analysis of 338 patients.
Meaning
Compared with patients with small cell lung cancer who received once-daily radiotherapy, those who received twice-daily radiotherapy were more likely to experience brain metastases after prophylactic cranial irradiation.
Importance
Although thoracic twice-daily radiotherapy (TDRT) is one of the standards of care for small cell lung cancer, its association with brain metastases remains unknown.
Objective
To investigate the association of TDRT vs once-daily radiotherapy (ODRT) with brain metastases after prophylactic cranial irradiation in patients with small cell lung cancer.
Design, Setting, and Participants
In this multicenter cohort study, data on 778 consecutive patients with small cell lung cancer who had undergone thoracic radiotherapy (609 received ODRT and 169 received TDRT), chemotherapy, and prophylactic cranial irradiation were retrieved from the databases of 8 hospitals in China between July 1, 2003, and June 30, 2016. A 1:1 propensity score matching approach was used to control for confounding between the ODRT and TDRT groups. Confounding covariates included 8 demographic variables and 8 treatment-related covariates. Data analysis was conducted from November 1, 2017, to May 31, 2018, and reanalyzed for revision.
Exposures
The ODRT group received 50 to 66 Gy given in 25 to 33 fractions. The TDRT group received 45 Gy given in 30 fractions.
Main Outcomes and Measures
The primary end point was brain metastases. Secondary end points included progression-free survival and overall survival.
Results
Of the 778 patients (median age, 55 years [interquartile range, 48-61 years]), 204 were women and 574 were men. At a median follow-up of 23.6 months (interquartile range, 14.2-38.2 months), 131 patients (16.8%) experienced brain metastases. The rate of brain metastasis at 3 years in the TDRT group was significantly higher than in the ODRT group (26.0% vs 16.9%; hazard ratio, 1.55; 95% CI, 1.06-2.26; P = .03). Of the 338 matched patients (169 in the ODRT group vs 169 in the TDRT group), 60 (17.8%) experienced brain metastases, with a rate at 3 years of 14.9% in the ODRT group vs 26.0% in the TDRT group (hazard ratio, 1.71; 95% CI, 1.02-2.88; P = .04). Progression-free survival was similar in both the whole cohort and the matched cohort. Median overall survival in the ODRT group tended to be significantly longer than in the TDRT group after matching (47.2 vs 32.8 months; hazard ratio, 1.41; 95% CI, 0.99-2.01; P = .06).
Conclusions and Relevance
In this study, patients with small cell lung cancer who received thoracic TDRT appeared to have a higher risk of brain metastases than those who received ODRT, which supports the need for further prospective randomized clinical trials, especially in China and other parts of Asia.
Twice-daily radiotherapy (TDRT) (1.5 Gy twice daily for a total of 45 Gy [to convert gray to rad, multiply by 100]) or once-daily radiotherapy (ODRT) (1.8-2.0 Gy once daily for a total of 60-70 Gy) is recommended for small cell lung cancer in the 2018 National Comprehensive Cancer Network guideline.1 However, the effect of TDRT on brain metastases remains unknown.
The NCCTG (North Central Cancer Treatment Group) 89-20-52 trial reported that the rates of brain metastasis for patients receiving TDRT were numerically higher than the rates of brain metastasis for patients receiving ODRT among 154 patients with limited disease (11% vs 9%; P = .68).2 In stage IIIA or IIIB non–small cell lung cancer, hyperfractionated accelerated radiotherapy was associated with higher rates of brain metastasis than was ODRT (20% vs 13%), but the calculated time point and P value were not reported.3 One retrospective study exploring risk factors for brain metastases after prophylactic cranial irradiation in patients with small cell lung cancer reported that, compared with patients receiving ODRT, patients receiving TDRT were more likely to develop brain metastases (3-year rates, 43% with TDRT vs 21% with ODRT; hazard ratio [HR], 2.171; 95% CI, 1.111-4.243; P = .02).4 However, according to the CONVERT (Concurrent Once-daily Versus Twice-daily Radiotherapy) trial using competing risk analysis, the incidence of brain metastases was similar in each arm (5-year rates, 18.3% with TDRT vs 15.9% with ODRT; HR, 1.15; 95% CI, 0.75-1.79; P = .42).5
To further investigate the association of thoracic TDRT and ODRT with brain metastases after prophylactic cranial irradiation, we conducted this retrospective study of patients with small cell lung cancer treated at 8 institutions in China, using methods that included competing risk analysis and propensity score matching.
In this multicenter cohort study, data on 778 consecutive patients with small cell lung cancer who had undergone thoracic radiotherapy (609 received ODRT and 169 received TDRT), chemotherapy, and prophylactic cranial irradiation were retrieved from the databases of 8 hospitals in China between July 1, 2003, and June 30, 2016. Criteria for eligibility for this study included the following: (1) pathologically or cytologically confirmed small cell lung cancer without a mixture of other pathologic types; (2) receipt of radiotherapy, chemotherapy, and once-daily prophylactic cranial irradiation before relapsing or progression from July 1, 2003, to June 30, 2016; (3) underwent contrast-enhanced cranial computed tomography or magnetic resonance imaging to rule out brain metastases prior to prophylactic cranial irradiation (a prior study4 showed that performance of computed tomography or magnetic resonance imaging before prophylactic cranial irradiation was not significantly related with brain metastases [at 3 years: computed tomography, 28% vs magnetic resonance imaging, 18%; P = .37], so we did not specify computed tomography or magnetic resonance imaging in this study); and (4) no evidence of prior malignant carcinoma during the past 5 years. Patients with incomplete medical records at diagnosis or treatment were excluded from this analysis (Figure 1). Disease stage was not the excluding criteria in this study because Slotman et al6,7 found that patients with an extensive stage of the disease also benefit from prophylactic cranial irradiation and thoracic radiotherapy. Instead, we matched and adjusted for disease stage in further analyses. This retrospective study was approved by the medical-record department of Shandong Cancer Hospital according to the institutional rules that reviewers should not take photographs, share the contents, or breach patients’ privacy. Those who disobey these rules would be revoked the reviewing permission for 3 to 6 months. We obeyed the rules strictly, and all personal information had been removed to protect patient privacy (patient informed consent was exempted). This study followed the International Society for Pharmacoeconomics and Outcomes Research (ISPOR) reporting guideline for comparative effectiveness studies according to the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statements.8
The most commonly used schedules were 50 to 66 Gy given in 25 to 33 fractions once daily for ODRT and 45 Gy given in 30 fractions twice daily for TDRT (eTable 1 in the Supplement). The mainly prophylactic cranial irradiation schedule was 25 Gy given in 10 fractions once daily. Patients who received 24 Gy given in 16 fractions twice daily were excluded.
The biologically effective dose (BED) of thoracic radiotherapy was calculated according to the linear-quadratic formula9: BED = (nd){1 + [d/(α/β)]} – [0.693t/(αTpot)], where n is the total number of fractions delivered, d is the dose per fraction (in units of gray), α/β = 10, α = 0.3 Gy, t is the total number of days in which radiotherapy was delivered, and Tpot is the potential doubling time (5.6 days).9,10
Response to chemoradiotherapy was assessed with the Response Evaluation Criteria in Solid Tumors version 1.1 criteria11 before prophylactic cranial irradiation but was not specified in this study because an earlier study found that response was not associated with brain metastasis (at 3 years: complete response, 29% vs partial response or stable disease, 27%; P = .84).4 The detailed follow-up strategy is presented in the eAppendix in the Supplement. Additional brain radiotherapy (radiosurgery or whole-brain radiotherapy, depending on whether oligometastasis in the brain or multiple brain metastasis) and chemotherapy were adopted for patients with brain metastases but not specified in this study.
The primary end point was brain metastases confirmed by cranial imaging at any time whether neurologic symptoms (eg, headache or vomiting) were present or not. The secondary end points were progression-free survival (progression of disease at the first time in any sites confirmed by imaging) and overall survival. All end points were analyzed as time-to-event data from the start of thoracic radiotherapy to the respective events, which were subject to censoring at the last follow-up (data cutoff was November 13, 2017) if no events were observed. The brain metastases were evaluated using competing risk analysis (the Gray test for univariate analysis and the Fine-Gray model for multivariable regression12,13), in which death without brain metastases was treated as a competing event. Both progression-free survival and overall survival were analyzed using the Kaplan-Meier method and Cox proportional hazards regression models. Six clinically important covariates (year of diagnosis, performance status, disease stage, thoracic radiotherapy, combination of chemoradiotherapy, and timing of prophylactic cranial irradiation) were included for multiple analysis.
A 1:1 optimal propensity score–matched method was used to control confounding between the ODRT (control) and TDRT (treated) groups14 to essentially estimate the average treatment effect on the treated, instead of the average treatment effect on the entire sample. Propensity scores (ie, the conditional probability of receiving TDRT) were calculated using a multivariable logistic regression model. The covariates used to calculate propensity scores included 8 demographic variables (treating site, year of diagnosis, age at diagnosis, sex, performance status, smoking history, laterality, and disease stage) and 8 treatment-related covariates (surgery, combination of chemoradiotherapy, type of initial chemotherapy regimen, types of chemotherapy regimen involved, chemotherapy cycles, thoracic radiotherapy time from diagnosis to start of therapy, timing of prophylactic cranial irradiation, and dose classification of prophylactic cranial irradiation), which were summarized and compared between the TDRT and ODRT groups using χ2 tests, both prior to and after matching. All tests were 2-sided, and P < .05 was considered statistically significant. Statistical analyses were performed from November 1, 2017, to May 31, 2018, and reanalyzed for revision using IBM SPSS, version 22.0 (IBM Corp) and R, version 2.15.3 (R Project for Statistical Computing).
Of the 894 consecutive patients queried, 778 met the study criteria with complete medical records and were included (Figure 1). Of the 778 patients (median age, 55 years [interquartile range, 48-61 years]), 204 (26.2%) were women, 490 (63.0%) were smokers (among the 574 male patients, 478 [83.3%] were smokers), 321 (41.3%) underwent radiotherapy sequentially after 2 to 4 cycles of chemotherapy (sequential chemoradiotherapy) rather than concurrently (concurrent chemoradiotherapy) largely owing to performance status or age, 609 (78.3%) received ODRT, and the other 169 (21.7%) received TDRT at treating physicians’ discretions based on their department’s inclinations (in some departments, physicians prefer ODRT for all of their patients, while in other departments, physicians prefer TDRT) (Table 1). The median duration of thoracic radiotherapy was 64 days (interquartile range, 42-102 days).
Risk of Brain Metastases and Survival Analyses
Of the 778 patients, 131 (16.8%) developed brain metastases at a median follow-up time of 23.6 months (interquartile range, 14.2-38.2 months), with a 3-year rate of 18.5% (95% CI, 15.6%-21.7%). Univariate analyses showed that, compared with patients treated with ODRT, those treated with TDRT were more likely to experience brain metastases after prophylactic cranial irradiation (3-year rate, 16.9% in the ODRT group vs 26.0% in the TDRT group; HR, 1.55; 95% CI, 1.06-2.26; P = .03) (Figure 2A). Disease stage (HR, 1.64; 95% CI, 1.03-2.62; P = .03) was significantly associated with brain metastases (Table 1). Multivariable analysis confirmed that patients who received TDRT (HR, 1.57; 95% CI, 1.04-2.37; P = .03) or those with extensive disease (HR, 1.69; 95% CI, 1.03-2.77; P = .04) had higher rates of brain metastasis. No significant difference between ODRT and TDRT was observed in overall survival (HR, 1.15; 95% CI, 0.88-1.50; P = .31) or progression-free survival (HR, 1.10; 95% CI, 0.87-1.37; P = .44). Multiple analysis showed that patients with poorer performance status (overall survival: HR, 1.38; 95% CI, 1.03-1.83; P = .03; progression-free survival: HR, 1.23; 95% CI, 0.97-1.56; P = .08) or those who underwent prophylactic cranial irradiation before the end of chemoradiotherapy (overall survival: HR, 1.37; 95% CI, 1.05-1.78; P = .02; progression-free survival: HR, 1.34; 95% CI, 1.07-1.68; P = .01) had poorer survival. In addition, patients who received a diagnosis in earlier years (2003-2010) or with extensive disease experienced shorter progression-free survival (Table 2).
Propensity score matching was used to further evaluate the role of TDRT and ODRT. A total of 338 patients were matched successfully. As shown in Table 3, patients’ clinical features, excluding treatment site, were balanced between the TDRT and ODRT groups after matching.
After a median follow-up of 25.9 months (interquartile range, 15.7-35.2 months), 60 of the 338 matched patients (17.8%) developed brain metastases, with a 3-year rate of 14.9% for the ODRT group and 26.0% for the TDRT group (HR, 1.71; 95% CI, 1.02-2.88; P = .04) (eTable 2 in the Supplement; Figure 2B). A total of 123 patients in the matched cohort (36.4%) died, with median overall survival of 47.2 months in the ODRT group and 32.8 months in the TDRT group (HR, 1.41; 95% CI, 0.99-2.01; P = .06) (eFigure in the Supplement). A total of 188 patients in the matched cohort (55.6%) experienced progression, with median progression-free survival of 20.1 months in the ODRT group vs 18.8 months in the TDRT group (HR, 1.16; 95% CI, 0.87-1.55; P = .30) (eFigure in the Supplement).
For the matched cohort, the median BED was lower in the TDRT group than in the ODRT group (43.1 vs 51.8 Gy; P < .001). The period from the start of any therapy to the end of radiotherapy (SER) was shorter in the TDRT group than in the ODRT group (median, 81 vs 108 days; P < .001). Neither BED nor SER was associated with brain metastases, progression-free survival, or overall survival. The differences in overall survival between the ODRT and TDRT groups became more obvious after adjusting for BED and SER (HR, 1.69; 95% CI, 1.05-2.71; P = .03) (eTable 3 in the Supplement).
Among the 5 excluded patients who received twice-daily prophylactic cranial irradiation (24 Gy given in 16 fractions twice daily) (screened among the 894 patients but not included among the 778 patients) (Figure 1), 3 (60%) experienced brain metastases. One of these 5 patients also received thoracic TDRT; he developed brain metastases. In addition, the asymptomatic brain metastasis ratio was 65.6% in the ODRT group vs 60.5% in the TDRT group (P = .59) before matching and 57.1% in the ODRT group vs 60.5% in the TDRT group (P = .80) after matching.
Other Prognostic Factors and Subgroup Analyses
Subgroup analyses based on disease stage, year of diagnosis, and timing of prophylactic cranial irradiation were performed among the 338 matched patients. Patients who received TDRT showed higher risks of developing brain metastases when adjusting by disease stage (HR, 1.71; 95% CI, 1.02-2.87; P = .04). Possibly because of reduced sample size and statistical power, the rates of brain metastasis in the TDRT group were only numerically higher in either limited-stage or extensive-stage subgroups (eTable 4 in the Supplement).
For patients who received a diagnosis of small cell lung cancer in recent years (2011-2016), no significant difference in the rate of brain metastasis was observed (HR, 1.47; 95% CI, 0.80-2.68; P = .21). Although for those who received a diagnosis in earlier years (2003-2010), the rate of brain metastasis was significantly higher in the TDRT group (HR, 3.05; 95% CI, 1.09-8.53; P = .03). After adjusting for year of diagnosis, TDRT continued to be significantly associated with higher risks of brain metastasis (HR, 1.78; 95% CI, 1.05-3.00; P = .03) (eTable 5 in the Supplement).
In addition, the rate of brain metastasis in earlier or recent years by stratum of ODRT and TDRT was analyzed to see whether time itself would affect the results. It showed that, for patients with ODRT, the rate of brain metastasis of those who received a diagnosis in earlier years was not significantly different from those who received a diagnosis in recent years (HR, 0.97; 95% CI, 0.37-2.55; P = .95). Although for patients with TDRT, the rate of brain metastasis was significantly higher for those who received a diagnosis in earlier years than for those who received a diagnosis recently (HR, 0.46; 95% CI, 0.22-0.96; P = .04). After adjustment for ODRT and TDRT, time of diagnosis was not significantly associated with risks of brain metastases (HR, 0.61; 95% CI, 0.34-1.10; P = .10) (eTable 6 in the Supplement).
For patients with late prophylactic cranial irradiation, the rates of brain metastasis tended to be higher in the TDRT group but marginally significant (HR, 1.81; 95% CI, 0.99-3.31; P = .05), and the rates of brain metastasis were not significantly different among those who received early prophylactic cranial irradiation (HR, 1.39; 95% CI, 0.49-3.99; P = .54). Patients treated with TDRT showed a trend of higher, but marginally significant, risks of developing brain metastases when adjusting by the timing of prophylactic cranial irradiation (HR, 1.71; 95% CI, 1.01-2.89; P = .05) (eTable 7 in the Supplement).
This multicenter study revealed that, compared with ODRT, TDRT was associated with higher incidences of brain metastases, as shown in both multivariable analysis based on the whole cohort of 778 patients and the propensity score–matched cohort of 338 patients. Additional subgroup analyses also suggest that such disparity may be independent of disease stages and timing of prophylactic cranial irradiation.
Our analysis therefore suggests that ODRT may be superior to TDRT in controlling brain metastases in patients with small cell lung cancer, with multiple unverified, hypothesis-generating underlying mechanisms. One of the possibilities may be related to the impairment of the blood–spinal cord barrier or the blood-brain barrier. Irradiation disrupts the blood–spinal cord barrier, with an associated increase in vascular permeability in the early period after irradiation (24 hours).15,16 Because spinal cord tissue and vascular endothelial cells are considered late reaction tissues with a very slow rate of turnover,17,18 the repair of sublethal injury in spinal cord tissue and vascular endothelial cells appears to be somewhat longer.3 Twice-daily radiotherapy involves the delivery of the target dose in a shorter time interval between fractions and provides healthy tissues with less time to repair sublethal radiation damage, which leads to an accumulation of incomplete repair and results in an asymptomatic biological response characterized by sequential physiological changes in the thoracic spinal cord.16,19,20 Sublethal injury with protractedly less time for repair leads to more severe disruption of the permeability of the thoracic blood–spinal cord barrier and results in more transmigration of residual tumor cells during TDRT compared with ODRT. The transmigrated tumor cells metastasize to the brain along with cerebrospinal fluid, form metastatic niches, and generate colonization in the brain months later.21
This inference can also be supported by the subgroup analyses spanning 13 years, during which time significant advancements in radiotherapy technique and supportive care have been made; thus, we have adjusted for the year of diagnosis to minimize the potential confounding in our study. It showed that, for patients who received a diagnosis of small cell lung cancer in earlier years, the rate of brain metastasis was obviously higher in the TDRT group, which remained significant after adjusting years. We even compared the rate of brain metastasis in earlier and recent years by stratum of ODRT and TDRT to see the association of time itself. Again, the results showed that, for patients undergoing TDRT, the rate of brain metastasis was significantly higher among those who received a diagnosis in earlier years. However, after adjustment for ODRT and TDRT, time of diagnosis was not significantly associated with risk of brain metastases. In other words, TDRT was more detrimental in earlier years. The improved radiologic techniques and better supportive care over the years might minimize the difference of brain metastases between ODRT and TDRT. Thus, improved techniques can decrease the level of injury to the thoracic blood–spinal cord barrier and mask the difference. This finding may also explain the results of studies from Western countries, such as the CONVERT study, in which there was no difference in the rates of brain metastasis between patients who underwent TDRT and those who underwent ODRT.5
In addition, the incidence of brain metastases among patients who received prophylactic cranial irradiation twice daily was much higher than among patients who received once-daily prophylactic cranial irradiation, which further indicated that irradiating the whole brain twice daily would more obviously injure the blood-brain barrier. In addition, data from other studies, such as the RTOG (Radiation Therapy Oncology Group) 0212 on prophylactic cranial irradiation, also showed that the rate of brain metastasis at 1 year was 10.6% in the twice-daily arm (36 Gy given in 24 fractions) vs 6.2% in the once-daily arm (36 Gy given in 18 fractions) (total, 21% in the twice-daily arm vs 10% in the once-daily arm)22; although the P value was not reported, the rate in the group that received twice-daily prophylactic cranial irradiation was obviously higher in terms of numerical value.
The timing of prophylactic cranial irradiation was controversial. Data from Lee et al23 showed the trend that the overall incidence of brain metastases was higher in the group that received late prophylactic cranial irradiation (offered irradiation after 5-6 courses of chemotherapy) than in the group that received early irradiation group (offered after 2-3 courses of chemotherapy) (23.6% vs 14.3%; P = .08). There was no difference in overall survival between the 2 groups. The pooled analysis conducted by Schild et al24 also showed that the timing was not associated with subsequent survival across all patients (HR, 1.00; 95% CI, 0.99-1.01; P = .76). According to Aupérin et al,25 via classifying the interval between the initiation of induction therapy and prophylactic cranial irradiation into less than 4 months, 4 to 6 months, and longer than 6 months, they identified a trend toward a reduction in the rate of brain metastasis with earlier prophylactic cranial irradiation after the initiation of chemotherapy without an overall survival difference. Sas-Korczyńska et al26 found that early prophylactic cranial irradiation (performed during chemoradiotherapy) was more effective compared with irradiation applied after combined therapy, which decreased the rate of brain metastasis from 20% to 7.3% (P = .009). However, the prophylactic administration of cranial irradiation concurrent with systemic therapy inevitably increased the risk of neurotoxic effects and hematotoxic effects.10,27,28 Some physicians do not like to apply prophylactic cranial irradiation too early because they think that prophylactic cranial irradiation decreases the rate of brain metastasis via eliminating micrometastases that cannot be detected by radiologic methods, rather than preventing metastases.29 Undergoing prophylactic cranial irradiation too early attenuates the effects since micrometastases have not developed yet. In our study, early prophylactic cranial irradiation did not decrease the rate of brain metastasis significantly compared with late irradiation, but it significantly shortened overall survival and progression-free survival. In line with the National Comprehensive Cancer Network guideline,1 our data support that prophylactic cranial irradiation should be administered after the resolution of the acute toxic effects of initial chemoradiotherapy.
Previous studies have shown that the BED and SER may be associated with overall survival10,30,31; we also found that the BED was lower and the SER was shorter in the TDRT group in our study. However, the multivariable analysis that jointly evaluated BED, SER, and TDRT and ODRT showed that BED and SER had no significant association with outcomes, which further supports the hypothesis that the observed difference in the incidences of brain metastases may be attributed to the frequency of radiotherapy.
In our study, the lower incidence of brain metastases in the ODRT group did not translate into improved overall survival in the whole cohort. In fact, such an observation is not rare and has been reported before,32-34 which could be explained by the potential confounding of effective subsequent therapies with higher treatment-related financial costs.35 However, after matching with balanced cases, the longer overall survival in the ODRT group became marginally significant, which further indicates that the effect of ODRT vs TDRT is of clinical significance.
In addition, our data showed that patients receiving ODRT lived longer than patients in previous studies.36 The longer overall survival resulted in a higher long-term incidence of brain metastases and might also have contributed to the observation of the significant difference in the incidence of brain metastases in our series. A possible cause for the longer overall survival in our study may be that all our patients were Chinese, whereas less than 1% of patients in the CONVERT trial36 were of Asian origin. Faivre-Finn et al36 also discussed that their results might not be applicable to other races/ethnicities. It is not unusual that different races/ethnicities may show different responses to the same treatment regimen. A case in point is that tyrosine kinase inhibitors are effective for Chinese patients but are less effective for European or American patients37,38 because there are more Chinese patients with an epidermal growth factor receptor mutation. In addition, our patients were younger with fewer smokers, which was in line with official Chinese data.39,40 According to Jia et al,39 among the enrolled 14 106 male patients, 11 750 (83.3%) were smokers. In another study that included 3320 patients, 2223 (67.0%) were smokers.40 Other reasons were briefly discussed in an earlier study.4
In summary, TDRT was introduced decades ago to clinical practice based on radiobiological principles.41 It is time to review real-world experiences and reconsider its value. Our findings may shed light on the benefit of TDRT with the use of the longest feasible interfraction interval20 (eg, 12 hours between fractions in the twice-daily regimen). Benefit may even be seen in stopping the use of TDRT for patients with small cell lung cancer, especially in China or other parts of Asia where twice-daily irradiation is logistically less preferred compared with once-daily irradiation because more and more ODRT schedules have shown noninferior outcomes.42-44
This study has several limitations. First, owing to the retrospective nature of this study, a fraction of patients with missing data had to be excluded from our analysis, which may have led to bias (such as incorrect estimates of difference between types of radiotherapy), limited the generalizability of our findings, and reduced the power to detect clinically meaningful differences in clinical outcomes. Second, despite the fact that we made significant efforts to minimize the potential selection biases using regression and propensity score matching, the current analysis could still be subject to unobserved confounding. Third, the centers are not balanced even after matching. However, considering that patients from the same institution had the same treatment-related or follow-up policy and that the detection rate of asymptomatic brain metastasis via imaging is similar between the ODRT group and the TDRT group in both the whole cohort and the matched cohort, we do not think centers would affect the results. In fact, our data showed that there was only very limited variability in brain metastasis rates across centers.
In this study, compared with thoracic ODRT, TDRT appears to be associated with a higher risk of brain metastases after prophylactic cranial irradiation in Chinese patients with small cell lung cancer. These findings may motivate more in vitro and in vivo research to further investigate the underlying mechanisms and may also affect the clinical option of the thoracic radiotherapy schedule, especially in China or other parts of Asia.
Accepted for Publication: January 4, 2019.
Published: May 17, 2019. doi:10.1001/jamanetworkopen.2019.0103
Open Access: This is an open access article distributed under the terms of the CC-BY License. © 2019 Zeng H et al. JAMA Network Open.
Corresponding Authors: Jinming Yu, MD, PhD, Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, 440 Jiyan Rd, Jinan 250117, Shandong, China (sdyujinming@163.com); Shuanghu Yuan, MD, PhD, Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, 440 Jiyan Rd, Jinan 250117, Shandong, China (yuanshuanghu@sina.com).
Author Contributions: Drs J. Yu and Yuan had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs J. Yu and Yuan contributed equally to this work. Drs H. Zeng, R. Li, C. Hu, and Qiu contributed equally to this work.
Concept and design: H. Zeng, H. Yu, M. Hu, H. Li, Lu, Zhu, J. Yu, Yuan.
Acquisition, analysis, or interpretation of data: H. Zeng, R. Li, C. Hu, Qiu, Ge, Zhang, P. Zeng, Xiao, Miao, Wei, Ni, Shen, H. Li, Yue, Fan, X. Hu, Kong, J. Yu, Yuan.
Drafting of the manuscript: H. Zeng, C. Hu, H. Yu, M. Hu, Xiao, Wei, Ni, Shen, H. Li, Lu, Kong, Yuan.
Critical revision of the manuscript for important intellectual content: H. Zeng, R. Li, C. Hu, Qiu, Ge, H. Yu, Zhang, P. Zeng, Miao, Yue, Fan, Zhu, X. Hu, Kong, J. Yu, Yuan.
Statistical analysis: H. Zeng, R. Li, C. Hu, Miao, Ni, Shen, H. Li, Kong, Yuan.
Obtained funding: Kong, J. Yu, Yuan.
Administrative, technical, or material support: H. Zeng, R. Li, Qiu, Ge, H. Yu, H. Li, Yue, Lu, Kong, J. Yu, Yuan.
Supervision: H. Yu, Ni, H. Li, Kong, J. Yu, Yuan.
Conflict of Interest Disclosures: Dr H. Zeng reported receiving grants 81372413 and 81472813 from the National Natural Science Foundation of China and grant ZR2015HZ004 from Shandong Provincial Key Project of Translational Medicine during the conduct of the study. Dr C. Hu reported receiving personal fees from Varian Medical Systems outside the submitted work. Dr H. Yu reported receiving grants and personal fees from China National Natural Scientific Fund during the conduct of the study. Dr Xiao reported receiving grant 81372413 from the National Natural Science Foundation of China during the conduct of the study. Dr J. Yu reported receiving grant ZR2015HZ004 from Shandong Provincial Key Project of Translational Medicine during the conduct of the study. No other disclosures were reported.
Funding/Support: This project was supported by the grants from the National Natural Science Foundation of China and Shandong Provincial Key Project of Translational Medicine.
Role of the Funder/Sponsor: The funding sources had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Additional Contributions: We thank the patients and their families who took part in this study and the caregivers of the study participants. Huiqing Li, PhD, Department of Epidemiology, College of Public Health of Shandong University, The Institute of Basic Medicine of Shandong Academy of Medical Sciences, and Yi Liu, PhD, Department of Biostatistics, School of Public Health of Shandong University, consulted on this study. Amy Miller, MA, CCRP, PMP, Department of Radiation Oncology, Indiana University School of Medicine, provided assistance with the manuscript. Drs Li and Liu and Ms Miller did not receive any compensation for their assistance.
2.Schild
SE, Bonner
JA, Shanahan
TG,
et al. Long-term results of a phase III trial comparing once-daily radiotherapy with twice-daily radiotherapy in limited-stage small-cell lung cancer.
Int J Radiat Oncol Biol Phys. 2004;59(4):943-951. doi:
10.1016/j.ijrobp.2004.01.055PubMedGoogle ScholarCrossref 3.Belani
CP, Wang
W, Johnson
DH,
et al; Eastern Cooperative Oncology Group. Phase III study of the Eastern Cooperative Oncology Group (ECOG 2597): induction chemotherapy followed by either standard thoracic radiotherapy or hyperfractionated accelerated radiotherapy for patients with unresectable stage IIIA and B non–small-cell lung cancer.
J Clin Oncol. 2005;23(16):3760-3767. doi:
10.1200/JCO.2005.09.108PubMedGoogle ScholarCrossref 6.Slotman
B, Faivre-Finn
C, Kramer
G,
et al; EORTC Radiation Oncology Group and Lung Cancer Group. Prophylactic cranial irradiation in extensive small-cell lung cancer.
N Engl J Med. 2007;357(7):664-672. doi:
10.1056/NEJMoa071780PubMedGoogle ScholarCrossref 10.Schild
SE, Bonner
JA, Hillman
S,
et al. Results of a phase II study of high-dose thoracic radiation therapy with concurrent cisplatin and etoposide in limited-stage small-cell lung cancer (NCCTG 95-20-53).
J Clin Oncol. 2007;25(21):3124-3129. doi:
10.1200/JCO.2006.09.9606PubMedGoogle ScholarCrossref 15.Li
YQ, Chen
P, Jain
V, Reilly
RM, Wong
CS. Early radiation-induced endothelial cell loss and blood-spinal cord barrier breakdown in the rat spinal cord.
Radiat Res. 2004;161(2):143-152. doi:
10.1667/RR3117PubMedGoogle ScholarCrossref 17.van der Kogel
AJ. Radiation-induced damage in the central nervous system: an interpretation of target cell responses.
Br J Cancer Suppl. 1986;7:207-217.
PubMedGoogle Scholar 18.Hirst
DG, Denekamp
J, Hobson
B. Proliferation studies of the endothelial and smooth muscle cells of the mouse mesentery after irradiation.
Cell Tissue Kinet. 1980;13(1):91-104.
PubMedGoogle Scholar 20.Thames
HD, Ang
KK, Stewart
FA, van der Schueren
E. Does incomplete repair explain the apparent failure of the basic LQ model to predict spinal cord and kidney responses to low doses per fraction?
Int J Radiat Biol. 1988;54(1):13-19. doi:
10.1080/09553008814551461PubMedGoogle ScholarCrossref 22.Wolfson
AH, Bae
K, Komaki
R,
et al. Primary analysis of a phase II randomized trial Radiation Therapy Oncology Group (RTOG) 0212: impact of different total doses and schedules of prophylactic cranial irradiation on chronic neurotoxicity and quality of life for patients with limited-disease small-cell lung cancer.
Int J Radiat Oncol Biol Phys. 2011;81(1):77-84. doi:
10.1016/j.ijrobp.2010.05.013PubMedGoogle ScholarCrossref 24.Schild
SE, Foster
NR, Meyers
JP,
et al; North Central Cancer Treatment Group. Prophylactic cranial irradiation in small-cell lung cancer: findings from a North Central Cancer Treatment Group pooled analysis.
Ann Oncol. 2012;23(11):2919-2924. doi:
10.1093/annonc/mds123PubMedGoogle ScholarCrossref 25.Aupérin
A, Arriagada
R, Pignon
JP,
et al; Prophylactic Cranial Irradiation Overview Collaborative Group. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission.
N Engl J Med. 1999;341(7):476-484. doi:
10.1056/NEJM199908123410703PubMedGoogle ScholarCrossref 26.Sas-Korczyńska
B, Korzeniowski
S, Wójcik
E. Comparison of the effectiveness of ‘late’ and ‘early’ prophylactic cranial irradiation in patients with limited-stage small cell lung cancer.
Strahlenther Onkol. 2010;186(6):315-319. doi:
10.1007/s00066-010-2088-3PubMedGoogle ScholarCrossref 29.Hu
X, Chen
M. Prophylactic cranial irradiation for limited-stage small cell lung cancer: controversies and advances [in Chinese].
Zhongguo Fei Ai Za Zhi. 2013;16(7):373-377.
PubMedGoogle Scholar 30.De Ruysscher
D, Pijls-Johannesma
M, Bentzen
SM,
et al. Time between the first day of chemotherapy and the last day of chest radiation is the most important predictor of survival in limited-disease small-cell lung cancer.
J Clin Oncol. 2006;24(7):1057-1063. doi:
10.1200/JCO.2005.02.9793PubMedGoogle ScholarCrossref 32.Gore
EM, Bae
K, Wong
SJ,
et al. Phase III comparison of prophylactic cranial irradiation versus observation in patients with locally advanced non–small-cell lung cancer: primary analysis of Radiation Therapy Oncology Group study RTOG 0214.
J Clin Oncol. 2011;29(3):272-278. doi:
10.1200/JCO.2010.29.1609PubMedGoogle ScholarCrossref 33.De Ruysscher
D, Dingemans
AC, Praag
J,
et al. Prophylactic cranial irradiation versus observation in radically treated stage III non–small-cell lung cancer: a randomized phase III NVALT-11/DLCRG-02 study.
J Clin Oncol. 2018;36(23):2366-2377. doi:
10.1200/JCO.2017.77.5817PubMedGoogle ScholarCrossref 34.Takahashi
T, Yamanaka
T, Seto
T,
et al. Prophylactic cranial irradiation versus observation in patients with extensive-disease small-cell lung cancer: a multicentre, randomised, open-label, phase 3 trial.
Lancet Oncol. 2017;18(5):663-671. doi:
10.1016/S1470-2045(17)30230-9PubMedGoogle ScholarCrossref 36.Faivre-Finn
C, Snee
M, Ashcroft
L,
et al; CONVERT Study Team. Concurrent once-daily versus twice-daily chemoradiotherapy in patients with limited-stage small-cell lung cancer (CONVERT): an open-label, phase 3, randomised, superiority trial.
Lancet Oncol. 2017;18(8):1116-1125. doi:
10.1016/S1470-2045(17)30318-2PubMedGoogle ScholarCrossref 37.Wu
YL, Zhong
WZ, Li
LY,
et al. Epidermal growth factor receptor mutations and their correlation with gefitinib therapy in patients with non-small cell lung cancer: a meta-analysis based on updated individual patient data from six medical centers in mainland China.
J Thorac Oncol. 2007;2(5):430-439. doi:
10.1097/01.JTO.0000268677.87496.4cPubMedGoogle ScholarCrossref 38.Thatcher
N, Chang
A, Parikh
P,
et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non–small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer).
Lancet. 2005;366(9496):1527-1537. doi:
10.1016/S0140-6736(05)67625-8PubMedGoogle ScholarCrossref 39.Jia
M, Li
J, Lin
H, Zou
X, Zhao
P. Effect of smoking on lung cancer histology and its epidemiology in Chinese male [in Chinese].
Zhongguo Fei Ai Za Zhi. 2017;20(8):516-521.
PubMedGoogle Scholar 40.Jiang
W, Kang
Y, Shi
GY,
et al. Comparisons of multiple characteristics between young and old lung cancer patients.
Chin Med J (Engl). 2012;125(1):72-80.
PubMedGoogle Scholar 42.Grønberg
BH, Halvorsen
TO, Fløtten
Ø,
et al; Norwegian Lung Cancer Study Group. Randomized phase II trial comparing twice daily hyperfractionated with once daily hypofractionated thoracic radiotherapy in limited disease small cell lung cancer.
Acta Oncol. 2016;55(5):591-597. doi:
10.3109/0284186X.2015.1092584PubMedGoogle ScholarCrossref 43.Xia
B, Hong
LZ, Cai
XW,
et al. Phase 2 study of accelerated hypofractionated thoracic radiation therapy and concurrent chemotherapy in patients with limited-stage small-cell lung cancer.
Int J Radiat Oncol Biol Phys. 2015;91(3):517-523. doi:
10.1016/j.ijrobp.2014.09.042PubMedGoogle ScholarCrossref