Outcomes of initial therapy for synchronous brain metastases from small cell lung cancer: a single-institution retrospective analysis (2024)

  • Journal List
  • Transl Lung Cancer Res
  • v.13(5); 2024 May 31
  • PMC11157380

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Outcomes of initial therapy for synchronous brain metastases from small cell lung cancer: a single-institution retrospective analysis (1)

Link to Publisher's site

Transl Lung Cancer Res. 2024 May 31; 13(5): 1110–1120.

Published online 2024 May 29. doi:10.21037/tlcr-23-641

PMCID: PMC11157380

Kevin V. Chaung,1 Michael Z. Kharouta,1,2 Andrew J. Gross,1 Pingfu Fu,3 Mitchell Machtay,1,4 Tiffany R. Hodges,5 Andrew E. Sloan,5,6 Tithi Biswas,1,7 Afshin Dowlati,8 and Serah ChoiOutcomes of initial therapy for synchronous brain metastases from small cell lung cancer: a single-institution retrospective analysis (2)1,9

Author information Article notes Copyright and License information PMC Disclaimer

Associated Data

Supplementary Materials

Abstract

Small cell lung cancer (SCLC) has a propensity for brain metastases, which is associated with poor prognosis. We sought to determine predictors of overall survival (OS) and brain progression-free survival (bPFS) in SCLC patients with synchronous brain metastases at the time of initial SCLC diagnosis. A total of 107 SCLC patients with synchronous brain metastases treated at a single institution were included in this retrospective analysis. These patients had brain lesions present on initial staging imaging. Survival was estimated using the Kaplan-Meier method with log-rank test. Factors predictive of OS and bPFS were analyzed using Cox proportional hazards regression model. Median OS for the entire cohort was 9 months (interquartile range, 4.2–13.8 months) and median bPFS was 7.3 months (interquartile range, 3.5–11.1 months). OS was 30.3% at 1 year and 14.4% at 2 years, while bPFS was 22.0% at 1 year and 6.9% at 2 years. The median number of brain lesions at diagnosis was 3 (interquartile range, 2–8), and the median size of the largest metastasis was 2.0 cm (interquartile range, 1.0–3.3 cm). Increased number of brain lesions was significantly associated with decreased OS. Patients who received both chemotherapy and whole brain radiation therapy (WBRT) had improved OS (P=0.02) and bPFS (P=0.005) compared to those who had either chemotherapy or WBRT alone. There was no significant difference in OS or bPFS depending on the sequence of therapy or the dose of WBRT. Thirteen patients underwent upfront brain metastasis resection, which was associated with improved OS (P=0.02) but not bPFS (P=0.09) compared to those who did not have surgery. The combination of chemotherapy and WBRT was associated with improved OS and bPFS compared to either modality alone. Upfront brain metastasis resection was associated with improved OS but not bPFS compared to those who did not have surgery.

Keywords: Small cell lung cancer (SCLC), brain metastases, radiation therapy, chemotherapy, craniotomy

Introduction

Small cell lung cancer (SCLC) is a high-grade neuroendocrine malignancy that generally has a poor prognosis and a high incidence of brain metastases. Approximately 15% of patients have synchronous brain metastases at the time of initial SCLC diagnosis, and the prevalence of brain metastases increases to over 50% at 2 years (1,2). Whole brain radiation therapy (WBRT) has historically been the standard of care for SCLC brain metastases, but emerging data suggests that patients treated with upfront stereotactic radiosurgery (SRS) instead of WBRT have comparable overall survival (OS) outcomes (3,4). Surgery does not typically have a role in the management of SCLC brain metastases, although surgical resection can simultaneously give pathologic diagnosis and relief of neurologic symptoms. SCLC patients were excluded from the three major randomized trials evaluating the role of surgical resection of single brain metastases (5-7). The outcomes of patients following treatments can vary significantly. Using an institutional database, we sought to identify predictors of OS and brain progression-free survival (bPFS) in patients with synchronous brain metastases from SCLC. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-641/rc).

Methods

The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). All patients treated for extensive-stage SCLC (ES-SCLC) at University Hospitals Cleveland Medical Center between 1998 and 2018 were identified using a University Hospitals Institutional Review Board (IRB) approved database (No. CHRV0021). There were minimal risks to the patients for study participation, thus obtaining consents was not required for this retrospective study. All patients with synchronous brain metastases diagnosed with initial staging head computed tomography (CT) or brain magnetic resonance imaging (MRI) scans, with or without extracranial metastases, were included in this retrospective analysis. OS was defined as the duration from date of pathologic diagnosis of SCLC to date of death and was censored at the date of last follow-up for survivors. bPFS was defined as the duration from date of pathologic diagnosis of SCLC to date of last follow-up, intracranial progression, or death (whichever occurred first). All available CT or brain MRI imaging and radiology reports were reviewed for bPFS. Our institution’s standard practice is to perform surveillance brain MRI with and without contrast every 3 months. Survival estimates were generated using the Kaplan-Meier method with log-rank test. Factors associated with OS and bPFS were analyzed using Cox proportional hazards model. Variables that were significant in univariate analyses (P<0.10) were included in multivariate Cox models to adjust for potential confounders. Modified Schoenfeld residuals were used to determine the validity of the proportional hazards assumption. All statistical analysis was performed using R version 4.0.2 (R Foundation for Statistical Computing, Vienna, Austria) using the “survival” and “survminer” packages.

Results

Patient characteristics

We identified 836 ES-SCLC patients treated at our institution between 1998 and 2018, and 107 had synchronous brain metastases (Figure 1). The median follow-up was 9.0 months (interquartile range, 4.8–13.9 months). Table 1 shows patient characteristics and demographics for the entire cohort. At the time of SCLC diagnosis, the median age was 62.7 years (range, 34.2–86.7 years). Fifty-one (47.7%) were male and 56 (52.3%) were female. Seventy-two (67.3%) were Caucasian, 24 (22.4%) were African-American, and 11 (10.3%) had unknown race. Sixty-four (59.8%) had extracranial metastases in addition to brain metastases at initial diagnosis. The median number of brain metastases was three, and the median size of the largest brain metastasis for each patient was 2.0 cm (interquartile range, 1.0–3.3 cm). Fifty-nine (55.1%) had neurologic symptoms at presentation.

Open in a separate window

Figure 1

Study schema of retrospective study. SCLC, small cell lung cancer; WBRT, whole brain radiation therapy.

Table 1

Characteristics/demographicsValues
Age (years), median [range]62.7 [34.2–86.7]
Sex, n (%)
Male51 (47.7)
Female56 (52.3)
Race, n (%)
Caucasian72 (67.3)
African-American24 (22.4)
Unknown11 (10.3)
T-stage, n (%)
T123 (21.5)
T216 (15.0)
T317 (15.9)
T430 (28.0)
Tx8 (7.5)
Unknown13 (12.1)
N-stage, n (%)
N04 (3.7)
N17 (6.5)
N250 (46.7)
N330 (28.0)
Nx1 (0.9)
Unknown15 (14.0)
Number of brain metastases, median [range]3 [1–innumerable]
Size of largest brain metastases (cm),
median [range]
2.0 [0.2–6.7]
No extracranial metastases, n (%)43 (40.2)
Neurological symptoms at diagnosis, n (%)59 (55.1)
WBRT, n (%)90 (84.1)
WBRT dose (Gy), median [range]30 [20–40]
Chemotherapy, n (%)93 (86.9)
Upfront craniotomy, n (%)13 (12.1)
Salvage radiation after WBRT, n (%)15 (14.0)

Open in a separate window

WBRT, whole brain radiation therapy.

As part of their initial therapy, 90 (84.1%) patients received WBRT with a median dose of 30 Gy (range, 20–40 Gy), and 93 (86.9%) received chemotherapy. Seventy-six (71.0%) received both chemotherapy and WBRT, 17 (15.9%) received chemotherapy without WBRT, and 14 (13.1%) received WBRT without chemotherapy. Of the 76 patients who received both chemotherapy and WBRT, 30 (39.5%) received chemotherapy first, 33 (43.4%) received WBRT first, and 13 (17.1%) received chemotherapy and WBRT concurrently (defined as starting WBRT within 14 days of the start of the first cycle of chemotherapy). Nine patients received immunotherapy in addition to chemotherapy. Thirteen of 107 patients (12.1%) underwent upfront craniotomy and brain metastasis resection; 10 of these patients went on to receive both chemotherapy and WBRT, one had chemotherapy alone, and two had WBRT alone.

Performance status data was available for 66 out of 107 patients (62%), including 12 out of the 13 patients (92%) who received upfront craniotomy. Of the 66 patients with recorded performance status, 18 (27%) had Karnofsky performance status (KPS) <70, while 48 (73%) had KPS ≥70. Five craniotomy patients had KPS <70, while seven had KPS ≥70.

Prognostic factors for OS and bPFS

For the entire cohort, median OS was 9.0 months (interquartile range, 4.2–13.8 months), and median bPFS was 7.3 months (interquartile range, 3.5–11.1 months) (Figure 2). There was no significant association between OS and year of diagnosis (Figure S1). Table 2 shows prognostic factors for OS and bPFS on univariate and multivariate analysis. On univariate analysis of OS, lower age [hazard ratio (HR) =1.03, P=0.004], fewer brain metastases (HR =1.01, P=0.07), receipt of both chemotherapy and WBRT (HR =0.38, P<0.001), upfront craniotomy (HR =0.41, P<0.001), and having no extracranial metastases at initial diagnosis (HR =0.67, P=0.056) were significant favorable predictors. On multivariate analysis, fewer brain metastases (HR =1.01, P=0.04), receipt of both chemotherapy and WBRT (HR =0.53, P=0.02), and upfront craniotomy (HR =0.44, P=0.02) remained significant factors for OS. For univariate analysis of bPFS, lower age (HR =1.02, P=0.03), fewer brain metastases (HR =1.01, P=0.052), receipt of both chemotherapy and WBRT (HR =0.44, P<0.001), and upfront craniotomy (HR =0.55, P=0.058) were significant favorable predictors. On multivariate analysis, fewer brain metastases (HR =1.01, P=0.03) and receipt of both chemotherapy and WBRT (HR =0.47, P=0.005) remained significant favorable predictors for bPFS.

Open in a separate window

Figure 2

Kaplan-Meier curves for OS and bPFS for the SCLC synchronous brain metastases patient cohort (n=107 patients). OS, overall survival; bPFS, brain progression-free survival; SCLC, small cell lung cancer.

Table 2

Prognostic factors for OS and bPFS

Patient variableValues (n=107)OS univariateOS multivariatebPFS univariatebPFS multivariate
HR (95% CI)P valueHR (95% CI)P valueHR (95% CI)P valueHR (95% CI)P value
Age (years), median62.71.03 (1.01–1.05)0.0041.01
(0.99–1.03)
0.411.02 (1.00–1.04)0.031.00
(0.98–1.03)
0.92
Sex, n (%)
Male51 (47.7)1.35 (0.91–2.01)0.141.23 (0.83–1.82)0.30
Female56 (52.3)Ref
Race, n (%)
Caucasian72 (67.3)RefRef
African-American24 (22.4)0.98 (0.60–1.60)0.930.98 (0.60–1.59)0.93
Unknown11 (10.3)0.74 (0.38–1.45)0.380.74 (0.38–1.45)0.38
Smoking status, n
Active82RefRef
Never30.40 (0.095–1.68)0.210.40 (0.10–1.68)0.21
Former211.01 (0.61–1.66)0.981.01 (0.61–1.66)0.98
Unknown1
T stage, n
T123RefRef
T2160.94 (0.48–1.83)0.850.88 (0.44–1.75)0.71
T3171.47 (0.78–2.80)0.241.70 (0.89–3.25)0.11
T4300.91 (0.51–1.61)0.740.91 (0.52–1.58)0.73
Tx80.88 (0.39–1.99)0.770.93 (0.42–2.09)0.87
Unknown13
N stage, n
N04RefRef
N171.00 (0.26–3.90)>0.990.80 (0.20–3.12)0.75
N2500.64 (0.20–2.10)0.460.59 (0.18–1.93)0.39
N3300.97 (0.29–3.21)0.960.71 (0.21–2.38)0.58
Nx11.63 (0.17–15.93)0.681.06 (0.11–10.31)0.96
Unknown13
Number of brain metastases, median31.01 (0.99–1.02)0.071.01
(1.00–1.02)
0.041.01 (0.99–1.02)0.0521.01
(1.00–1.02)
0.03
Size of largest brain metastases (cm), median2.00.90 (0.78–1.04)0.150.94 (0.81–1.09)0.40
Chemotherapy + WBRT, n (%)76 (71.0)0.38 (0.24–0.60)<0.0010.53
(0.31–0.92)
0.020.44 (0.29–0.68)<0.0010.47
(0.27–0.79)
0.005
Upfront craniotomy, n (%)13 (12.1)0.41 (0.22–0.79)<0.0010.44
(0.23–0.87)
0.020.55 (0.30–1.02)0.0580.58
(0.31–1.08)
0.09
No extracranial metastases, n (%)43 (40.2)0.67 (0.44–1.01)0.0560.91
(0.58–1.43)
0.680.74 (0.50–1.12)0.14
Symptomatic brain metastases, n (%)59 (55.1)0.98 (0.66–1.45)0.901.16 (0.79–1.72)0.45
Salvage RT after WBRT, n (%)15 (14.0)0.29 (0.15–0.53)<0.00010.58 (0.33–1.01)0.054

Open in a separate window

OS, overall survival; bPFS, brain progression-free survival; HR, hazard ratio; CI, confidence interval; Ref, reference; WBRT, whole brain radiation therapy; RT, radiation therapy.

Patients who received both chemotherapy and WBRT had significantly improved OS and bPFS compared to patients who had either therapy alone. There was no significant difference in OS or bPFS among patients who started chemotherapy first, WBRT first, or both concurrently. WBRT dose (30 Gy/10 fractions vs. 37.5 Gy/15 fractions) was not a significant predictor for OS or bPFS (Figure 3). Of the 90 patients who received WBRT, 21 had intracranial progression at 1 year, and 29 by 2 years. Fifteen of these patients had salvage brain radiation after their initial WBRT; nine had Gamma Knife radiosurgery, four had repeat WBRT, and two had both Gamma Knife radiosurgery followed by repeat WBRT for further progression.

Open in a separate window

Figure 3

Kaplan-Meier curves for OS and bPFS for patients who received WBRT with 37.5 Gy in 15 fractions versus 30 Gy in 10 fractions. OS, overall survival; bPFS, brain progression-free survival; WBRT, whole brain radiation therapy.

Thirteen of 107 patients (12.1%) received upfront craniotomy and brain tumor resection. Table 3 shows the characteristics for each patient who received upfront surgery as well as the indications for surgery. All presented with neurologic symptoms (altered mental status, motor dysfunction, aphasia, ataxia, dysarthria, cognitive/behavioral changes, seizures, or signs of increased intracranial pressure), and only one had histologically proven SCLC prior to surgery. Median OS for this surgery cohort was 18.7 months (interquartile range, 6.6–35.7 months). The surgical cohort had significantly longer OS but not bPFS compared to the non-surgical cohort (Figure 4). Eleven of the 13 patients had both chemotherapy and WBRT following surgery. All but one surgical patient received WBRT, and that patient died only 3.3 months after initial diagnosis.

Table 3

Demographics/characteristics of patients who underwent upfront craniotomy for resection of synchronous brain metastases from small cell lung cancer

PatientAge (years)SexOverall survival (months)Reason for surgeryNumber of brain metastases at diagnosisLargest brain metastases size (cm)KPSTNM stageExtracranial metastatic sitesTime from craniotomy to start of chemo
or WBRT (days)
144.3M10.3Right frontal mass with acute left hemiparesis and AMS64.360T3N2M1cLiver, adrenal, kidney18
259.6M38.8Cerebellar mass with ataxia and dysarthria33.0T2aN2M1cLiver, bone15
362.7F18.7Cerebellar mass with headache, nausea, seizure103.490T1aNxM1cNone31
467.5M13.8Cerebellar mass with headache, ataxia, memory loss, nausea and vomiting24.680TxN2M1cNone22
565.9F25.3Cerebellar mass with ataxia13.330–40TxN2M1bNone20
653.3F32.7Pineal area mass with headache and diplopia12.690TxN2M1cAdrenal16
781.2M5.0Left frontal mass with confusion and aphasia46.750–60TxN3M1cPeritoneum, lymph node72
864.3M8.1Cerebellar mass with seizures, behavioral changes, headache, nausea and vomiting93.550–60T1bN3M1cNone16
953.8F61.5Right cerebral masses with left arm paresis64.470–80T4N2M1cNone27
1062.5F23.2Right frontal mass with left arm paresis46.490T1aN2M1cNone23
1186.7F3.3Cerebellar mass with ataxia and dysarthria15.150–60T1bN1M1cAdrenal, bone42
1264.7F4.2Left occipital mass with vertigo, headache, aphasia26.470–80TxN3M1cNone39
1368.2F39.0Right parietal mass with ataxia, left visual field cut, dizziness125.570–80T1bN2M1cNone34

Open in a separate window

KPS, Karnofsky performance status; TNM, TNM classification of malignant tumors; WBRT, whole brain radiation therapy; M, male; AMS, altered mental status; F, female.

Open in a separate window

Figure 4

Kaplan-Meier curves for OS and bPFS for patients who received upfront craniotomy (n=13 patients) for resection of SCLC synchronous brain metastases versus patients who did not undergo craniotomy (n=94 patients). OS, overall survival; bPFS, brain progression-free survival; SCLC, small cell lung cancer.

Subset analysis was performed on the cohort of 66 patients with recorded performance status using multivariable Cox regression to estimate the effect of upfront craniotomy on OS after controlling for the effect of performance status. Table 4 shows the results of this subset analysis. Upfront craniotomy remained significantly associated with improved OS (HR =0.37, P=0.006).

Table 4

Results of multivariate Cox regression of overall survival in the subset of patients with performance status data available (n=66)

FactorHazard ratio (95% CI)P value
Upfront craniotomy vs. no upfront craniotomy0.37 (0.18, 0.76)0.006
KPS (≥70 vs. <70)0.3 (0.16, 0.57)<0.001

Open in a separate window

CI, confidence interval; KPS, Karnofsky performance status.

Discussion

In this retrospective study of SCLC patients with synchronous brain metastases at initial diagnosis, we sought to identify factors associated with OS and bPFS. Of the 107 patients in this cohort, 13 had upfront craniotomy prior to chemotherapy or WBRT. On multivariate analysis, upfront craniotomy was a significant favorable predictor for OS but not bPFS. Receipt of both chemotherapy and WBRT was a predictor for improved OS and bPFS compared to receipt of either chemotherapy or WBRT alone. For patients who had both chemotherapy and WBRT, there was no significant difference in OS or bPFS among patients who started both concurrently (defined as starting within 14 days of each other), started chemotherapy before WBRT, or started WBRT before chemotherapy.

A recent study based on Surveillance, Epidemiology, and End Results (SEER) data included 33,169 SCLC patients, of whom 5,711 had synchronous brain metastases. This analysis found median OS of 5.0 months for SCLC patients with synchronous brain metastases. This is lower than the OS reported in our cohort (9.0 months), though this may be due to differences in treatment regimens and/or patient characteristics. This study also analyzed the impact of different treatment regimens. Similar to our findings, patients who received both radiation and chemotherapy had improved OS compared to receipt of either modality alone. However, data on surgical resection of brain metastases was not reported in this study (8).

Focal therapies such as SRS and surgical resection have historically not been used for SCLC brain metastases because of its radiosensitive nature and concern for diffuse spread leading to a short interval to distant intracranial failure. The role of radiation for the treatment for SCLC brain metastases is evolving (3,9-12). Two recent studies, the FIRE-SCLC multicenter cohort study and a systematic review and meta-analysis showed no survival detriment for upfront SRS compared to WBRT for selected SCLC patients (3,4). As such, the role of upfront SRS for the treatment of SCLC brain metastases is currently being evaluated in the NRG Oncology cooperative groups clinical trial, NRG-CC009. There remains a paucity of data on surgical resection for SCLC brain metastases aside from case reports (13-15). Prior randomized trials of surgical resection for solitary brain metastases have shown survival improvement compared to WBRT alone, but these trials excluded SCLC and other radiosensitive histologies (5-7,16). Current National Comprehensive Cancer Network (NCCN) guidelines do not list surgical resection as a standard treatment option for SCLC brain metastases (17). We do not view our study as suggestive for a change in the standard of care or guidelines, but rather as hypothesis-generating and warranting further exploration of potential patient characteristics that would help identify patients who may significantly benefit from upfront craniotomy for synchronous SCLC brain metastases.

Our retrospective analysis suggests that surgical resection of symptomatic SCLC brain metastases does not negatively impact survival, as the surgical cohort had a significantly longer OS compared to the overall cohort. There is likely selection bias; the surgical cohort was small (12% of patients) with a higher proportion of patients without extracranial metastases compared to the non-surgical cohort (61.5% vs. 37.2%), and these patients were healthy enough to undergo major surgery. However, the proportions of patients with single brain metastases were similar between cohorts (23.1% vs. 21.3%), and some of the longest-surviving surgical patients had more than three lesions. SCLC brain metastasis surgical specimens may open opportunities for genomic characterization of SCLC brain metastases as has been the case for brain metastases for NSCLC and other primary histologies (18,19). Such molecular studies may help elucidate why SCLC has such a high propensity for brain metastasis compared to other solid tumors.

Platinum doublet chemotherapy has long been the standard systemic treatment for ES-SCLC, and this may also be effective for intracranial disease. The optimal combination and sequence of chemotherapy and radiotherapy for SCLC brain metastases remains unclear. NCCN guidelines allow upfront chemotherapy with deferred WBRT if the brain metastases are asymptomatic (17). Our analysis showed improved OS and bPFS for patients who received both chemotherapy and WBRT as initial therapy compared to either modality alone. Selection bias may explain this, as patients with shorter survival or poorer performance status may not have lived long enough or have been eligible for chemotherapy. Whether these patients received WBRT before, during, or after the first cycle of chemotherapy did not significantly impact OS or bPFS.

WBRT dose (30 Gy/10 fractions vs. 37.5 Gy/15 fractions) was not a significant predictor of OS or bPFS in our study. A post hoc analysis of the N107C trial showed that WBRT 37.5 Gy/15 fractions did not improve cognitive outcomes, PFS, or OS compared to 30 Gy/10 fractions (20). This trial excluded radiosensitive histologies, and our analysis showed similar results for SCLC.

This study has multiple limitations. In particular, performance status data was not available for 38% of the entire patient cohort, and this is an important factor for brain metastasis prognosis (21-23). Our subset analysis on the patients with recorded performance status found that upfront craniotomy remained significantly associated with improved OS even when controlling for performance status. The proportion of patients with missing performance status certainly raises the possibility of selection bias. Moreover, we do not have patient quality of life data. There was also a lack of data on neurologic death vs. death from other causes, which would help elucidate the difference in OS seen between the surgical and non-surgical cohorts. Because this study included patients who were diagnosed with ES-SCLC between 1998 and 2018, the majority of patients were treated without immunotherapy, which is now a standard component of upfront systemic therapy for ES-SCLC after randomized trials showed OS benefit (17,24,25). The few patients in our cohort who did receive immunotherapy were treated with ipilimumab or nivolumab, whereas current standard systemic therapy regimens now incorporate atezolizumab or durvalumab following the publication of the IMpower133 and CASPIAN trials. However, IMpower133 included patients with treated asymptomatic brain metastases, but not untreated ones, limiting evaluation of the impact of atezolizumab on intracranial disease. While the CASPIAN trial did include patients with untreated brain metastases, they only made up about 10% of the cohort. Our study included patients with synchronous brain metastases at initial diagnosis of SCLC, representing a population that was not studied in IMpower133 and was a small minority in the CASPIAN trial. More prospective data is needed to evaluate the efficacy of immunotherapy for SCLC brain metastases. Other changes in treatment protocols between 1998 and 2018 could have impacted outcomes, but a linear regression model found no significant association between OS and year of diagnosis in our study.

Conclusions

In this retrospective analysis of SCLC patients with synchronous brain metastases at initial diagnosis, the combination of chemotherapy and WBRT was associated with improved OS and bPFS compared to either modality alone. Upfront brain metastasis resection was associated with increased OS but not bPFS. Further study is needed to better identify which patients with SCLC brain metastases may benefit from upfront surgical resection.

Supplementary

The article’s supplementary files as

Click here to view.(118K, pdf)

Click here to view.(133K, pdf)

Click here to view.(840K, pdf)

Click here to view.(292K, pdf)

Acknowledgments

Some data from this manuscript was included in a poster presentation at the 2020 American Society for Radiation Oncology (ASTRO) Annual Meeting. The authors would like to thank Dr. Prashant Vempati for his critical review of the manuscript.

Funding: This work was supported by the National Institutes of Health (Grant No. K12CA076917 to S.C.).

Notes

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). All patients treated for extensive-stage SCLC (ES-SCLC) at University Hospitals Cleveland Medical Center between 1998 and 2018 were identified using a University Hospitals Institutional Review Board (IRB) approved database (No. CHRV0021). There were minimal risks to the patients for study participation, thus obtaining consents was not required for this retrospective study.

Footnotes

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-641/rc

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-641/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-641/coif). T.R.H. reports receiving consulting fees from Medtronics which is not relevant to the work in this paper. T.B. reports funding from National Cancer Institute (NCI) Cancer Therapy Evaluation Program (CTEP) as national principle investigator (PI) of a clinical trial 04/22-3/23, which is not relevant to this study. A.D. reports participation in data safety monitoring board or advisory board on Ipsen, Amgen, Jazz, BMS, Astra Zeneca. S.C. reports receiving funding from the National Institutes of Health 01/20-11/23 (grant No. K12CA076917 to S.C. for salary support and research funds); participation in advisory boards for Telix Pharmaceuticals, Seagen Inc. and GT Medical. A.E.S. reports funding from U01 CS236215 06/19-5/23 (PI: Sloan), R01 CA 217956 06/17-05/22 (PI: Brady-Kalnay), R21 CA256573 01/21-12/22 (PI: Sloan), P01 CA CA245705-01 09/20-08/25 (PI: Lathia), Merck Case 3316 12/17-11/21 (PI: Sloan), Coulter Translational Research Fund 09/17-08/22 (PI: Brady-Kalnay), Jobs Ohio 5/22-11/23 (PI: Brady-Kalnay); options for <1% of company shares of Surgical Theater; consulting fees from Medtronic (Visualase), Monteris Medical Inc. and Surgical Theater; patents on the usage of PS-Binding CAR-T Cells and the procoagulant function of cancer stem cells. The other authors have no conflicts of interest to declare.

References

1. Kromer C, Xu J, Ostrom QT, et al.Estimating the annual frequency of synchronous brain metastasis in the United States 2010-2013: a population-based study.J Neurooncol2017;134:55-64. 10.1007/s11060-017-2516-7 [PubMed] [CrossRef] [Google Scholar]

2. Seute T, Leffers P, ten Velde GP, et al.Detection of brain metastases from small cell lung cancer: consequences of changing imaging techniques (CT versus MRI).Cancer2008;112:1827-34. 10.1002/cncr.23361 [PubMed] [CrossRef] [Google Scholar]

3. Rusthoven CG, Yamamoto M, Bernhardt D, et al.Evaluation of First-line Radiosurgery vs Whole-Brain Radiotherapy for Small Cell Lung Cancer Brain Metastases: The FIRE-SCLC Cohort Study.JAMA Oncol2020;6:1028-37. 10.1001/jamaoncol.2020.1271 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

4. Gaebe K, Li AY, Park A, et al.Stereotactic radiosurgery versus whole brain radiotherapy in patients with intracranial metastatic disease and small-cell lung cancer: a systematic review and meta-analysis.Lancet Oncol2022;23:931-9. 10.1016/S1470-2045(22)00271-6 [PubMed] [CrossRef] [Google Scholar]

5. Patchell RA, Tibbs PA, Walsh JW, et al.A randomized trial of surgery in the treatment of single metastases to the brain.N Engl J Med1990;322:494-500. 10.1056/NEJM199002223220802 [PubMed] [CrossRef] [Google Scholar]

6. Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al.Treatment of single brain metastasis: radiotherapy alone or combined with neurosurgery?Ann Neurol1993;33:583-90. 10.1002/ana.410330605 [PubMed] [CrossRef] [Google Scholar]

7. Mintz AH, Kestle J, Rathbone MP, et al.A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis.Cancer1996;78:1470-6. 10.1002/(sici)1097-0142(19961001)78:7<1470::aid-cncr14>3.0.co;2-x [PubMed] [CrossRef] [Google Scholar]

8. Zhou G, Zhang Z, Yu P, et al.Predictive value of clinical characteristics on risk and prognosis of synchronous brain metastases in small-cell lung cancer patients: A population-based study.Cancer Med2023;12:1195-203. 10.1002/cam4.4978 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

9. Gjyshi O, Lin SH, Pezzi TA, et al.Care Patterns for Stereotactic Radiosurgery in Small Cell Lung Cancer Brain Metastases.Clin Lung Cancer2022;23:185-90. 10.1016/j.cllc.2021.07.003 [PubMed] [CrossRef] [Google Scholar]

10. Jiang W, Haque W, Verma V, et al.Stereotactic radiosurgery for brain metastases from newly diagnosed small cell lung cancer: practice patterns and outcomes.Acta Oncol2019;58:491-8. 10.1080/0284186X.2018.1562207 [PubMed] [CrossRef] [Google Scholar]

11. Robin TP, Jones BL, Amini A, et al.Radiosurgery alone is associated with favorable outcomes for brain metastases from small-cell lung cancer.Lung Cancer2018;120:88-90. 10.1016/j.lungcan.2018.03.027 [PubMed] [CrossRef] [Google Scholar]

12. Rusthoven CG, Staley AW, Gao D, et al.Comparison of first-line radiosurgery for small-cell and non-small cell lung cancer brain metastases (CROSS-FIRE).J Natl Cancer Inst2023;115:926-36. 10.1093/jnci/djad073 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

13. Imai R, Hayakawa K, Sakurai H, et al.Small cell lung cancer with a brain metastasis controlled for 5 years: a case report.Jpn J Clin Oncol2001;31:116-8. 10.1093/jjco/hye024 [PubMed] [CrossRef] [Google Scholar]

14. Jesien-Lewandowicz E, Spych M, Fijuth J, et al.Solitary brain metastasis of an occult and stable small-cell lung cancer in a schizophrenic patient: a 3-year control.Lung Cancer2010;69:245-8. 10.1016/j.lungcan.2010.05.002 [PubMed] [CrossRef] [Google Scholar]

15. Drpa G, Popovic F, Nikolic I, et al.Small cell lung cancer with solitary brain metastasis treated with complete resection.Precision Radiation Oncology2018;2:61-3. [Google Scholar]

16. Noordijk EM, Vecht CJ, Haaxma-Reiche H, et al.The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age.Int J Radiat Oncol Biol Phys1994;29:711-7. 10.1016/0360-3016(94)90558-4 [PubMed] [CrossRef] [Google Scholar]

17. National Comprehensive Cancer Network. Small Cell Lung Cancer (Version 2.2024). Accessed December 10, 2023. Available online: https://www.nccn.org/professionals/physician_gls/pdf/sclc.pdf

18. Wang H, Ou Q, Li D, et al.Genes associated with increased brain metastasis risk in non-small cell lung cancer: Comprehensive genomic profiling of 61 resected brain metastases versus primary non-small cell lung cancer (Guangdong Association Study of Thoracic Oncology 1036).Cancer2019;125:3535-44. 10.1002/cncr.32372 [PubMed] [CrossRef] [Google Scholar]

19. Brastianos PK, Twohy E, Anders CK, et al.Alliance A071701: Genomically guided treatment trial in brain metastases.J Clin Oncol2020;38:TPS2573. [Google Scholar]

20. Trifiletti DM, Ballman KV, Brown PD, et al.Optimizing Whole Brain Radiation Therapy Dose and Fractionation: Results From a Prospective Phase 3 Trial (NCCTG N107C [Alliance]/CEC.3).Int J Radiat Oncol Biol Phys2020;106:255-60. Erratum in: Int J Radiat Oncol Biol Phys 2020;106:1111. [PMC free article] [PubMed] [Google Scholar]

21. Gaspar L, Scott C, Rotman M, et al.Recursive partitioning analysis (RPA) of prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain metastases trials.Int J Radiat Oncol Biol Phys1997;37:745-51. 10.1016/s0360-3016(96)00619-0 [PubMed] [CrossRef] [Google Scholar]

22. Sperduto PW, Mesko S, Li J, et al.Survival in Patients With Brain Metastases: Summary Report on the Updated Diagnosis-Specific Graded Prognostic Assessment and Definition of the Eligibility Quotient.J Clin Oncol2020;38:3773-84. 10.1200/JCO.20.01255 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

23. Sperduto PW, De B, Li J, et al.Graded Prognostic Assessment (GPA) for Patients With Lung Cancer and Brain Metastases: Initial Report of the Small Cell Lung Cancer GPA and Update of the Non-Small Cell Lung Cancer GPA Including the Effect of Programmed Death Ligand 1 and Other Prognostic Factors.Int J Radiat Oncol Biol Phys2022;114:60-74. 10.1016/j.ijrobp.2022.03.020 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

24. Horn L, Mansfield AS, Szczęsna A, et al.First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer.N Engl J Med2018;379:2220-9. 10.1056/NEJMoa1809064 [PubMed] [CrossRef] [Google Scholar]

25. Paz-Ares L, Dvorkin M, Chen Y, et al.Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial.Lancet2019;394:1929-39. 10.1016/S0140-6736(19)32222-6 [PubMed] [CrossRef] [Google Scholar]

Articles from Translational Lung Cancer Research are provided here courtesy of AME Publications

Outcomes of initial therapy for synchronous brain metastases from small cell lung cancer: a single-institution retrospective analysis (2024)

References

Top Articles
Latest Posts
Article information

Author: Kimberely Baumbach CPA

Last Updated:

Views: 5969

Rating: 4 / 5 (61 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Kimberely Baumbach CPA

Birthday: 1996-01-14

Address: 8381 Boyce Course, Imeldachester, ND 74681

Phone: +3571286597580

Job: Product Banking Analyst

Hobby: Cosplaying, Inline skating, Amateur radio, Baton twirling, Mountaineering, Flying, Archery

Introduction: My name is Kimberely Baumbach CPA, I am a gorgeous, bright, charming, encouraging, zealous, lively, good person who loves writing and wants to share my knowledge and understanding with you.