Skip to main content
Erschienen in: Radiation Oncology 1/2018

Open Access 01.12.2018 | Research

Comorbidity indexing for prediction of the clinical outcome after stereotactic body radiation therapy in non-small cell lung cancer

verfasst von: Julia Dreyer, Michael Bremer, Christoph Henkenberens

Erschienen in: Radiation Oncology | Ausgabe 1/2018

Abstract

Purpose

To determine the prognostic impact of comorbidity and age in medically inoperable early-stage non-small cell lung cancer (NSCLC) treated with stereotactic body radiotherapy (SBRT) using the age-adjusted Charlson Comorbidity Index (aCCI).

Patients and methods

Between November 2008 and January 2015, 196 consecutive patients with medically inoperable NSCLC were treated with SBRT at a single institution. The prescribed isocenter dose was either 60.0 Gray (Gy) in six fractions for central lung cancer or 56.25 Gy in three fractions for peripheral lung cancer. Baseline comorbidities were retrospectively retrieved according to available outclinic medical records as well as the hospital information system. The aCCI was scored for each patient and subjected according to outcome and toxicity as well as all of the single items of the aCCI and other clinical parameters using univariate and multivariate analysis.

Results

Thirty-one point 6 % (62/196) of patients were deceased, of whom 17.3% (34/196) died due to lung cancer and 14.3% (28/196) due to comorbidities. The median overall survival (OS) was 15.0 months (95% CI [11.9–18.1]), whereas the median cancer-specific survival (CSS) was not reached. An aCCI ≥7 compared with an aCCI ≤6 was significantly associated with an increased risk of death (HR 1.79, 95% CI [1.02–2.80], p = 0.04) and cancer-specific death (HR 9.26, 95% CI [4.83–24.39], p < 0.001), respectively. Neither OS nor CCS were significantly associated with age, sex, side (left vs. right), lobe, localization (central vs. peripheral), packyears, TNM, or any item of the aCCI. Considering the 14.3% (28/196) of deceased patients who died due to comorbidities, aCCI ≥9 was significantly associated with non-cancer-related death (HR 3.12, 95% CI [1.22–8.33], p = 0.02). The observed cumulative rate of radiation pneumonitis (RP) ≥2 was 12.7% (25/196). The aCCI had no statistical association with RP.

Conclusion

Advanced age and numerous comorbidities characterizing this patient population were successfully assessed using the aCCI in terms of survival. Therefore, we recommend that age and comorbidity be indexed using the aCCI as a simple scoring system for all patients treated with SBRT for lung cancer.
Hinweise
First author: Julia Dreyer
Co-Author: Michael Bremer
Last/Senior author: Christoph Henkenberens
Abkürzungen
aCCI
Age-adjusted Charlson Comorbidity Index
CIRS-G
Cumulative Illness Rating Scale for Geriatrics
COPD
Chronic obstructive pulmonary disease
CSS
Cancer-specific survival
CT
Computed tomography
CTCAE
Common Toxicity for Criteria Adverse Events
CWT
Chest wall toxicity
FDG
18F-fluorodeoxyglucose
G
Grading
Gy
Gray
ITV
Internal target volume
LENT-SOMA
Late Effects on Normal Tissue-subjective, objective, management scales
NSCLC
Non-small cell lung cancer
OS
Overall survival
PET
Positron emission tomography
PTV
Planning target volume
RE
Radiation esophagitis
RP
Radiation pneumonitis
SBRT
Stereotactic body radiotherapy

Introduction

Lobectomy remains the standard of care for early-stage non-small cell lung cancer (NSCLC) in medically fit patients [1], but approximately 20% of patients are medically inoperable due to comorbidities, old age, or both [2]. Among the strategies to improve control rates, stereotactic body radiotherapy (SBRT) is the most favored. Numerous reports have indicated extremely good local control after SBRT with an excellent toxicity profile [35].
However, the reported overall survival rates after SBRT for early-stage NSCLC tend to be worse than local control. This has frequently been attributed to competing comorbidities because patients are treated with SBRT instead of surgery due to their comorbidities [69]. The choice against surgery and in favor SBRT has been found to depend on local practice [6] and patient-specific factors [79]. Baseline comorbidities and their prognostic impacts on the clinical outcome have not been assessed using a simple and objective comorbidity score. With this study, we aimed to make another step towards this goal. The objective of this retrospective study cohort was therefore to use the age-adjusted Charlson Comorbidity Index (aCCI) [10], as it is tempting to use given its simplicity, to investigate the impact of comorbidities on the outcome of NSCLC treated with SBRT.

Patients and methods

Patients

Between November 2008 and January 2015, 196 patients with medically inoperable NSCLC were treated with SBRT at a single institution. Patient were collected by reviewing the available outclinic medical records and the medical records of the hospital information system. Comorbidities were encoded using the aCCI (Table 1). The selection criteria were medically unfit for surgery or declination of surgery and staging of tumor and distant metastasis based on positron emission tomography (PET) computed tomography (CT) and biopsy of the tumor if the medical condition allowed bronchoscopy or CT guided biopsy.
Table 1
Age-adjusted Charlson Comorbidity Index (aCCI) [10]
Score
Comorbid condition
1
Myocardial infarction
Congestive heart failure
Cerebral vascular disease
Peripheral vascular disease
Dementia
COPD
Connective tissue disease
Peptic ulcer disease
Mild liver disease
Agea
2
Diabetes
Hemiplegia
Moderate(Severe renal disease
Diabetes with end-organ damage
Solid tumor
Leukemia
Lymphoma
3
Moderate/severe liver disease
6
Metastatic solid tumor
Acquired immunodeficiency syndomre
a1pint is added to aged 41–50 years, 2 points for those aged 51–60 years, 3 points for those 61–70 years, and 4 points for those 71 years or older

Radiotherapy

Patients were fixed in a stereotactic body frame system with a customized vacuum pillow (Elekta, Stockholm, Sweden) using abdominal compression and free breathing. The gross tumor volume was defined based on CT findings in lung and soft tissue windows including all small spiculae. Slow scan cone beam computed tomography was performed to determine the internal target volume (ITV) until October 2014, and 4-dimensional CT was used after that. We added a margin of 4 mm in all directions to the ITV to define the planning target volume (PTV). SBRT treatment planning was conducted with Oncentra Masterplan (Elekta, Stockholm, Sweden). Irradiation was performed as multifield irradiation using a linac accelerator every second day. The prescribed isocenter dose for peripheral located tumors was 18.75 Gy (PTV border covered by the 67% isodose), and the total dose was 56.25 in three fractions. Centrally located tumors usually received an isocenter dose of 7.5 Gy (PTV boarder covered by the 80% isodose), and the total dose was 60.0 Gy. Dosimetric calculation was conducted using a pencil beam algorithm with heterogeneity correction. The constraints for RT planning are described elsewhere [5, 11]. In some patients, the dose was individually adjusted to the dose exposure of organs at risk. The detailed patient characteristics are summarized in Table 2.
Table 2
Patient characteristics
 
n = 196 (%); median
range
Sex
 female
73 (37.1)
 
 male
123 (62.9)
 
Medically inoperable
182 (92.8)
 
Medically operable
14 (7.2)
 
Localization
 -central
83 (42.3)
 
 -peripheral
113 (57.7)
 
Side
 -left
86 (43.9)
 
 -right
110 (56.1)
 
Grading (G)
 G1
7 (3.6)
 
 G2
74 (37.8)
 
 G3
42 (21.4)
 
Stage according to UICC (7th edition)
 
 -I
113 (57.7)
 
 -II
68 (34.6)
 
 -IIIa
15 (7.7)
 
Histology
 -Adenocarcinoma
49 (39.9)
 
 -Squamous cell carcinoma
71 (57.7)
 
 -Large cell carcinoma
3 (2.4)
 
 -No biopsy due to comorbidities
73 (37.2)
 
Age
67
29–86
 0–50
6 (3.1)
 
 50–65
63 (32.1)
 
 66–80
101 (51.5)
 
 > 80
26 (13.3)
 
aCCI
7
3–16
 0–3
4 (2.0)
 
 4–6
63 (32.1)
 
 7–9
62 (31.6)
 
 10–12
44 (22.4)
 
 > 12
23 (11.9)
 
Hypertension
119 (60.7)
 
Diabetes with or without end-organ damage
52 (26.6)
 
Moderate/severe renal damage
63 (32.1)
 
COPD
167 (85.2)
 
 -Gold 1 + 2
51 (26.0)
 
 -Gold 3
61 (31.1)
 
 -Gold 4
55 (28.1)
 
Peripheral vascular disease
49 (25)
 
Myocardial infarction
31 (15.8)
 
Congestive heart failure
71 (36.2)
 
Cerebral vascular disease
13 (6.6)
 
Mild liver disease
9 (4.5)
 
Isocenter Dose
 -peripheral tumor
18.75
18–20
 -central tumor
7.5
7–9
Packyears
40
0–120

Follow-up

Follow-up visits were performed every 3 months and included CT of the chest and abdomen. 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) was performed when CT was suspicious for relapse. The date of relapse was determined as the date when FDG-PET was assessed as positive for local and/or distant relapse by experienced nuclear physicians or when biopsy proved relapse in medically fit patients. Overall survival (OS) was defined as the period from the last day of SBRT to the date of death from any cause. Lung cancer death was defined as death resulting from the progression of lung cancer (local and/or distant), and non-lung cancer death was defined as death of any other cause due to comorbidities. Locoregional relapse was defined as any relapse within the lung or mediastinum, and distant metastases were defined as lung cancer lesions outside the lung and mediastinum.

Toxicity

Toxicity was assessed weekly during SBRT by anamnesis and physical examination. Acute toxicity was defined from the start of SBRT up to 90 days after the last day of irradiation and was graded according to the Common Toxicity for Criteria Adverse Events (CTCAE V 4.0) [12]. Late toxicity was defined as symptoms > 90 days after the last fraction of SBRT and was classified according to the Late Effects on Normal Tissue-Subjective, Objective, Management scales (LENT-SOMA) [13].

Statistics

The outcomes were statistically assessed using Kaplan Meier analysis with log-rank test and Cox regression analysis.
Toxicity was statistically assessed with univariate analyses using the Chi-squared-test for non-parametric parameters and Student’s t-test for parametric parameters. Multivariate logistic regression analysis included all significant parameter from the univariate analysis using backwards elimination to determine the parameters that contributed the most to toxicity. The factors evaluated were age, sex, histology, grading, side, localization, TNM stage, packyears, aCCI and all single items of the aCCI. Statistical analysis was performed with a commercially available software package (SPSS V.24, IBM, Armonk, NY, USA).

Results

Outcome

The median overall survival was 15.0 (3.0–64.0) months for all patients and the median follow-up was 24.0 months (6–64.0) for patients who were alive (66.8% [131/196]. Concerning all patients, 31.6% (62/196) were deceased and 1.6% (3/196) were lost to follow-up. Seventeen point 3 % (34/196) of patients died due to lung cancer, 6.1% (12/196) due to locoregional failure and 11.2% (22/196) due to distant extrapulmonary metastases. Furthermore, 14.3% (28/196) of patients died due to comorbidities. The detailed results are shown in Table 3.
Table 3
Descriptive outcome analysis
Status
n
%
Alive
 
131
66.8
Deceased
 
62
31.6
Unknown
 
3
1.6
Death from lung cancer
 
34
17.3
 Locoregional failure
12
6.2
 Distant progression
22
11.2
Death from comorbidities
28
14.3
 cardiovascular
 
8
4.1
 lung
 
6
3.1
 infection
 
4
2.0
 stroke
 
3
1.5
 other
 
7
3.6
The median OS was 15.0 months (95% CI [11.9–18.1], Fig. 1a), whereas the median cancer-specific survival (CSS) was not reached (Fig. 1b). In addition, 45.2% (28/62) of the deceased patients died from competing comorbidities and 54.8% (34/62) from lung cancer. Neither OS (Fig. 2a) nor CCS (Fig. 2b) was significantly worse for central tumors compared with peripheral tumors (HR 1.05, 95% CI [0.64–1.70], p = 0.85; HR 1.40, 95% CI [0.73–2.70], p = 0.31). Considering the survival of the presented patient cohort divided by the median aCCI of 7, aCCI ≥7 compared with a aCCI of ≤6 was found to be significantly associated with an increased hazard for death (HR 1.79, 95%CI [1.02–2.80],p = 0.04) and cancer-specific death (HR 9.26, 95% CI [4.83–24.39], p < 0.001), respectively. The corresponding Kaplan Meier curves of the OS and CCS are shown in Fig. 3. Neither OS nor CCS was significantly associated with age, sex, side (left vs. right), lobe, localization (central vs. peripheral), packyears, TNM, or any item of the aCCI. Considering the 14.3% (28/196) of deceased patients who died due to comorbidities, aCCI ≥9 was significantly associated with non-cancer-related death (HR 3.12, 95% CI [1.22–8.33], p = 0.02).

Toxicity

Due to the low number of events, the frequencies of acute and late toxicity were assessed cumulatively. We observed no fatal toxicity related to SBRT.
Radiation pneumonitis (RP) of grade 1 occurred in 34.7% (68/196), of grade 2 in 11.2% (22/196), of grade 3 in 1.0% (2/196), of grade 4 in 0.5% (1/196) and of grade 5 in 0% (0/196) of patients, respectively. This resulted in a cumulative RP ≥2 rate of 12.7% (25/196). Univariate analysis revealed that tumors located on the right lung side (p = 0.01) were associated with clinically relevant RP ≥ grade 2. Age, sex, lobe, localization (central vs. peripheral), packyears, TNM, aCCI nor any item of the aCCI were statistically associated with RP ≥ grade 2.
In total, 7.7% (15/196) of patients developed a radiation esophagitis (RE) grade ≥ 2. No patients (0/113) with peripheral tumors developed an RE grade ≥ 2, whereas 16.9% (14/83) of the patients with central tumors developed acute RE grade 2, and 1.2% (1/83) acute RE grade 3, respectively. No late RE ≥ grade 1 was observed. Univariate statistical analysis revealed no significant parameters associated with RE. Furthermore, in 2.1% (4/196), a mild chest wall toxicity (CWT) grade 1 with no need for narcotics was observed. No CWT ≥ grade 2 was observed. In addition, none of the assessed toxicities (RP, RE, CWT) were associated with items of the aCCI.

Discussion

To our best knowledge, the presented study represents the largest early-stage lung cancer population treated with SBRT to quantify the impact of baseline co-morbidities on the clinical outcome.
The observed median overall survival of 15.0 months was low, although this rate is consistent with other studies [1416]. Convincing data suggest that poor survival–despite high local control rates–is attributed to advanced age and competing comorbidities because subgroup analysis revealed that medically operable patients treated with SBRT had a much higher survival than medically inoperable patients treated with SBRT [1720], which was recently confirmed in a prospective single-arm phase 2 study conducted by the NRG Oncology Radiation Therapy Oncology Group [21]. Although a large randomized trial comparing surgery with SBRT for medically operable early stage NSCLC does not exist; SBRT is a good alternative to surgery [1721] with lower direct medical costs and better quality-adjusted life expectancies [22]. Furthermore, Eguchi et al. showed in a competing risks analysis of curative-intent resection of stage I lung cancer that high age was a significant parameter for worse short-term outcome and 1-point increase of the CCI (not age-adjusted) decreased the overall survival by 14% [23].
Therefore, it is lucid to assume that reported data from lung cancer patients treated with SBRT are biased by not precisely recorded high age, number and severity of comorbidities and not by the technique of SBRT itself, which may compromise survival. Therefore, the age-adjusted Charlson Comorbidity Index (aCCI) was used in this study to assess the prognostic significance of age and co-morbidity. The improved survival of early-stage NSCLC –particularly in medically unfit patients- is related to the widespread adoption of SBRT to the clinical routine [2427]. A direct comparison of the survival between lobectomy and SBRT is limited by the inherent unmeasured biases of databases warranting dedicated prospective trials [24, 25].
The results of our analysis provide some reassurance that it is indeed advanced age coupled with competing baseline comorbidity rather than overlooked treatment-related mortality that is largely responsible for the low observed OS and CSS rates post-SBRT. Patients with aCCI ≥7 had a significantly increased hazard for death and cancer-specific death compared with patients with an aCCI ≤6. The median aCCI was 7, suggesting usually three other competing comorbidities for a patient cohort with a median age of 70. This is considerably higher than in other reports [14, 28, 29], although some studies do not use the age-adjusted CCI [14, 29]. Nevertheless, in our analysis neither age nor any of the single comorbidities of the aCCI were significantly associated with outcome suggesting that an age-adjusted comorbidity index, such as the aCCI, should be used instead of an index that does not adjust for age because age is generally associated with poor survival after SBRT for lung cancer [30]. The CCI in general does not graduate severity of comorbidities in a precise way, and Extermann et al. were cautious about the CCI due to its tendency to underrate the functional status of older cancer patients [31]. In the particular case of SBRT, lung cancer patients are often not eligible for surgery due to chronic obstructive pulmonary disease (COPD) Gold III or Gold IV, which is the major reason for allocating to SBRT. The aCCI distinguishes chronic pulmonary disease as yes or no but does not take into account that patients with severe COPD Gold III or IV have a substantially reduced life expectancy, even without lung cancer [32, 33]. The severity of COPD classified according to the Gold criteria was not assessed, although that might have been a confounder that influenced survival analysis using aCCI. An alternative score is the Cumulative Illness Rating Scale for Geriatrics (CIRS-G), which allows a subjective grading of severity of comorbidities in elderly patients [34, 35], but the CIRS-G has never been used in a large patient cohort with mainly medically inoperable lung cancer patients treated with SBRT [20]. The CIRS-G is much more complex, more labor-intensive and less user-friendly than the aCCI because the user of the CIRS-G needs complex multidisciplinary knowledge, and sometimes the CIRS-G even requires further medical consultation [20, 34, 35]. Therefore, the aCCI is a more applicable and faster scoring system than the CIRS-G.
RP is considered to be the most important toxicity with rates of RP ≥2 ranging from 9 to 28% [36]. We observed no abnormally increased rate of clinically relevant RP ≥2 of 12.7% in the presented patient collective with mainly multimorbid patients. Several risk factors, such as age, sex, severity of COPD, baseline lung function and smoking status, have been reported with controversial results [22, 3739]. Statistical analyses showed that tumor location on the right lung was associated with RP ≥2, which was also observed by Chaudari et al. [37]. Basically, it can be assumed that the small patient cohorts and the low incidence of RP have introduced bias into the statistical results, and thus, the results of statistical analyses have to be interpreted with caution. Therefore, we cannot rule out that the statistical association with RP ≥2 and tumor on the right lung side might be a random result.
Some limitations of this study should be acknowledged. First, its retrospective character has inherent limitations and might have introduced a selection bias. Second, this study included a selected cohort with mainly medically inoperable patients. Therefore, caution should be applied when transferring the observed results to medically fit and operable patients. Third, the aCCI certainly does not include all outcome relevant comorbidities and does not grade comorbidities according to their severity in a precise way. Additionally, the aCCI was not developed specifically for carcinoma patients, but is a more general tool to estimate the prognosis of patients. Fourth, this study is based on clinical parameters, although dosimetric parameters also have impact on outcome and side effects [5, 22, 40, 41]. Fifth, the follow-up period of 24 months for patients who were alive and the number of deceased patients (cancer related and non-cancer related death) might be insufficient to make definitive statements about long-term cancer survival and survival of comorbidities. Therefore we cannot predict long-term outcome and claim that we successfully assessed age and numerous comorbidities in general. Ideally, there would be a validation cohort for this purpose.
Nevertheless, the observed results are robust and the aCCI was a simple tool for estimation of prognosis in medically unfit patients.

Conclusion

The results of the present study indicate that SBRT for early stage lung cancer is a well-tolerated treatment modality that offers long-term tumor control. The advanced age and numerous comorbidities characterizing this patient population were successfully assessed with the aCCI in terms of survival. Therefore, we recommend that age and comorbidity should be indexed using the aCCI as a simple score for all patients treated with SBRT for lung cancer.

Acknowledgements

We thank Mr. Bernhard Vaske for his statistical support.

Funding

Not applicable.

Availability of data and materials

Supporting data will not be provided, but can be shared question-related.
This retrospective study complied with the regulations of the local institutional review board and the principles of the Declaration of Helsinki.
All authors approved the submitted manuscript.

Competing interest

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Crino L, Weder W, van Meerbeck J, Felip E. ESMO guidelines working group. Early stage and locally advanced (non-metastatic) non-small-cell lung cancer: ESMO clinical practice Guidleines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v103–15.CrossRef Crino L, Weder W, van Meerbeck J, Felip E. ESMO guidelines working group. Early stage and locally advanced (non-metastatic) non-small-cell lung cancer: ESMO clinical practice Guidleines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v103–15.CrossRef
2.
Zurück zum Zitat Raz DJ, Zell Ja OSH, Gandara SR, Anton-Culver H, Jablons DM. Natural history of stage I non-small cell lung cancer: implications for early detection. Chest. 2007;132:193–9.CrossRef Raz DJ, Zell Ja OSH, Gandara SR, Anton-Culver H, Jablons DM. Natural history of stage I non-small cell lung cancer: implications for early detection. Chest. 2007;132:193–9.CrossRef
3.
Zurück zum Zitat Timmerman RD, Park C, Kavanagh BD. The north American experience with stereotactic body radiation therapy in non-small cell lung cancer. J Thorac Oncol. 2007;2:101–12.CrossRef Timmerman RD, Park C, Kavanagh BD. The north American experience with stereotactic body radiation therapy in non-small cell lung cancer. J Thorac Oncol. 2007;2:101–12.CrossRef
4.
Zurück zum Zitat 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 randomized trials. Lancet Oncol. 2015;16:630–7.CrossRef 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 randomized trials. Lancet Oncol. 2015;16:630–7.CrossRef
5.
Zurück zum Zitat Guckenberger M, Andratschke N, Alheit H, Holy R, Nestle U, Sauer O. Definition of stereotactic body radiotherapy: principles and practice for the treatment of stage I non-small cell lung cancer. Strahlenther Onkol. 2014;190:26–33.CrossRef Guckenberger M, Andratschke N, Alheit H, Holy R, Nestle U, Sauer O. Definition of stereotactic body radiotherapy: principles and practice for the treatment of stage I non-small cell lung cancer. Strahlenther Onkol. 2014;190:26–33.CrossRef
6.
Zurück zum Zitat Khakwani A, Al R, Powell HA, Tata LJ, Stanley RA, Baldwin DR, et al. Lung cancer survival in England: trends in non-small-cell lung cancer survival over the duration of the National Lung Cancer Audit. Br J Cancer. 2013;109:2058–65.CrossRef Khakwani A, Al R, Powell HA, Tata LJ, Stanley RA, Baldwin DR, et al. Lung cancer survival in England: trends in non-small-cell lung cancer survival over the duration of the National Lung Cancer Audit. Br J Cancer. 2013;109:2058–65.CrossRef
7.
Zurück zum Zitat Crabtree TD, Denlinger CE, Meyers BF, El Naqa I, Zoole J, Krupnick AS, et al. Stereotactic body radiation therapy versus surgical resection for stage I non-small cell lung cancer. J Thorac Cardiovasc Surg. 2010;140:377–86.CrossRef Crabtree TD, Denlinger CE, Meyers BF, El Naqa I, Zoole J, Krupnick AS, et al. Stereotactic body radiation therapy versus surgical resection for stage I non-small cell lung cancer. J Thorac Cardiovasc Surg. 2010;140:377–86.CrossRef
8.
Zurück zum Zitat Palma D, Senan S. Improving outcomes for high-risk patients with early- stage non-small-cell lung cancer. Insight from population-based data and the role of stereotactic ablative radiotherapy. Clin Lung Cancer. 2013;14:1–5.CrossRef Palma D, Senan S. Improving outcomes for high-risk patients with early- stage non-small-cell lung cancer. Insight from population-based data and the role of stereotactic ablative radiotherapy. Clin Lung Cancer. 2013;14:1–5.CrossRef
9.
Zurück zum Zitat Palma DA, Visser O, Lagerwaard FJ, Belderbos J, Slotman B, Senan S. Treatment of stage I NSCLC in elderly patients: a population-based matched-pair comparison of stereotactic radiotherapy versus surgery. Radiother Oncol. 2011;101:240–4.CrossRef Palma DA, Visser O, Lagerwaard FJ, Belderbos J, Slotman B, Senan S. Treatment of stage I NSCLC in elderly patients: a population-based matched-pair comparison of stereotactic radiotherapy versus surgery. Radiother Oncol. 2011;101:240–4.CrossRef
10.
Zurück zum Zitat Charlson ME, Szwatrowski TP, Peterson J, Gold J. Validation of a combined index. J Clin Epidemiol. 1994;47:1245–51.CrossRef Charlson ME, Szwatrowski TP, Peterson J, Gold J. Validation of a combined index. J Clin Epidemiol. 1994;47:1245–51.CrossRef
11.
Zurück zum Zitat Benedict SH, Yenice KM, Followill D, Galvin JM, Hinson W, Kavangh B, et al. Stereotactic body radiation therapy: the report of AAPM task group 101. Med Phys. 2010;37:4078–101.CrossRef Benedict SH, Yenice KM, Followill D, Galvin JM, Hinson W, Kavangh B, et al. Stereotactic body radiation therapy: the report of AAPM task group 101. Med Phys. 2010;37:4078–101.CrossRef
13.
Zurück zum Zitat LENT SOMA scales for all anatomic sites. Int J Radiat Oncol Biol Phys. 1995;31:1049–92. LENT SOMA scales for all anatomic sites. Int J Radiat Oncol Biol Phys. 1995;31:1049–92.
14.
Zurück zum Zitat Mohkles S, Nuyttens JJ, Maat AP, Birim Ö, Aerts JG, Bogers AJ, et al. Survival and treatment of non-small cell lung cancer stage I-II treated surgically or with stereotactic body radiotherapy: patient and tumor-specific factors affect the prognosis. Ann Surg Oncol. 2015;22:316–23.CrossRef Mohkles S, Nuyttens JJ, Maat AP, Birim Ö, Aerts JG, Bogers AJ, et al. Survival and treatment of non-small cell lung cancer stage I-II treated surgically or with stereotactic body radiotherapy: patient and tumor-specific factors affect the prognosis. Ann Surg Oncol. 2015;22:316–23.CrossRef
15.
Zurück zum Zitat Nyman J, Johansson KA, Hulten U. Stereotactic hypofractionated radiotherapy for stage I non-small cell lung cancer – mature results for medically inoperable patients. Lung Cancer. 2006;51:97–103.CrossRef Nyman J, Johansson KA, Hulten U. Stereotactic hypofractionated radiotherapy for stage I non-small cell lung cancer – mature results for medically inoperable patients. Lung Cancer. 2006;51:97–103.CrossRef
16.
Zurück zum Zitat Wulf J, Haedinger U, Oppitz U, Thiele W, Mueller G, Flentje M. Stereotactic radiotherapy for primary lung cancer and pulmonary metastases: a noninvasive treatment approach in medically inoperable patients. Int J Radiat Oncol Biol Phys. 2004;60:186–96.CrossRef Wulf J, Haedinger U, Oppitz U, Thiele W, Mueller G, Flentje M. Stereotactic radiotherapy for primary lung cancer and pulmonary metastases: a noninvasive treatment approach in medically inoperable patients. Int J Radiat Oncol Biol Phys. 2004;60:186–96.CrossRef
17.
Zurück zum Zitat Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, et al. Hypofractionated stereotactic radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study. J Thorac Oncol. 2007;2:94–100.CrossRef Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, et al. Hypofractionated stereotactic radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study. J Thorac Oncol. 2007;2:94–100.CrossRef
18.
Zurück zum Zitat Uematsu M, Shioda A, Suda A, Fukui T, Ozeki Y, Hama Y, et al. Computed tomography-guided frameless stereotactic radiotherapy for stage I non-small cell lung cancer: a 5-year experience. Int J Radiat Oncol Biol Phys. 2001;51:666–70.CrossRef Uematsu M, Shioda A, Suda A, Fukui T, Ozeki Y, Hama Y, et al. Computed tomography-guided frameless stereotactic radiotherapy for stage I non-small cell lung cancer: a 5-year experience. Int J Radiat Oncol Biol Phys. 2001;51:666–70.CrossRef
19.
Zurück zum Zitat Onishi H, Kuriyama K, Komiyama T, Tanaka S, Sano N, Marino K, et al. Clinical outcomes of stereotactic radiotherapy for stage I non-small cell lung cancer using a novel irradiation technique: patient self-controlled breath-hold and beam switching using a combination of linear accelerator and CT scanner. Lung Cancer. 2004;45:45–55.CrossRef Onishi H, Kuriyama K, Komiyama T, Tanaka S, Sano N, Marino K, et al. Clinical outcomes of stereotactic radiotherapy for stage I non-small cell lung cancer using a novel irradiation technique: patient self-controlled breath-hold and beam switching using a combination of linear accelerator and CT scanner. Lung Cancer. 2004;45:45–55.CrossRef
20.
Zurück zum Zitat Firat S, Bousamra M, Gore E, Byhardt RW. Comorbidity and KPS are independent prognostic factors in stage I non-small-cell lung cancer. Int J Radiat Oncol Biol Phys. 2002;52:1047–57.CrossRef Firat S, Bousamra M, Gore E, Byhardt RW. Comorbidity and KPS are independent prognostic factors in stage I non-small-cell lung cancer. Int J Radiat Oncol Biol Phys. 2002;52:1047–57.CrossRef
21.
Zurück zum Zitat Timmermann 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.CrossRef Timmermann 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.CrossRef
22.
Zurück zum Zitat Paix A, Noel G, Falcoz PE, Levy P. Cost-effectiveness analysis of stereotactic body radiotherapy and surgery for medically operable early stage non small cell lung cancer. Radiother Oncol. 2018;128:534–40.CrossRef Paix A, Noel G, Falcoz PE, Levy P. Cost-effectiveness analysis of stereotactic body radiotherapy and surgery for medically operable early stage non small cell lung cancer. Radiother Oncol. 2018;128:534–40.CrossRef
23.
Zurück zum Zitat Eguchi T, Bains S, Lee MC, Tan KS, Hristov B, Buitrago D, et al. Impact of increasing age on cause-specific mortality and morbidity in patients with stage I non-small-cell lung Cancer: a competing risks analysis. J Clin Oncol. 2017;35:281–90.CrossRef Eguchi T, Bains S, Lee MC, Tan KS, Hristov B, Buitrago D, et al. Impact of increasing age on cause-specific mortality and morbidity in patients with stage I non-small-cell lung Cancer: a competing risks analysis. J Clin Oncol. 2017;35:281–90.CrossRef
24.
Zurück zum Zitat Boyer MJ, Williams CD, Harpole DH, Onaitis MW, Kelley MJ, Salama JK. Improved survival of stage I non-small cell lung Cancer: a VA central registry analysis. J Thorac Oncol. 2017;12:1814–23.CrossRef Boyer MJ, Williams CD, Harpole DH, Onaitis MW, Kelley MJ, Salama JK. Improved survival of stage I non-small cell lung Cancer: a VA central registry analysis. J Thorac Oncol. 2017;12:1814–23.CrossRef
25.
Zurück zum Zitat Bryant AK, Mundt RC, Sandhu AP, Urbanic JJ, Sharabi AB, Gupta S, et al. Stereotactic body radiation therapy versus surgery for early lung Cancer among US veterans. Ann Thorac Surg. 2018;105:425–31.CrossRef Bryant AK, Mundt RC, Sandhu AP, Urbanic JJ, Sharabi AB, Gupta S, et al. Stereotactic body radiation therapy versus surgery for early lung Cancer among US veterans. Ann Thorac Surg. 2018;105:425–31.CrossRef
26.
Zurück zum Zitat Hague W, Verma V, Polamraju P, Farach A, Butler EB, The BS. Stereotactic body radiation therapy versus conventionally fractionated radiation therapy for early stage non-small cell lung cancer. Radiother Oncol. 2018; [Epub ahead of print]. Hague W, Verma V, Polamraju P, Farach A, Butler EB, The BS. Stereotactic body radiation therapy versus conventionally fractionated radiation therapy for early stage non-small cell lung cancer. Radiother Oncol. 2018; [Epub ahead of print].
27.
Zurück zum Zitat Von Reibnitz SF, Wu AJ, Treharne GC, Dick-Godfrey R, Foster A, et al. Stereotactic body radiation therapy (SBRT) improves local control and overall survival compared to conventionally fractionated radiation for stage I non-small cell lung cancer (NSCLC). Actac Oncol. 2018; [Epub ahead of print]. Von Reibnitz SF, Wu AJ, Treharne GC, Dick-Godfrey R, Foster A, et al. Stereotactic body radiation therapy (SBRT) improves local control and overall survival compared to conventionally fractionated radiation for stage I non-small cell lung cancer (NSCLC). Actac Oncol. 2018; [Epub ahead of print].
28.
Zurück zum Zitat Kopek N, Paludan M, Petersen J, Hansen A, Grau C, Hoyer M. Co-morbidity index predicts for mortality after stereotactic body radiotherapy for medically inoperable early-stage non-small cell lung cancer. Radiother Oncol. 2009;93:402–7.CrossRef Kopek N, Paludan M, Petersen J, Hansen A, Grau C, Hoyer M. Co-morbidity index predicts for mortality after stereotactic body radiotherapy for medically inoperable early-stage non-small cell lung cancer. Radiother Oncol. 2009;93:402–7.CrossRef
29.
Zurück zum Zitat Baker R, Han G, Sarangkasiri S, DeMarco M, Turke C, Stevens CW, et al. Clinical and Dosimetric predictors of radiation pneumonitis in a large series of patients treated with stereotactic body radiation therapy to the lung. Int J Radiat Oncol Biol Phys. 2013;85:190–5.CrossRef Baker R, Han G, Sarangkasiri S, DeMarco M, Turke C, Stevens CW, et al. Clinical and Dosimetric predictors of radiation pneumonitis in a large series of patients treated with stereotactic body radiation therapy to the lung. Int J Radiat Oncol Biol Phys. 2013;85:190–5.CrossRef
30.
Zurück zum Zitat Zheng X, Schipper M, Kidwell K, Lin J, Reddy R, Ren Y, et al. Survival outcome after stereotactic body radiation therapy and surgery for stage I non-small cell lung cancer: a meta-analysis. Int J Radiat Oncol Biol Phys. 2014;90:603–11.CrossRef Zheng X, Schipper M, Kidwell K, Lin J, Reddy R, Ren Y, et al. Survival outcome after stereotactic body radiation therapy and surgery for stage I non-small cell lung cancer: a meta-analysis. Int J Radiat Oncol Biol Phys. 2014;90:603–11.CrossRef
31.
Zurück zum Zitat Extermann M, Overcash J, Lyman GH, Parr J, Balducci L. Comorbidity and functional status are independent in older cancer patients. J Clin Oncol. 1998;16:1582–7.CrossRef Extermann M, Overcash J, Lyman GH, Parr J, Balducci L. Comorbidity and functional status are independent in older cancer patients. J Clin Oncol. 1998;16:1582–7.CrossRef
32.
Zurück zum Zitat Shavelle RM, Paculdo DR, Kush SJ, Dm M, Strauss DJ. Life expectancy and years of life lost in chronic obstructive pulmonary disease: findings from the NHANES III follow-up study. Int J Chron Obstruct Pul Dis. 2009;4:137–48.CrossRef Shavelle RM, Paculdo DR, Kush SJ, Dm M, Strauss DJ. Life expectancy and years of life lost in chronic obstructive pulmonary disease: findings from the NHANES III follow-up study. Int J Chron Obstruct Pul Dis. 2009;4:137–48.CrossRef
33.
Zurück zum Zitat Suissa S, Dell’Aniello S, Ernst P. Long-term natural history of chronic obstructive pulmonary disease: severe exacerbations and mortality. Thorax. 2012;67:957–63.CrossRef Suissa S, Dell’Aniello S, Ernst P. Long-term natural history of chronic obstructive pulmonary disease: severe exacerbations and mortality. Thorax. 2012;67:957–63.CrossRef
34.
Zurück zum Zitat Kirkhus L, Jordhøy M, Šaltytė Benth J, Rostoft S, Selbaek G, Jensen Hjermstad M, et al. Comparing comorbidity scales: attending physician score versus the cumulative illness rating scale for geriatrics. J Geriatric Oncol. 2016;7:90–8.CrossRef Kirkhus L, Jordhøy M, Šaltytė Benth J, Rostoft S, Selbaek G, Jensen Hjermstad M, et al. Comparing comorbidity scales: attending physician score versus the cumulative illness rating scale for geriatrics. J Geriatric Oncol. 2016;7:90–8.CrossRef
35.
Zurück zum Zitat Grønberg BH, Sundstrøm S, Kaasa S, Bremnes RM, Flotten O, Amundsen T, et al. Influence of comorbidity on survival, toxicity and health-related quality of life in patients with advanced non-small-cell lung cancer receiving platinum-doublet chemotherapy. Eur J Cancer. 2010;46:2225–34.CrossRef Grønberg BH, Sundstrøm S, Kaasa S, Bremnes RM, Flotten O, Amundsen T, et al. Influence of comorbidity on survival, toxicity and health-related quality of life in patients with advanced non-small-cell lung cancer receiving platinum-doublet chemotherapy. Eur J Cancer. 2010;46:2225–34.CrossRef
36.
Zurück zum Zitat Yamashita H, Takahashi W, Haga A, Nakagawa K. Radiation pneumonitis after stereotactic radiation therapy for lung cancer. World J Radiol. 2014;6:708–15.CrossRef Yamashita H, Takahashi W, Haga A, Nakagawa K. Radiation pneumonitis after stereotactic radiation therapy for lung cancer. World J Radiol. 2014;6:708–15.CrossRef
37.
Zurück zum Zitat Chaudhuri AA, Binkley MS, Rigdon J, Carter JN, Aggarwal S, Dudley SA. (2016) pre-treatment non-target lung FDG-PET uptake predicts symptomatic radiation pneumonitis following stereotactic ablative radiotherapy (SABR). Radiother Oncol. 2016;119:454–60.CrossRef Chaudhuri AA, Binkley MS, Rigdon J, Carter JN, Aggarwal S, Dudley SA. (2016) pre-treatment non-target lung FDG-PET uptake predicts symptomatic radiation pneumonitis following stereotactic ablative radiotherapy (SABR). Radiother Oncol. 2016;119:454–60.CrossRef
38.
Zurück zum Zitat Wang J, Cao J, Ji W, Arenberg D, Dai J, Stanton P, et al. Poor baseline pulmonary function may not increase the risk of radiation-induced lung toxicity. Int J Radiat Oncol Biol Phys. 2013;85:798–804.CrossRef Wang J, Cao J, Ji W, Arenberg D, Dai J, Stanton P, et al. Poor baseline pulmonary function may not increase the risk of radiation-induced lung toxicity. Int J Radiat Oncol Biol Phys. 2013;85:798–804.CrossRef
39.
Zurück zum Zitat Takeda A, Kunieda E, Ohashi T, Aoki Y, Oku Y, Enomoto T, et al. Severe COPD is correlated with mild radiation pneumonitis following stereotactic body radiotherapy. Chest. 2012;141:858–66.CrossRef Takeda A, Kunieda E, Ohashi T, Aoki Y, Oku Y, Enomoto T, et al. Severe COPD is correlated with mild radiation pneumonitis following stereotactic body radiotherapy. Chest. 2012;141:858–66.CrossRef
40.
Zurück zum Zitat Schanne DH, Nestle U, Allgäuer M, Andratschke N, Appold S, Dieckmann U, et al. Stereotactic body radiotherapy for centrally located stage I NSCLC: a multicenter analysis. Strahlenther Onkol. 2015;191:125–32.CrossRef Schanne DH, Nestle U, Allgäuer M, Andratschke N, Appold S, Dieckmann U, et al. Stereotactic body radiotherapy for centrally located stage I NSCLC: a multicenter analysis. Strahlenther Onkol. 2015;191:125–32.CrossRef
41.
Zurück zum Zitat Moustakis C, Blanck O, Ebrahimi Tazehmahalleh F, Ka Heng Chan M, Ernst I, Krieger T, et al. planning benchmark study for SBRT of early stage NSCLC : results of the DEGRO working group stereotactic radiotherapy. Strahlenther Onkol. 2017;193:780–90.CrossRef Moustakis C, Blanck O, Ebrahimi Tazehmahalleh F, Ka Heng Chan M, Ernst I, Krieger T, et al. planning benchmark study for SBRT of early stage NSCLC : results of the DEGRO working group stereotactic radiotherapy. Strahlenther Onkol. 2017;193:780–90.CrossRef
Metadaten
Titel
Comorbidity indexing for prediction of the clinical outcome after stereotactic body radiation therapy in non-small cell lung cancer
verfasst von
Julia Dreyer
Michael Bremer
Christoph Henkenberens
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2018
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-018-1156-1

Weitere Artikel der Ausgabe 1/2018

Radiation Oncology 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.