Vol 27, No 6 (2022)
Research paper
Published online: 2022-10-31

open access

Page views 3398
Article views/downloads 407
Get Citation

Connect on Social Media

Connect on Social Media

Contralateral esophageal sparing technique in definitive radiotherapy for non-small cell lung cancer: dosimetric parameters and normal tissue complication probability modeling

Małgorzata Łazar-Poniatowska1, Joanna Kamińska21, Krzysztof Konopa1, Rafał Dziadziuszko1, Jacek Jassem1
Rep Pract Oncol Radiother 2022;27(6):933-942.

Abstract

Background: The purpose of this study was to assess the benefit of the contralateral esophageal sparing technique (CEST) in definitive radiotherapy of non–small cell lung cancer (NSCLC).

Materials and methods: We retrospectively reviewed radiation plans for 13 patients who underwent definitive chemoradiation for locally advanced NSCLC. Alternative plans were prepared with the use of CEST, with an additional margin of 5 mm from planning treatment volume (PTV). Normal tissue complication probability (NTCP) analyses for the esophagus and tumor control probability (TCP) for the PTV were performed for original and CEST plans using the equivalent uniform dose (EUD)-based mathematical model.

Results: In all cases, the CEST plan allowed for the reduction of esophageal dose, with a mean of 3.8 Gy (range, 0.7 to 8.7 Gy). The mean reductions of V40 and V60 to the esophagus were 6.4 Gy (range, 2.1 to 17.2 Gy) and 1.9 Gy (range, 3.4 to 10.0 Gy), respectively. There was no substantial decrease  in the maximal dose to the esophagus. Reduction of NTCP was achieved for all patients (range, 5–73%), and TCP was not affected (–1.8 to +6.7%).

Conclusions: The application of CEST in definitive radiotherapy of locally advanced NSCLC allows for reducing selected dosimetric parameters to the esophagus without compromising TCP.

research paper

Reports of Practical Oncology and Radiotherapy

2022, Volume 27, Number 6, pages: 933–942

DOI: 10.5603/RPOR.a2022.0110

Submitted: 10.07.2022

Accepted: 03.10.2022

© 2022 Greater Poland Cancer Centre.

Published by Via Medica.

All rights reserved.

e-ISSN 2083–4640

ISSN 1507–1367

Contralateral esophageal sparing technique in definitive radiotherapy for non-small cell lung cancer: dosimetric parameters and normal tissue complication probability modeling

Małgorzata Łazar-Poniatowska1Joanna Kamińska12Krzysztof Konopa1Rafał Dziadziuszko1Jacek Jassem1
1Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
2Institute of Experimental Physics, Faculty of Mathematics, Physics and Informatics, University of Gdansk, Gdansk, Poland

Address for correspondence: Małgorzata Łazar-Poniatowska, MD, Department of Oncology and Radiotherapy, Medical University of Gdańsk, 17 Smoluchowskiego St., 80–214 Gdańsk, Poland, tel: +48 58 584 45 60; e-mail: poniatowska@gumed.edu.pl

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially

Abstract
Background: The purpose of this study was to assess the benefit of the contralateral esophageal sparing technique (CEST) in definitive radiotherapy of non–small cell lung cancer (NSCLC).
Materials and methods: We retrospectively reviewed radiation plans for 13 patients who underwent definitive chemoradiation for locally advanced NSCLC. Alternative plans were prepared with the use of CEST, with an additional margin of 5 mm from planning treatment volume (PTV). Normal tissue complication probability (NTCP) analyses for the esophagus and tumor control probability (TCP) for the PTV were performed for original and CEST plans using the equivalent uniform dose (EUD)-based mathematical model.
Results: In all cases, the CEST plan allowed for the reduction of esophageal dose, with a mean of 3.8 Gy (range, 0.7 to 8.7 Gy). The mean reductions of V40 and V60 to the esophagus were 6.4 Gy (range, 2.1 to 17.2 Gy) and 1.9 Gy (range, 3.4 to 10.0 Gy), respectively. There was no substantial decrease in the maximal dose to the esophagus. Reduction of NTCP was achieved for all patients (range, 5–73%), and TCP was not affected (–1.8 to +6.7%).
Conclusions: The application of CEST in definitive radiotherapy of locally advanced NSCLC allows for reducing selected dosimetric parameters to the esophagus without compromising TCP.
Key words: lung cancer; chemoradiation; esophageal sparing; dosimetric parameters; TCP; NTCP
Rep Pract Oncol Radiother 2022;27(6):933–942

Introduction

The objective of radiotherapy is to achieve maximal tumor control probability (TCP) with minimal normal tissue complication probability (NTCP). The relation between TCP and NTCP constitutes
a therapeutic window. Higher doses, higher radiation volumes, and the addition of systemic treatments result in increases in both values [1].

Several randomized clinical trials in locally advanced non–small cell lung cancer (NSCLC) have demonstrated that concurrent chemotherapy and radiotherapy result in better outcomes than radiotherapy alone or sequential use of both modalities [2–6], but at the cost of a higher incidence of acute esophagitis (4% vs. 18%) [4]. In the Radiation Therapy Oncology Group (RTOG) 0617 studies, the incidence of grade 3 esophagitis with concurrent chemoradiation in the standard dose arm was substantially lower (7%) than previously described, likely because of recent technical improvements in radiotherapy [7]. However, esophagitis remains an essential issue because resulting radiotherapy breaks may negatively affect long-term survival [8], and subsequent esophageal strictures decrease quality of life [9].

One of the approaches to decreasing esophageal toxicity is the contralateral esophageal sparing technique (CEST) in which the esophageal wall contralateral to gross disease is considered an avoidance structure [10]. The purpose of this modeling study was to explore whether this approach affects dosimetric parameters and esophageal NTCP in NSCLC patients administered definitive chemoradiation.

Materials and methods

We retrospectively reviewed radiation plans for patients with locally advanced NSCLC who underwent definitive chemoradiation. Radiotherapy plans were designed using four-dimensional (4D) computed tomography (CT) or breath-hold CT. For 4D-CT, the internal target volume (ITV) was defined as the gross tumor volume (GTV) plus the internal margin for respiratory tumor motion across the 10 phases of the breathing cycle. For the breath-hold technique, only GTV was contoured. The original clinical target volume was based on the automatic isotropic 5 mm margin around the ITV or GTV. The organs at risk (OARs) were defined according to the RTOG atlas and included the right and left lungs, combined lung volumes, spinal canal, entire esophagus, heart and, in selected cases, brachial plexus. Original plans were designed using the subvolume “esophagus PTV”. In the CEST plans, the contralateral esophagus (CE) was contoured as a separate avoidance structure 5 mm from the PTV edge, allowing for an additional margin to achieve steep dose fall-off across sections of the esophagus. Superior and inferior borders of CE were 2 cm around the PTV (Fig. 1). There was no lower limit of the CE size.

%c5%81azar-Poniatowska-1.png
Figure 1. Example of contralateral esophagus delineation. Structures displayed are: planning treatment volume (red), esophagus (blue), and contralateral esophagus (yellow)

The prescribed dose was 60–66 Gy in 30–33 fractions delivered with 6 MV photons and the intensity-modulated radiation therapy (IMRT) technique. The alternative CEST plans were prepared without compromising target coverage and respecting dose constraints to OARs. Efforts were made to minimize the dose to CE. CEST plans were prepared using the same physical parameters that is, the same radiation technique, the same number of beams, the same energy, and the same AAA 13.026 calculation model. For all patients, original and CEST plans were compared according to prescribed dosimetric parameters. The Dmean, V40, V60, and maximal dose (Dmax) to the esophagus, as well as the V95, V98, V107, and Dmax for PTV, were extracted from the histograms prepared in the treatment planning system (Eclipse, Aria 13.0, Varian Medical Systems). The NTCP analyses for the esophagus and TCP for the PTV were performed for original and CEST plans using an equivalent uniform dose (EUD)-based mathematical model provided by MATLAB modules [11]. This model can be used for both tumors and normal tissues according to the Niemierko formula [12, 13]:

155056.png

In this formula, {vi, Di} are bins of a histogram and a is a tissue-specific parameter. This parameter is negative for tumors and positive for normal structures. Parameter i is unitless and describes the i-th partial volume receiving the dose Di in Gray (Gy). The choice of parameter a determines the behavior of the EUD-based model. The EUD-based NTCP was calculated using the logistic function [13]:

155071.png

The tolerance dose 50 (TD50) is the 50% complication rate at a specific time interval, and γ50 is the normalized dose response gradient. The γ50 expressed as %/% is the increment in response in percentage points for a 1% increase in dose, defined at the 50% response level. To calculate TCP, the EUD was substituted in the following equation:

155065.png

The TCD50 is the dose to control 50% of the homogeneously irradiated tumor. Parameters for TCP and NTCP (Tab. 1) were selected based on the proposal of Emami et al. Esophageal toxicity in this report included severe complications such as critical stricture and perforation [14].

Table 1. Radiobiological parameters used in modeling of normal tissue control probability (NTCP) and tumor control probability (TCP)

NTCP

TCP

Parameter a

19

–12

Parameter γ50

4

2

TD50 (Gy)/TCD50(Gy)

68

60

α/β (Gy)

3

10

Results

We analyzed the treatment plans of 13 patients with locally advanced nonmetastatic NSCLC (12 primary tumors and 1 mediastinal relapse) and gross tumor within 1 cm of the esophagus, who underwent definitive sequential or concurrent chemoradiation (Tab. 2). The CEST planning showed no deterioration in parameters for PTV (Tab. 3). The mean esophageal dose with CEST was lower in all patients, with a mean reduction of 3.8 Gy (range, 0.7 to 8.7 Gy). The mean reduction of V40 and V60 to the esophagus was 6.4 Gy (range, 2.1 to 17.2 Gy) and 1.9 Gy (range, –3,4 to +10.0 Gy), respectively (Fig. 2). However, in no case was substantial reduction of the maximal dose to the esophagus achieved (Tab. 4). The delta parameter, defined as the percentage difference between the maximum dose in the CEST and original plan, varied from –1.5% (reduction) to +2.5% (increase).

Table 2. Patient characteristics

Patient

Histology

cTNM

Stage

Type of chemoradiation

Chemotherapy regimen

Total radiation dose [Gy] (dose levels)

1

Squamous cell carcinoma

cT4N2

IIIB

Concurrent

2 × CP/EP

66

2

Adenocarcinoma

cT1N2

IIIA

Concurrent

2 × CP/EP

66

3

Adenocarcinoma

cT2N2

IIIA

Concurrent

2 × CP/EP

66/60

4

Adenocarcinoma

cT3N2

IIIA

Sequential

3 × CP/GEM

60

5

Squamous cell carcinoma

cT3N2

IIIA

Sequential

4 × CP/VB

66

6

Adenocarcinoma

Mediastinal relapse

Radiotherapy alone

60

7

Squamous cell carcinoma

cT3N1

IIIA

Concurrent

1 × CP/EP

66

8

Squamous cell carcinoma

cT3N2

IIIA

Concurrent

2 × CP/EP

60

9

Squamous cell carcinoma

cT2N2

IIIA

Concurrent

2 × CP/EP

66/54

10

Adenocarcinoma

cT3N2

IIIA

Sequential

3 × CP/GEM

66

11

Squamous cell carcinoma

cT4N2

IIIB

Sequential

4 × CP/VB

62

12

Adenocarcinoma

cT4N2

IIIB

Concurrent

2 × CP/PEM

60

13

NSCLC-NOS

cT4N2

IIIB

Concurrent

3 × CBDCA/PCL

60

Table 3. Dosimetric planning target volume (PTV) parameters for original and contralateral esophageal sparing technique (CEST) plans

Patient

Plan

PTV

V95 (%)

Δ (%)

Dmax (%)

Δ (%)

1

Original

95.2

–0.1%

108.3

0.3%

CEST

95.1

108.6

2

Original

98.3

0.1%

105.7

0.0%

CEST

98.5

105.7

3

Original

79.8

0.7%

110.0

–0.9%

CEST

80.3

109.0

4

Original

98.7

–0.7%

107.3

2.5%

CEST

98.0

110.0

5

Original

98.6

–2.0%

106.9

2.0%

CEST

96.7

109.0

6

Original

99.6

0,0%

105.0

0.5%

CEST

99.6

105.5

7

Original

99.3

–0.8%

106.7

0.3%

CEST

98.5

107.0

8

Original

98.5

–0.2%

107.5

0.3%

CEST

98.2

107.8

9

Original

94.5

–0.5%

107.0

0.0%

CEST

94.0

107.0

10

Original

97.3

–0.3%

105.9

0.1%

CEST

97.0

106.0

11

Original

97.4

1.2%

106.3

–1.5%

CEST

98.6

104.7

12

Original

96.6

–1.1%

105.5

2.4%

CEST

95.6

108.0

13

Original

100.0

–0.2%

105.0

1.0%

CEST

99.8

106.0

SD

0.8%

1.2 %

%c5%81azar-Poniatowska-2-html.png
Figure 2. Comparison of esophageal parameters for original (ORG) and contralateral esophageal sparing technique (CEST) plans
Table 4. Dosimetric parameters for esophagus for original and contralateral esophageal sparing technique (CEST) plans

Patient

Plan

Esophagus

Dmean

Δ [Gy]

Dmax

Δ [Gy]

1

Original

30.5

–4.55

67.5

0.64

CEST

25.9

68.1

2

Original

23.5

–8.65

66.5

0.68

CEST

14.8

67.2

3

Original

26.0

–2.82

72.2

0.23

CEST

23.2

72.5

4

Original

29.1

–3.21

60.4

0.54

CEST

25.9

61

5

Original

31.4

–2.34

68.6

0.14

CEST

29.0

68.7

6

Original

28.9

–3.02

56.3

0.28

CEST

25.9

56.6

7

Original

24.7

–4.06

68

0.43

CEST

20.7

68.4

8

Original

27.9

–5.88

59.6

0.05

CEST

22.1

59.7

9

Original

17.6

–0.66

68.6

–0.06

CEST

17.0

68.6

10

Original

30.6

–4.07

64.8

0.58

CEST

26.5

65.3

11

Original

26.1

–2.43

63.9

0.98

CEST

23.7

64.9

12

Original

35.2

–5.10

63

–0.15

CEST

30.1

62.8

13

Original

33.0

–2.26

65.3

–0.8

CEST

30.7

64.5

SD

2.01

0.46

The differences in the mean heart dose, mean lung dose, and maximal spinal cord dose were negligible for all patients (Tab. 5). Figure 3 shows an example of the dose-volume histogram of original and CEST plans for a single patient. For the CEST plan, the mean heart dose and the mean lung dose were comparable with the original plans. The reduction of NTCP was reached in all patients (range, 5%–73%), and the TCP was not affected (–1.8% to +6.7%; Fig. 4 and 5).

Table 5. Dosimetric parameters for selected organs at risk for original and contralateral esophageal sparing technique (CEST) plans

Patient

Plan

Heart

Lungs

Spinal cord

Dmean [Gy]

Δ [Gy]

Dmean

Δ [Gy]

Dmax

Δ [Gy]

1

Original

12.7

–0.59

20.1

0.27

45.4

1.33

CEST

12.2

20.4

46.7

2

Original

7.0

–0.41

18.3

–0.21

50.1

0.12

CEST

6.5

18.1

50.2

3

Original

3.8

0.04

17.4

48.4

0.70

CEST

3.9

17.4

49.1

4

Original

17.9

0.7

18.8

–0.14

44.7

0.42

CEST

18.6

18.7

45.2

5

Original

3.5

1.18

17.6

–0.13

47.7

0.07

CEST

4.6

17.4

47.7

6

Original

17.1

–0.09

16.7

–0.15

45.3

0.57

CEST

17.0

16.6

45.8

7

Original

3.2

0.33

15.6

0.05

47.1

1.55

CEST

3.5

15.7

48.6

8

Original

11.6

–0.09

19.1

–0.1

45

2.39

CEST

11.5

19.0

47.4

9

Original

6.3

14.7

–0.05

37.3

–0.20

CEST

6.3

14.6

37.1

10

Original

13.0

0.15

19.1

–0.07

48.1

0.23

CEST

13.1

19.0

48.3

11

Original

38.0

0.98

12.9

–0.14

45.6

–0.45

CEST

39.0

12.8

45.2

12

Original

13.8

1.20

18.9

40.5

2.40

CEST

15.0

18.9

42.9

13

Original

22.5

–0.63

19.3

0.20

14.2

0.60

CEST

21.9

19.5

14.8

SD

0.63

0.14

0.91

%c5%81azar-Poniatowska-3.png
Figure 3. Example of dose-volume histogram for original (squares) and contralateral esophageal sparing technique (CEST) plan (triangle lines) for a single patient. Structures displayed are planning target volume (PTV) (red), esophagus (blue), lungs (violet), and heart (pink)

155299.png
Figure 4. Normal tissue complication probability (NTCP) comparison for the original and contralateral esophageal sparing technique (CEST) plans
155332.png
Figure 5. Tumor control probability (TCP) comparison for the original and contralateral esophageal sparing technique (CEST) plans

Discussion

Acute esophagitis accompanying combined chemoradiotherapy for lung cancer remains a significant clinical problem. Pharmacologic prevention of esophagitis has proved unsuccessful [15]. Crucial elements of reducing the risk of esophagitis include the thorough definition of target volumes and dosimetric parameters and improvements in the planning technique and dose delivery.

In 1991, Emami et al. published normal tissue constraints for use in radiotherapy practice [14]. With a particular emphasis on partial volume effects, tolerance doses were proposed for irradiation of one-third, two-thirds, or whole organs. Further studies involving large patient populations and using three-dimensional dosimetry was the basis of subsequent Quantitative Analysis of Normal Tissue Effects which defines volumetric parameters predictive for radiation esophagitis [16]. Recommended dosimetric constraints included V35, V50, and V70 for grade2 esophagitis and the median dose (MD) for grade3 toxicity. The MD has been widely adopted as a parameter; however, its predictive value for esophagitis in particular studies has been inconsistent [17–20].

The advent of IMRT allowed for the reduction of the esophagus dose while maintaining adequate target coverage. In addition, interfraction motion does not compromise the quality of treatment plans [21]. IMRT appears to be particularly beneficial in node-positive patients and in cases with target volumes located close to the esophagus. While meeting all normal-tissue constraints in node-positive patients, IMRT can deliver RT doses that are 25%–30% greater compared with three-dimensional radiotherapy [22].

The primary objective of the present study was to use NTCP and TCP modeling to evaluate whether CEST potentially allows for decreasing esophageal toxicity compared with conventional treatment.

A prospective CEST trial showed promising results, with no incidence of grade3 esophagitis [10]. The authors suggested the following CE dose constraints: maximum dose 60 Gy (to 0.03 cc),
V55 < 0.5 cc, and V45 < 2.5 cc, with acceptable deviations being 63 Gy, 3 cc, and 7.5 cc, respectively. In another phase 1 nonrandomized clinical trial including 27 participants, the CE-sparing technique was associated with reduced risk of esophagitis among patients treated uniformly with chemoradiation (to 70 Gy) [23]. There was no grade 3 or higher esophagitis despite the tumor being located within 1 cm of the esophagus. The 2-year progression-free survival and overall survival rates were 57% [95% confidence interval (CI): 33–75%] and 67% (95% CI: 45–82%), respectively.

In the 1980s, probabilistic radiobiology models introduced concepts of serial and parallel tissue organization and functional subunits [24]. According to this concept, the esophagus is an organ with functional subunits arranged in the longitudinal axis and high-dose irradiation of the entire cross-section of the esophagus can result in whole organ dysfunction. The CEST concept suggests that avoiding high radiation doses to the entire cross-section of the esophagus may lead to more efficient regeneration of the esophageal mucosa. It has been also speculated that preserving the contralateral esophageal wall converts this organ from a serial to parallel one [23].

In our study, there was no substantial reduction of the maximal esophageal dose. Evaluation of this aspect revealed that D max reduction would affect parameters of the PTV coverage with prescribed dose, thus being unacceptable. On the other hand, the increase in the maximal dose was elevating the NTCP, regardless of the obtained improvement in volumetric parameters such as mean dose, V40, V60. It can be explained by the nature of the assumptions used in the modeling formulas strictly related to paradigm of esophagus as a serial organ [10, 25].

The limitations of our study are a relatively small number of radiation treatment plans, the shortcomings of the NTCP modeling and its assumptions, and lack of testing of internal and external model validity. Nevertheless, our results show that CEST, with its additional margin (5 mm from PTV), allows for the reduction of selected dosimetric parameters to the esophagus in radiotherapy of NSCLC. The usefulness of CEST in reducing esophageal toxicity should be assessed in prospective clinical trials.

Conflict of interests

None declared

Funding

Study was supported by a statutory grant 02-0023/07/171 of the Medical University of Gdańsk, Poland

Data sharing statement

The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials.

References

  1. Chang DS, Lasley FD, Das IJ, Mendonca MS, Dynlacht JR. Therapeutic ratio. In: Chang DS. ed. Basic Radiotherapy Physics and Biology. Springer International Publishing 2014: 265–275.
  2. Curran WJ, Paulus R, Langer CJ, et al. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst. 2011; 103(19): 1452–1460, doi: 10.1093/jnci/djr325, indexed in Pubmed: 21903745.
  3. Furuse K, Fukuoka M, Kawahara M, et al. Phase III study of concurrent versus sequential thoracic radiotherapy in combination with mitomycin, vindesine, and cisplatin in unresectable stage III non-small-cell lung cancer. J Clin Oncol. 1999; 17(9): 2692–2699, doi: 10.1200/JCO.1999.17.9.2692, indexed in Pubmed: 10561343.
  4. Aupérin A, Le Péchoux C, Rolland E, et al. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol. 2010; 28(13): 2181–2190, doi: 10.1200/JCO.2009.26.2543, indexed in Pubmed: 20351327.
  5. Le Chevalier T, Arriagada R, Quoix E, et al. Radiotherapy alone versus combined chemotherapy and radiotherapy in nonresectable non-small-cell lung cancer: first analysis of a randomized trial in 353 patients. J Natl Cancer Inst. 1991; 83(6): 417–423, doi: 10.1093/jnci/83.6.417, indexed in Pubmed: 1847977.
  6. Jeremic B, Shibamoto Y, Acimovic L, et al. Hyperfractionated radiation therapy with or without concurrent low-dose daily carboplatin/etoposide for stage III non-small-cell lung cancer: a randomized study. J Clin Oncol. 1996; 14(4): 1065–1070, doi: 10.1200/JCO.1996.14.4.1065, indexed in Pubmed: 8648358.
  7. Bradley JD, Paulus R, Komaki R, et al. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): a randomised, two-by-two factorial phase 3 study. Lancet Oncol. 2015; 16(2): 187–199, doi: 10.1016/S1470-2045(14)71207-0, indexed in Pubmed: 25601342.
  8. Cox JD, Pajak TF, Asbell S, et al. Interruptions of high-dose radiation therapy decrease long-term survival of favorable patients with unresectable non-small cell carcinoma of the lung: analysis of 1244 cases from 3 Radiation Therapy Oncology Group (RTOG) trials. Int J Radiat Oncol Biol Phys. 1993; 27(3): 493–498, doi: 10.1016/0360-3016(93)90371-2, indexed in Pubmed: 8226140.
  9. Jiang ZQ, Yang K, Komaki R, et al. Long-term clinical outcome of intensity-modulated radiotherapy for inoperable non-small cell lung cancer: the MD Anderson experience. Int J Radiat Oncol Biol Phys. 2012; 83(1): 332–339, doi: 10.1016/j.ijrobp.2011.06.1963, indexed in Pubmed: 22079735.
  10. Al-Halabi H, Paetzold P, Sharp GC, et al. A Contralateral Esophagus-Sparing Technique to Limit Severe Esophagitis Associated With Concurrent High-Dose Radiation and Chemotherapy in Patients With Thoracic Malignancies. Int J Radiat Oncol Biol Phys. 2015; 92(4): 803–810, doi: 10.1016/j.ijrobp.2015.03.018, indexed in Pubmed: 26104934.
  11. Warkentin B, Stavrev P, Stavreva N, et al. A TCP-NTCP estimation module using DVHs and known radiobiological models and parameter sets. J Appl Clin Med Phys. 2004; 5(1): 50–63, doi: 10.1120/jacmp.v5i1.1970, indexed in Pubmed: 15753933.
  12. Niemierko A. A generalized concept of equivalent uniform dose (EUD). In: Proceedings of the 41st AAPM Annual Meeting; 1999. Med Phys 1999;26:1100 [abstract].
  13. Niemierko A. A unified model of tissue response to radiation. In: Proceedings of the 41st AAPM Annual Meeting; 1999. Med Phys; 1999;26:1100 [abstract].
  14. Emami B, Lyman J, Brown A, et al. Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 1991; 21(1): 109–122, doi: 10.1016/0360-3016(91)90171-y, indexed in Pubmed: 2032882.
  15. Lawrence Y, Paulus R, Langer C, et al. The addition of amifostine to carboplatin and paclitaxel based chemoradiation in locally advanced non-small cell lung cancer: Long-term follow-up of Radiation Therapy Oncology Group (RTOG) randomized trial 9801. Lung Cancer. 2013; 80(3): 298–305, doi: 10.1016/j.lungcan.2013.02.008, indexed in Pubmed: 23477890.
  16. Bentzen SM, Constine LS, Deasy JO, et al. Quantitative Analyses of Normal Tissue Effects in the Clinic (QUANTEC): an introduction to the scientific issues. Int J Radiat Oncol Biol Phys. 2010; 76(3 Suppl): S3–S9, doi: 10.1016/j.ijrobp.2009.09.040, indexed in Pubmed: 20171515.
  17. Singh AK, Lockett MA, Bradley JD. Predictors of radiation-induced esophageal toxicity in patients with non-small-cell lung cancer treated with three-dimensional conformal radiotherapy. Int J Radiat Oncol Biol Phys. 2003; 55(2): 337–341, doi: 10.1016/s0360-3016(02)03937-8, indexed in Pubmed: 12527046.
  18. Qiao WB, Zhao YH, Zhao YB, et al. Clinical and dosimetric factors of radiation-induced esophageal injury: radiation-induced esophageal toxicity. World J Gastroenterol. 2005; 11(17): 2626–2629, doi: 10.3748/wjg.v11.i17.2626, indexed in Pubmed: 15849822.
  19. Palma DA, Senan S, Oberije C, et al. Predicting esophagitis after chemoradiation therapy for non-small cell lung cancer: an individual patient data meta-analysis. Int J Radiat Oncol Biol Phys. 2013; 87(4): 690–696, doi: 10.1016/j.ijrobp.2013.07.029, indexed in Pubmed: 24035329.
  20. Werner-Wasik M, Pequignot E, Leeper D, et al. Predictors of severe esophagitis include use of concurrent chemotherapy, but not the length of irradiated esophagus: a multivariate analysis of patients with lung cancer treated with nonoperative therapy. Int J Radiat Oncol Biol Phys. 2000; 48(3): 689–696, doi: 10.1016/s0360-3016(00)00699-4, indexed in Pubmed: 11020565.
  21. Niedzielski J, Bluett JB, Williamson RT, et al. Analysis of esophageal-sparing treatment plans for patients with high-grade esophagitis. J Appl Clin Med Phys. 2013; 14(4): 4248, doi: 10.1120/jacmp.v14i4.4248, indexed in Pubmed: 23835390.
  22. Shaitelman SF, Vicini FA, Grills IS, et al. Potential for reduced toxicity and dose escalation in the treatment of inoperable non-small-cell lung cancer: a comparison of intensity-modulated radiation therapy (IMRT), 3D conformal radiation, and elective nodal irradiation. Int J Radiat Oncol Biol Phys. 2003; 57(3): 875–890, doi: 10.1016/s0360-3016(03)00743-0, indexed in Pubmed: 14529795.
  23. Kamran SC, Yeap BY, Ulysse CA, et al. Assessment of a Contralateral Esophagus-Sparing Technique in Locally Advanced Lung Cancer Treated With High-Dose Chemoradiation: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol. 2021; 7(6): 910–914, doi: 10.1001/jamaoncol.2021.0281, indexed in Pubmed: 33830168.
  24. Schultheiss TE, Orton CG, Peck RA. Models in radiotherapy: volume effects. Med Phys. 1983; 10(4): 410–415, doi: 10.1118/1.595312, indexed in Pubmed: 6888354.
  25. Withers HR, Taylor J, Maciejewski B. Treatment volume and tissue tolerance. Int J Radiat Oncol Biol Phys. 1988; 14(4): 751–759, doi: 10.1016/0360-3016(88)90098-3, indexed in Pubmed: 3350731.



Reports of Practical Oncology and Radiotherapy