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

Open Access 01.12.2018 | Research

Automated VMAT planning for postoperative adjuvant treatment of advanced gastric cancer

verfasst von: Abdul Wahab M. Sharfo, Florian Stieler, Oskar Kupfer, Ben J. M. Heijmen, Maarten L. P. Dirkx, Sebastiaan Breedveld, Frederik Wenz, Frank Lohr, Judit Boda-Heggemann, Daniel Buergy

Erschienen in: Radiation Oncology | Ausgabe 1/2018

Abstract

Background

Postoperative/adjuvant radiotherapy of advanced gastric cancer involves a large planning target volume (PTV) with multi-concave shapes which presents a challenge for volumetric modulated arc therapy (VMAT) planning. This study investigates the advantages of automated VMAT planning for this site compared to manual VMAT planning by expert planners.

Methods

For 20 gastric cancer patients in the postoperative/adjuvant setting, dual-arc VMAT plans were generated using fully automated multi-criterial treatment planning (autoVMAT), and compared to manually generated VMAT plans (manVMAT). Both automated and manual plans were created to deliver a median dose of 45 Gy to the PTV using identical planning and segmentation parameters. Plans were evaluated by two expert radiation oncologists for clinical acceptability. AutoVMAT and manVMAT plans were also compared based on dose-volume histogram (DVH) and predicted normal tissue complication probability (NTCP) analysis.

Results

Both manVMAT and autoVMAT plans were considered clinically acceptable. Target coverage was similar (manVMAT: 96.6 ± 1.6%, autoVMAT: 97.4 ± 1.0%, p = 0.085). With autoVMAT, median kidney dose was reduced on average by > 25%; (for left kidney from 11.3 ± 2.1 Gy to 8.9 ± 3.5 Gy (p = 0.002); for right kidney from 9.2 ± 2.2 Gy to 6.1 ± 1.3 Gy (p <  0.001)). Median dose to the liver was lower as well (18.8 ± 2.3 Gy vs. 17.1 ± 3.6 Gy, p = 0.048). In addition, Dmax of the spinal cord was significantly reduced (38.3 ± 3.7 Gy vs. 31.6 ± 2.6 Gy, p <  0.001). Substantial improvements in dose conformity and integral dose were achieved with autoVMAT plans (4.2% and 9.1%, respectively; p <  0.001). Due to the better OAR sparing in the autoVMAT plans compared to manVMAT plans, the predicted NTCPs for the left and right kidney and the liver-PTV were significantly reduced by 11.3%, 12.8%, 7%, respectively (p ≤ 0.001). Delivery time and total number of monitor units were increased in autoVMAT plans (from 168 ± 19 s to 207 ± 26 s, p = 0.006) and (from 781 ± 168 MU to 1001 ± 134 MU, p = 0.003), respectively.

Conclusions

For postoperative/adjuvant radiotherapy of advanced gastric cancer, involving a complex target shape, automated VMAT planning is feasible and can substantially reduce the dose to the kidneys and the liver, without compromising the target dose delivery.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13014-018-1032-z) contains supplementary material, which is available to authorized users.
Abkürzungen
autoVMAT
Fully automated volumetric modulated arc therapy (plans)
CI
Conformity index
CRT
Chemoradiotherapy
DVH
Dose-volume histogram
EUD
Equivalent uniform dose
HI
Homogeneity index
IMRT
Intensity-modulated radiation therapy
INT-0116
Intergroup Study 0116
LTCP
Logarithmic tumor control probability
manVMAT
Manually generated volumetric modulated arc therapy (plans)
MU
Monitor units
NTCP
Normal tissue complication probability
OAR
Organ at risk
PTV
Planning target volume
RTOG
Radiation Therapy Oncology Group
TPS
Treatment planning system
TV
Target volume
VMAT
Volumetric modulated arc therapy

Background

In the Intergroup Study 0116 (INT-0116) study, adjuvant chemoradiotherapy (CRT) improved overall survival and progression-free survival in patients with gastric cancer compared to surgery alone [1, 2]. The benefit of combined modality treatment was later confirmed in Asian studies with D2-resected patients [35]. European trials in gastroesophageal junction cancers showed survival benefits compared to surgery with neoadjuvant CRT [6] or perioperative chemotherapy without radiotherapy [7, 8]. Randomized trials comparing adjuvant CRT to (perioperative) chemotherapy indicated clinical benefits for CRT in subgroups, but failed in showing a consistent benefit in European and Asian trials [3, 5, 9, 10]. Therefore, the role of adjuvant CRT in gastric carcinoma compared to other approaches, such as perioperative chemotherapy, is not yet clearly defined [9]. The use of modern radiotherapy approaches might improve the risk−/benefit ratio in favor of radiotherapy in gastric carcinoma. Recent data showed the feasibility of using intensity-modulated radiation therapy (IMRT) or volumetric modulated arc therapy (VMAT) for this purpose [3, 1012]. Postoperative radiotherapy of advanced gastric cancer involves a large target volume with multi-concave shapes which presents a challenge for VMAT planning with a risk of protocol deviations. In a similar setting, radiotherapy for esophageal carcinoma indicated that the experience of the treatment planner may largely affect plan quality, as shown by large differences in plan quality among planners within one institute [13]. In such a setting, automated treatment planning might have several advantages, including facilitated central review in clinical trials as well as improvement of less experienced treatment planner’s performance.
Over the years, in-house developed as well as commercial algorithms have attempted to automate the trial-and-error process in order to create optimal plans, reduce user variability and improve the quality and efficiency of the resulting plans. Knowledge-based planning uses a model library of previously generated plans to predict new treatment plan parameters [14, 15], while multi-criterial optimization generates a set of Pareto-optimal plans [16, 17]. Erasmus-iCycle is an optimizer for fully automated multi-criterial beam profile optimization and beam angle selection for coplanar and non-coplanar IMRT, developed at the Erasmus MC Cancer Institute [1822]. In combination with the Monaco treatment planning system (TPS), Erasmus-iCycle is currently used in clinical practice for IMRT and VMAT plan generation for prostate, head and neck, cervical and lung cancer patients [1922]. Several studies on postoperative gastric cancer patients have dosimetrically evaluated different radiotherapy techniques [2326]. To our knowledge, no study has been published considering the feasibility and advantages of automated treatment planning for this treatment site. In this study, we investigated to what extent automated treatment planning using Erasmus-iCycle results in improved VMAT plan quality for advanced gastric cancer patients compared to plan generation by an expert planner.

Methods

Patients

A total of 20 patients with advanced gastric cancer who received radiotherapy treatment were included in the study. Clinical details are shown in Additional file 1: Table S1. Written informed consent was obtained from all patients for anonymized usage of treatment planning data. Our study protocol for retrospective evaluation of automated planning using Erasmus-iCycle was approved by the ethics committee of Heidelberg University, Medical Faculty Mannheim (2016-806R-MA).

Treatment plan generation

All manually and automatically generated dual-arc VMAT plans (manVMAT and autoVMAT, respectively) were prescribed to deliver a median dose of 45 Gy to the target in 25 fractions. The primary planning objective was achieving adequate PTV coverage while maximally sparing the organs at risk. All plans were generated for delivery at a VersaHD linear accelerator (Elekta AB, Stockholm, Sweden), equipped with an Agility multi-leaf collimator and a photon beam energy of 10 MV.
All manVMAT plans were generated by expert treatment planners with the Monaco TPS, version 5.11 (Elekta AB, Stockholm, Sweden) using template-based optimization cost-functions. The employed optimization template is set according to the current clinical guidelines used at the Medical University of Mannheim. For each patient, the optimization cost-functions’ parameters were iteratively tweaked to improve the dose distribution and achieve optimal target coverage while respecting OAR constraints. As explained below, also the final autoVMAT plans were generated with Monaco, for which we used the same software version. All Monte Carlo dose calculations in Monaco were performed using a 1% dose variance, and a dose grid resolution of 3 mm. A maximum of 140 control points per arc was allowed. Also the other segmentation settings were kept identical for manual and automated plan generation.

Automated multi-criterial VMAT plan generation with Erasmus-iCycle/Monaco

The in-house developed Erasmus-iCycle/Monaco platform for fully automated multi-criteria plan generation [1822] was configured to generate clinically deliverable VMAT plans for gastric cancer. For each of the study patients, Erasmus-iCycle was used to first automatically generate a Pareto-optimal plan with clinically favorable trade-offs between treatment objectives using a “wish-list” developed for gastric cancer (as detailed below in Table 1). Based on this Erasmus-iCycle plan, a patient-specific Monaco template was then created fully automatically, to be used in the Monaco TPS for automated generation of a deliverable dual-arc VMAT plan that mimicked the initial Erasmus-iCycle plan. The applied wish-list contains constraints to be strictly fulfilled, and clinical plan objectives with ascribed priorities to be met as closely as possible or superseded (see Table 1 for details). Three constraints were used to control the maximum dose in the PTV and the patient (i.e., the outline of the patient’s external surface including the PTV, delineated OARs and the unspecified tissues), as well as the dose conformity outside the PTV. In order to achieve a homogeneous and adequate PTV dose coverage, the highest priority objective was given to the PTV using the Logarithmic Tumor Control Probability function (LTCP) [27], followed by a shell around the PTV to ensure a steep gradient outside the PTV (priority 2). In line with the clinical practice, minimizing the mean dose in the kidneys was the organ-at-risk (OAR) objective with highest priority (priority 3), followed by the mean dose in the liver (priority 4). Subsequently, equivalent uniform dose (EUD) objectives with volume effect parameters (k = 6 and 12) [28], focusing on reduction of the midrange and high dose in the heart and spinal cord were used (priorities 5 and 6), respectively. To control the dose conformity and entrance dose, a shell at 18 mm from the PTV, as well as a skin ring of 21 mm wide from the body contour towards the patient’s internal were defined (priority 7). Additionally, dose-volume objectives for the kidneys, liver, and the lungs were used with lower priorities.
Table 1
Applied wish-list for automatic VMAT plan generation for gastric cancer patients
Constraints
 Volume
Type
Limit
  PTV
Maximum
105% of D p
  PTV Shell 39 mm
Maximum
50% of D p
  Patient
Maximum
105% of D p
Objectives
 Priority
Volume
Type
Goal
Parameters
  1
PTV
↓ LTCP
0.4
D p =45 Gy,α = 4
  2
PTV Shell 3 mm
↓ Maximum
90% of D p
 
  3
Left and Right Kidney
↓ Mean
8 Gy
 
  4
Liver
↓ Mean
15 Gy
 
  5
Heart
↓ EUD
15 Gy
k = 6
  6
Spinal Cord
↓ EUD
25 Gy
k = 12
  7
PTV Shell 18 mm
↓ Maximum
40% of D p
 
Skin Ring
↓ Maximum
25% of D p
21 mm
  8
Left and Right Kidney
↓ Volume-Dose
25%
12 Gy
Liver
↓ Volume-Dose
30%
24 Gy
  9
Left and Right Lung
↓ Volume-Dose
50%
20 Gy
Abbreviations: D p prescribed dose, LTCP Logarithmic Tumor Control Probability, α cell sensitivity parameter to achieve adequate target coverage, EUD Equivalent Uniform Dose, k volume parameter

Plan evaluation and comparison

All plans were evaluated by expert radiation oncologists (FL, JBH, and DB) for clinical acceptability. For fair dosimetric plan comparisons, all manVMAT and autoVMAT plans were first normalized to obtain equal median PTV dose (i.e., D50% = 45 Gy). In accordance with the International Commission on Radiation Units and Measurements Report No. 83, near-minimum and near-maximum doses (D98% and D2%, respectively) in the PTV were evaluated, from which the homogeneity index (HI = (D2% - D98%)/D50%) was computed. Additionally, dose conformity was estimated by calculating the conformity index (CI = (TVRI)2/(TV*VRI)), i.e., ratio of the target volume covered by the reference isodose level (TVRI) to the target volume (TV) and volume of the reference isodose (VRI). Quantitative analyses of OAR doses included Dmean, D30%, D50%, D60% in the kidneys and the liver-PTV, mean and maximum doses (Dmean, and Dmax) in the spinal cord and the heart, volumes of the kidneys receiving more than 12 Gy (V12Gy) and 20 Gy (V20Gy) [29], volumes of the liver-PTV receiving more than 24 Gy (V24Gy) and 30 Gy (V30Gy), and Dmean in the lungs and volumes of the lungs receiving more than 5 Gy (V5Gy) and 20 Gy (V20Gy) [30]. In addition, integral patient doses were evaluated by assessing patient Dmean and volumes receiving V5Gy, V11.25Gy and V22.5Gy. Estimated treatment delivery time and total number of monitor units (MUs) for each plan were also quantified.
From the DVHs, normal tissue complication probabilities (NTCPs) for the kidneys and the liver were estimated using the Lyman-Burman-Kutcher NTCP model for late effects, considering tolerance dose TD50/5 = 12 Gy, n = 0.70, m = 0.26 for the kidneys [29], and TD50/5 = 30 Gy, n = 0.32, m = 0.15 for liver failure [31], where TD50/5 refers to the dose to the whole organ which lead to complication in 50% of the population at 5 years, m relates to the steepness of the dose-response curve, and n represents the volume effect in the LKB model. For this purpose, all plans were first normalized to 1.5 Gy per fraction using α/β = 2.5 Gy.
Differences in dosimetric parameters between manVMAT and autoVMAT plans were analyzed using SPSS software v.21 (SPSS, Inc., Chicago, USA) and presented as the mean ± 1 standard deviation. Paired two-sided Wilcoxon Signed-rank tests were performed to assess statistical significance of observed differences, considering p <  0.05 statistically significant.

Results

Target volume dosimetric evaluations

All automatically and manually generated VMAT plans were clinically acceptable and achieved adequate target coverage. Clinical acceptability of the plans was evaluated by radiation oncologists with experience in gastric cancer treatment (JBH, FL and DB). Differences between the autoVMAT and manVMAT plans in V95% or D98% were not statistically significant (Table 2). However, autoVMAT plans exhibited a significantly lower near-maximum dose in the PTV, resulting in a better target dose homogeneity (0.09 ± 0.01 vs. 0.10 ± 0.02 (p = 0.003)). For autoVMAT, the dose conformity was significantly improved as well (0.91 ± 0.02 vs. 0.88 ± 0.03 (p <  0.001)). As an example, Fig. 1a shows the resulting dose distributions from the autoVMAT and manVMAT plans for patient 8. As is evident from this figure and the corresponding dose-volume histograms in Fig. 1b, AutoVMAT resulted in favorable dose conformity and better OAR sparing.
Table 2
Comparison of dosimetric parameters between autoVMAT and manVMAT plans. Population mean values for the 20 study patients and corresponding standard deviations are reported
Structure
Parameter
autoVMAT
manVMAT – autoVMAT
Mean ± SD
Mean diff. ± SD
p-value
PTV
V95% (%)
97.4 ± 1.0
- 0.8 ± 1.7
0.085
D98% (Gy)
42.5 ± 0.5
- 0.2 ± 0.7
0.14
D2% (Gy)
46.3 ± 0.1
0.5 ± 0.2
<  0.001
HI
0.09 ± 0.01
0.02 ± 0.02
0.003
CI
0.91 ± 0.02
- 0.04 ± 0.03
<  0.001
Left Kidney
Dmean (Gy)
12.7 ± 4.3
1.8 ± 1.6
0.001
D30% (Gy)
14.9 ± 8.1
2.1 ± 3.3
0.013
D50% (Gy)
8.9 ± 3.5
2.4 ± 2.2
0.002
D60% (Gy)
7.3 ± 1.9
2.4 ± 1.4
<  0.001
V12Gy (%)
32.2 ± 14.0
11.7 ± 7.7
<  0.001
V20Gy (%)
19.2 ± 13.2
1.0 ± 3.9
0.117
NTCP (%)
45.1 ± 35.8
11.3 ± 10.9
0.001
Right Kidney
Dmean (Gy)
7.5 ± 1.7
3.0 ± 1.2
<  0.001
D30% (Gy)
7.8 ± 1.7
4.1 ± 1.6
<  0.001
D50% (Gy)
6.1 ± 1.3
3.1 ± 1.3
<  0.001
D60% (Gy)
5.5 ± 1.2
2.7 ± 1.1
<  0.001
V12Gy (%)
12.4 ± 4.8
18.9 ± 8.4
<  0.001
V20Gy (%)
3.8 ± 2.8
3.0 ± 2.8
0.001
NTCP (%)
4.2 ± 3.8
12.8 ± 7.2
<  0.001
Liver - PTV
Dmean (Gy)
18.6 ± 3.3
1.8 ± 2.8
0.02
D30% (Gy)
21.9 ± 4.1
2.8 ± 3.2
0.003
D50% (Gy)
17.1 ± 3.6
1.7 ± 3.3
0.048
D60% (Gy)
15.1 ± 3.4
1.2 ± 3.3
0.14
V24Gy (%)
25.4 ± 11.6
7.1 ± 8.8
0.004
V30Gy (%)
12.9 ± 6.0
4.7 ± 4.6
0.001
NTCP (%)
31.1 ± 23.2
7.0 ± 7.0
0.001
Heart
Dmean (Gy)
12.7 ± 4.7
0.7 ± 1.8
0.117
Dmax (Gy)
46.8 ± 0.6
0.8 ± 1.2
0.009
Spinal cord
Dmean (Gy)
13.9 ± 2.6
2.9 ± 1.5
<  0.001
Dmax (Gy)
31.6 ± 2.6
6.7 ± 3.2
<  0.001
Left Lung
Dmean (Gy)
7.3 ± 3.2
0.6 ± 0.3
<  0.001
V5Gy (%)
32.7 ± 15.6
1.4 ± 1.0
<  0.001
V20Gy (%)
14.0 ± 7.2
2.2 ± 1.8
<  0.001
Right Lung
Dmean (Gy)
5.4 ± 3.3
0.3 ± 0.5
0.013
V5Gy (%)
31.2 ± 16.8
1.1 ± 2.1
0.01
V20Gy (%)
7.0 ± 7.6
0.8 ± 1.8
0.078
Patient
Dmean (Gy)
9.4 ± 1.9
0.5 ± 0.3
<  0.001
V5Gy (%)
40.2 ± 8.9
1.6 ± 0.8
<  0.001
V11.25Gy (%)
30.0 ± 6.6
1.8 ± 1.7
0.001
V22.5Gy (%)
16.2 ± 3.3
1.5 ± 1.1
<  0.001
Abbreviations: HI homogeneity index, CI conformity index, NTCP normal tissue complication probability

Organs at risk dosimetric evaluations

Figure 2 shows the observed absolute differences in dosimetric parameters for each of the study patients. Overall, autoVMAT plans had more favorable dose distributions, resulting in reduced dose delivery to the kidneys, liver, spinal cord, heart and lungs, without deteriorating the PTV dose coverage (Table 2 and Fig. 2). Median doses (D50%) to the left kidney, right kidney, and liver-PTV were significantly reduced by 28% (from 11.3 ± 2.1 Gy to 8.9 ± 3.5 Gy, p = 0.002), 39% (from 9.2 ± 2.2 Gy to 6.1 ± 1.3 Gy, p <  0.001), and 11% (from 18.8 ± 2.3 Gy to 17.1 ± 3.6 Gy, p = 0.048), respectively. The V20Gy for the left and right kidney with autoVMAT were lower than those for the manVMAT plans (Table 2). In addition, the V30Gy for the liver-PTV was significantly decreased with autoVMAT plans by 36% (from 17.7 ± 6.2% to 12.9 ± 6.0%, p = 0.001). The maximum dose in the spinal cord was on average reduced by 6.7 ± 3.2 Gy (p <  0.001) with autoVMAT. Furthermore, the integral dose in the patient and the dose conformity were also significantly better with autoVMAT (Table 2).

NTCPs evaluations

The published sets of parameters used for calculations of NTCPs for the kidneys and liver-PTV resulted in overall lower probability of late complications with autoVMAT compared to manVMAT plans. In 16/20 patients, the predicted NTCPs were lowest with autoVMAT for the left kidney, whereas all 20 patients had lower predicted NTCPs with autoVMAT for the right kidney (Fig. 3). For 17/20 patients, the NTCP predictions for liver-PTV were lower for the autoVMAT plans. The average NTCP values resulting from autoVMAT, given in Table 2, were significantly reduced by 11.3%, 12.8% and 7% for the left and right kidney and the liver-PTV, respectively.

Planning and treatment delivery times

All manVMAT plans in this study were generated using template-based manual planning. Hands-on tweaking time of the optimization cost functions was on average 30 min (range 5-60 min). The optimization and dose calculation of autoVMAT plans in Erasmus-iCycle/Monaco was fully automated and therefore did not require any hands-on time.
Compared to manVMAT plans a significantly more MUs were required for delivery of the autoVMAT plans (1001 ± 134 MU vs. 781 ± 168 MU, p = 0.003), resulting in a prolonged (estimated) treatment delivery time (207 ± 26 s vs. 168 ± 19 s, p = 0.006).

Discussion

Gastric cancer is the third leading cause of cancer death in both sexes with yearly 723,000 deaths worldwide [32]. Protocol deviations can reduce clinical efficacy of radiotherapy in complex treatment geometries, like gastric carcinoma. In the centralized review of the INT-0116 study, minor or major protocol deviations were observed in 35% of patients [2]. Even after correction of errors, major protocol deviations were still found in 6.5% of the plans that were actually irradiated [1, 2]. The clinical impact of protocol deviations in INT-0116 has not been reported. However, in other complex geometries it was shown that protocol violations during radiation therapy were correlated with reduced overall survival [33]. Modern radiotherapy approaches such as IMRT and VMAT have several dosimetric advantages but they are associated with increased complexity [9, 3436]. Very high protocol deviations are therefore possible; for example, RTOG 0529 showed 81% protocol deviations at first plan review [37]. Albeit in small numbers, RTOG 0022 showed local failure rates of 50% (2/4) vs. 6% (3/49) in oropharyngeal cancer patients treated with and without protocol deviations, respectively [38]. Protocol deviations may, amongst other things, be caused by inaccurate delineation or sub-optimal treatment planning. For gastric cancer, the percentage of these deviations is unknown. In a recent report on RTOG 0933 by Gondi et al. [39], unacceptable radiotherapy protocol deviations were observed in 25% of cases at a rapid review process: 11/21 cases had contouring deviations, 5/21 cases had unacceptable planning deviations and 5/21 had unacceptable deviations of contouring and planning. Also in a recent study of Habraken et al. [40] on hepatocellular carcinoma patients, it was demonstrated that pretreatment plan review was important to reduce unacceptable protocol deviations. Additionally, they demonstrated that automated treatment planning could be used to identify sub-optimal treatment plans. This might be especially relevant in trials in which low numbers of patients are eligible or small numbers of patients are treated in a participating hospital.
Over the past years, several studies have investigated the advantages of automated treatment planning compared to manual planning using in-house developed or commercial algorithms [1822, 4143]. Treatment planning with Erasmus-iCycle/Monaco is fully automated, and it has been successfully validated for clinical use in several treatment sites including prostate [19], head-and-neck [20], cervix [21], and lung [22]. Knowledge-based planning using a model library of previously generated plans to predict treatment plan parameters for a new patient was configured and tested in pelvic anatomy [41], lung [42], and esophageal cancer [43].
Numerous dosimetric studied have evaluated different radiotherapy techniques for gastric cancer by comparing IMRT, VMAT, helical tomotherapy, 3DCRT and proton therapy [2326]. Until now, no study has been published showing the possibility/advantages of automated treatment planning for postoperative gastric cancer patients. In this study Erasmus-iCycle was used to automatically generate VMAT plans for gastric cancer patients. For this site, manual treatment optimization is a large challenge, even for an expert planner, due to the multi-concave shape, the extent of the target volume, and the close proximity to many radiosensitive organs (i.e., kidneys, liver, heart, and spinal cord). Another difficulty in postoperative radiotherapy in gastric cancer patients is that radiation tolerance doses of OARs are relatively low. Compared to the manVMAT plans, generated by an expert planner, plan quality was significantly better for the autoVMAT plans, only requiring slightly longer treatment delivery times. Specifically, dose conformity and sparing of organs at risk were improved, while the clinical importance of the observed longer treatment delivery time and the increased MUs is considered to be low. As a result of higher modulation, the number of MUs was significantly higher in the autoVMAT than in manVMAT plans, this could lead to challenges in radiation delivery. The integral dose was significantly reduced in autoVMAT plans, this may be beneficial specially for young patients to avoid secondary tumors, and most notably showed significantly reduced NTCP for liver and both kidneys, although it is not yet clear to what extent the observed dosimetric advantages of autoVMAT vs. an experienced planner will really translate into a clinical benefit. Automated treatment planning can improve the efficiency of the treatment planning process and reduce its user dependency, this in turns might lead to more consistent and uniform outcomes in treatment planning studies and clinical trials. Apart from the improved plan quality and reduced predicted complications, automated planning also eliminated the planning hands-on time required for planning.

Conclusion

Automated treatment planning is of great value for complex treatment sites like for gastric cancer. Compared to manual planning by an expert planner, plan quality could be largely improved, while drastically reducing treatment planning workload.

Funding

This work was in part funded by a research grant of Elekta AB, Stockholm, Sweden.

Availability of data and materials

The dataset generated and analyzed during the current study are available from the corresponding author on reasonable request.
The study was approved by the ethics committee of Heidelberg University, Medical Faculty Mannheim (2016-806R-MA). Written informed consent for scientific usage of clinical data was obtained from all patients.
Consent for scientific usage of clinical data was obtained from all patients included in the study.

Competing interests

Erasmus MC Cancer Institute has research collaborations with Accuray Inc., Sunnyvale, USA.
AWMS reports grants from Elekta AB, Stockholm, Sweden, during the conduct of the study; FS reports personal fees from Elekta AB, Sweden, outside the submitted work; OK has nothing to disclose; BJMH reports grants from Elekta AB, Stockholm, Sweden, during the conduct of the study; grants from Accuray Inc., Sunnyvale, USA, outside the submitted work; MLPD reports grants from Elekta AB, Stockholm, Sweden, during the conduct of the study; SB reports grants from Elekta AB, Stockholm, Sweden, during the conduct of the study; grants from Accuray Inc., Sunnyvale, USA, outside the submitted work; FW reports research grants and travel support from Elekta, during the conduct of the study; FL has nothing to disclose; JBH reports teaching honoraria from Elekta AB, Stockholm, Sweden, during the conduct of the study; DB reports personal fees from Siemens AG, personal fees from NB Capital ApS, outside the submitted work.

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 Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed
2.
Zurück zum Zitat Smalley SR, Benedetti JK, Haller DG, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.CrossRefPubMedPubMedCentral Smalley SR, Benedetti JK, Haller DG, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Zhu WG, Xua DF, Pu J, et al. A randomized, controlled, multicenter study comparing intensity-modulated radiotherapy plus concurrent chemotherapy with chemotherapy alone in gastric cancer patients with D2 resection. Radiother Oncol. 2012;104:361–6.CrossRefPubMed Zhu WG, Xua DF, Pu J, et al. A randomized, controlled, multicenter study comparing intensity-modulated radiotherapy plus concurrent chemotherapy with chemotherapy alone in gastric cancer patients with D2 resection. Radiother Oncol. 2012;104:361–6.CrossRefPubMed
4.
Zurück zum Zitat Lee J, Lim DH, Kim S, et al. Phase III trial comparing capecitabine plus cisplatin versus capecitabine plus cisplatin with concurrent capecitabine radiotherapy in completely resected gastric cancer with D2 lymph node dissection: the ARTIST trial. J Clin Oncol. 2012;30:268–73.CrossRefPubMed Lee J, Lim DH, Kim S, et al. Phase III trial comparing capecitabine plus cisplatin versus capecitabine plus cisplatin with concurrent capecitabine radiotherapy in completely resected gastric cancer with D2 lymph node dissection: the ARTIST trial. J Clin Oncol. 2012;30:268–73.CrossRefPubMed
5.
Zurück zum Zitat Park SH, Sohn TS, Lee J, et al. Phase III trial to compare adjuvant chemotherapy with Capecitabine and cisplatin versus concurrent chemoradiotherapy in gastric Cancer: final report of the adjuvant chemoradiotherapy in stomach tumors trial, including survival and subset analyses. J Clin Oncol. 2015;33:3130–6.CrossRefPubMed Park SH, Sohn TS, Lee J, et al. Phase III trial to compare adjuvant chemotherapy with Capecitabine and cisplatin versus concurrent chemoradiotherapy in gastric Cancer: final report of the adjuvant chemoradiotherapy in stomach tumors trial, including survival and subset analyses. J Clin Oncol. 2015;33:3130–6.CrossRefPubMed
6.
Zurück zum Zitat van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366:2074–84.CrossRefPubMed van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366:2074–84.CrossRefPubMed
7.
Zurück zum Zitat Ychou M, Boige V, Pignon JP, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed Ychou M, Boige V, Pignon JP, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed
8.
Zurück zum Zitat Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed
9.
Zurück zum Zitat Buergy D, Lohr F, Baack T, et al. Radiotherapy for tumors of the stomach and gastroesophageal junction--a review of its role in multimodal therapy. Radiat Oncol. 2012;7:192.CrossRefPubMedPubMedCentral Buergy D, Lohr F, Baack T, et al. Radiotherapy for tumors of the stomach and gastroesophageal junction--a review of its role in multimodal therapy. Radiat Oncol. 2012;7:192.CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat Boda-Heggemann J, Weiss C, Schneider V, et al. Adjuvant IMRT/XELOX radiochemotherapy improves long-term overall- and disease-free survival in advanced gastric cancer. Strahlenther Onkol. 2013;189:417–23.CrossRefPubMed Boda-Heggemann J, Weiss C, Schneider V, et al. Adjuvant IMRT/XELOX radiochemotherapy improves long-term overall- and disease-free survival in advanced gastric cancer. Strahlenther Onkol. 2013;189:417–23.CrossRefPubMed
12.
Zurück zum Zitat Dikken JL, van Sandick JW, Maurits Swellengrebel HA, et al. Neo-adjuvant chemotherapy followed by surgery and chemotherapy or by surgery and chemoradiotherapy for patients with resectable gastric cancer (CRITICS). BMC Cancer. 2011;11:329.CrossRefPubMedPubMedCentral Dikken JL, van Sandick JW, Maurits Swellengrebel HA, et al. Neo-adjuvant chemotherapy followed by surgery and chemotherapy or by surgery and chemoradiotherapy for patients with resectable gastric cancer (CRITICS). BMC Cancer. 2011;11:329.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Berry SL, Boczkowski A, Ma R, Mechalakos J, Hunt M. Interobserver variability in radiation therapy plan output: results of a single-institution study. Pract Radiat Oncol. 2016;6:442–9.CrossRefPubMedPubMedCentral Berry SL, Boczkowski A, Ma R, Mechalakos J, Hunt M. Interobserver variability in radiation therapy plan output: results of a single-institution study. Pract Radiat Oncol. 2016;6:442–9.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Appenzoller LM, Michalski JM, Thorstad WL, Mutic S, Moore KL. Predicting dose-volume histograms for organs-at-risk in IMRT planning. Med Phys. 2012;39:7446–61.CrossRefPubMed Appenzoller LM, Michalski JM, Thorstad WL, Mutic S, Moore KL. Predicting dose-volume histograms for organs-at-risk in IMRT planning. Med Phys. 2012;39:7446–61.CrossRefPubMed
15.
Zurück zum Zitat Yuan L, Ge Y, Lee WR, Yin FF, Kirkpatrick JP, Wu QJ. Quantitative analysis of the factors which affect the interpatient organ-at-risk dose sparing variation in IMRT plans. Med Phys. 2012;39:6868–78.CrossRefPubMed Yuan L, Ge Y, Lee WR, Yin FF, Kirkpatrick JP, Wu QJ. Quantitative analysis of the factors which affect the interpatient organ-at-risk dose sparing variation in IMRT plans. Med Phys. 2012;39:6868–78.CrossRefPubMed
16.
Zurück zum Zitat Monz M, Kufer KH, Bortfeld TR, Thieke C. Pareto navigation: algorithmic foundation of interactive multi-criteria IMRT planning. Phys Med Biol. 2008;53:985–98.CrossRefPubMed Monz M, Kufer KH, Bortfeld TR, Thieke C. Pareto navigation: algorithmic foundation of interactive multi-criteria IMRT planning. Phys Med Biol. 2008;53:985–98.CrossRefPubMed
17.
Zurück zum Zitat Breedveld S, Storchi PR, Keijzer M, Heemink AW, Heijmen BJM. A novel approach to multi-criteria inverse planning for IMRT. Phys Med Biol. 2007;52:6339–53.CrossRefPubMed Breedveld S, Storchi PR, Keijzer M, Heemink AW, Heijmen BJM. A novel approach to multi-criteria inverse planning for IMRT. Phys Med Biol. 2007;52:6339–53.CrossRefPubMed
18.
Zurück zum Zitat Breedveld S, Storchi PRM, Voet PWJ, Heijmen BJM. Icycle: integrated, multicriterial beam angle, and profile optimization for generation of coplanar and noncoplanar IMRT plans. Med Phys. 2012;39:951–63.CrossRefPubMed Breedveld S, Storchi PRM, Voet PWJ, Heijmen BJM. Icycle: integrated, multicriterial beam angle, and profile optimization for generation of coplanar and noncoplanar IMRT plans. Med Phys. 2012;39:951–63.CrossRefPubMed
19.
Zurück zum Zitat PWJ V, MLP D, Breedveld S, Al-Mamgani A, Incrocci L, BJM H. Fully automated volumetric modulated arc therapy plan generation for prostate cancer patients. Int J Radiat Oncol Biol Phys. 2014;88:1175–9.CrossRef PWJ V, MLP D, Breedveld S, Al-Mamgani A, Incrocci L, BJM H. Fully automated volumetric modulated arc therapy plan generation for prostate cancer patients. Int J Radiat Oncol Biol Phys. 2014;88:1175–9.CrossRef
20.
Zurück zum Zitat Voet PWJ, Dirkx MLP, Breedveld S, Fransen D, Levendag PC, Heijmen BJM. Toward fully automated multicriterial plan generation: a prospective clinical study. Int J Radiat Oncol Biol Phys. 2013;85:866–72.CrossRefPubMed Voet PWJ, Dirkx MLP, Breedveld S, Fransen D, Levendag PC, Heijmen BJM. Toward fully automated multicriterial plan generation: a prospective clinical study. Int J Radiat Oncol Biol Phys. 2013;85:866–72.CrossRefPubMed
21.
Zurück zum Zitat Sharfo AWM, Breedveld S, Voet PWJ, et al. Validation of fully automated VMAT plan generation for library-based plan-of-the-day cervical cancer radiotherapy. PLoS One. 2016;11:e0169202.CrossRefPubMedPubMedCentral Sharfo AWM, Breedveld S, Voet PWJ, et al. Validation of fully automated VMAT plan generation for library-based plan-of-the-day cervical cancer radiotherapy. PLoS One. 2016;11:e0169202.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Della Gala G, Dirkx ML, Hoekstra N, et al. Fully automated VMAT treatment planning for advanced-stage NSCLC patients. Strahlenther Onkol. 2017;193:402–9.CrossRefPubMedPubMedCentral Della Gala G, Dirkx ML, Hoekstra N, et al. Fully automated VMAT treatment planning for advanced-stage NSCLC patients. Strahlenther Onkol. 2017;193:402–9.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Li Z, Zeng J, Wang Z, Zhu H, Wei Y. Dosimetric comparison of intensity modulated and volumetric arc radiation therapy for gastric cancer. Oncol Lett. 2014;8:1427–34.CrossRefPubMedPubMedCentral Li Z, Zeng J, Wang Z, Zhu H, Wei Y. Dosimetric comparison of intensity modulated and volumetric arc radiation therapy for gastric cancer. Oncol Lett. 2014;8:1427–34.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Zhang T, Liang ZW, Han J, Bi JP, Yang ZY, Ma H. Double-arc volumetric modulated therapy improves dose distribution compared to static gantry IMRT and 3D conformal radiotherapy for adjuvant therapy of gastric cancer. Radiat Oncol. 2015;19:114.CrossRef Zhang T, Liang ZW, Han J, Bi JP, Yang ZY, Ma H. Double-arc volumetric modulated therapy improves dose distribution compared to static gantry IMRT and 3D conformal radiotherapy for adjuvant therapy of gastric cancer. Radiat Oncol. 2015;19:114.CrossRef
25.
Zurück zum Zitat Onal C, Dolek Y, Akkus YB. Dosimetric comparison of 3-dimensional conformal radiotherapy, volumetric modulated arc therapy, and helical tomotherapy for postoperative gastric cancer patients. Jpn J Radiol. 2018;36:30–9.CrossRefPubMed Onal C, Dolek Y, Akkus YB. Dosimetric comparison of 3-dimensional conformal radiotherapy, volumetric modulated arc therapy, and helical tomotherapy for postoperative gastric cancer patients. Jpn J Radiol. 2018;36:30–9.CrossRefPubMed
26.
Zurück zum Zitat Mondlane G, Gubanski M, Lind PA, Ureba A, Siegbahn A. Comparison of gastric-cancer radiotherapy performed with volumetric modulated arc therapy or single-field uniform-dose proton therapy. Acta Oncol. 2017;56:832–8.CrossRefPubMed Mondlane G, Gubanski M, Lind PA, Ureba A, Siegbahn A. Comparison of gastric-cancer radiotherapy performed with volumetric modulated arc therapy or single-field uniform-dose proton therapy. Acta Oncol. 2017;56:832–8.CrossRefPubMed
27.
Zurück zum Zitat Alber M, Reemtsen R. Intensity modulated radiotherapy treatment planning by use of a barrier-penalty multiplier method. Opt Methods Softw. 2007;22:391–411.CrossRef Alber M, Reemtsen R. Intensity modulated radiotherapy treatment planning by use of a barrier-penalty multiplier method. Opt Methods Softw. 2007;22:391–411.CrossRef
28.
Zurück zum Zitat Niemierko A. Reporting and analyzing dose distributions: a concept of equivalent uniform dose. Med Phys. 1997;24:103–10.CrossRefPubMed Niemierko A. Reporting and analyzing dose distributions: a concept of equivalent uniform dose. Med Phys. 1997;24:103–10.CrossRefPubMed
29.
Zurück zum Zitat Dawson LA, Kavanagh BD, Paulino AC, et al. Radiation-associated kidney injury. Int J Radiat Oncol Biol Phys. 2010;76:S108–15.CrossRefPubMed Dawson LA, Kavanagh BD, Paulino AC, et al. Radiation-associated kidney injury. Int J Radiat Oncol Biol Phys. 2010;76:S108–15.CrossRefPubMed
31.
Zurück zum Zitat Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. Int J Radiat Oncol Biol Phys. 2002;53:810–21.CrossRefPubMed Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. Int J Radiat Oncol Biol Phys. 2002;53:810–21.CrossRefPubMed
32.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed
33.
Zurück zum Zitat Wong KK, All S, Waxer J, et al. Radiotherapy after high-dose chemotherapy with autologous hematopoietic cell rescue: quality assessment of head start III. Pediatr Blood Cancer. 2017;64:e26529.CrossRef Wong KK, All S, Waxer J, et al. Radiotherapy after high-dose chemotherapy with autologous hematopoietic cell rescue: quality assessment of head start III. Pediatr Blood Cancer. 2017;64:e26529.CrossRef
34.
Zurück zum Zitat Wieland P, Dobler B, Mai S, et al. IMRT for postoperative treatment of gastric cancer: covering large target volumes in the upper abdomen: a comparison of a step-and-shoot and an arc therapy approach. Int J Radiat Oncol Biol Phys. 2004;59:1236–44.CrossRefPubMed Wieland P, Dobler B, Mai S, et al. IMRT for postoperative treatment of gastric cancer: covering large target volumes in the upper abdomen: a comparison of a step-and-shoot and an arc therapy approach. Int J Radiat Oncol Biol Phys. 2004;59:1236–44.CrossRefPubMed
35.
Zurück zum Zitat Lohr F, Dobler B, Mai S, et al. Optimization of dose distributions for adjuvant locoregional radiotherapy of gastric cancer by IMRT. Strahlenther Onkol. 2003;179:557–63.CrossRefPubMed Lohr F, Dobler B, Mai S, et al. Optimization of dose distributions for adjuvant locoregional radiotherapy of gastric cancer by IMRT. Strahlenther Onkol. 2003;179:557–63.CrossRefPubMed
36.
Zurück zum Zitat Haneder S, Michaely HJ, Schoenberg SO, et al. Assessment of renal function after conformal radiotherapy and intensity-modulated radiotherapy by functional 1H-MRI and 23Na-MRI. Strahlenther Onkol. 2012;188:1146–54.CrossRefPubMed Haneder S, Michaely HJ, Schoenberg SO, et al. Assessment of renal function after conformal radiotherapy and intensity-modulated radiotherapy by functional 1H-MRI and 23Na-MRI. Strahlenther Onkol. 2012;188:1146–54.CrossRefPubMed
37.
Zurück zum Zitat Kachnic LA, Winter K, Myerson RJ, et al. RTOG 0529: a phase 2 evaluation of dose-painted intensity modulated radiation therapy in combination with 5-fluorouracil and mitomycin-C for the reduction of acute morbidity in carcinoma of the anal canal. Int J Radiat Oncol Biol Phys. 2013;86:27–33.CrossRefPubMed Kachnic LA, Winter K, Myerson RJ, et al. RTOG 0529: a phase 2 evaluation of dose-painted intensity modulated radiation therapy in combination with 5-fluorouracil and mitomycin-C for the reduction of acute morbidity in carcinoma of the anal canal. Int J Radiat Oncol Biol Phys. 2013;86:27–33.CrossRefPubMed
38.
Zurück zum Zitat Eisbruch A, Harris J, Garden AS, et al. Multi-institutional trial of accelerated hypofractionated intensity-modulated radiation therapy for early-stage oropharyngeal cancer (RTOG 00-22). Int J Radiat Oncol Biol Phys. 2010;76:1333–8.CrossRefPubMed Eisbruch A, Harris J, Garden AS, et al. Multi-institutional trial of accelerated hypofractionated intensity-modulated radiation therapy for early-stage oropharyngeal cancer (RTOG 00-22). Int J Radiat Oncol Biol Phys. 2010;76:1333–8.CrossRefPubMed
39.
Zurück zum Zitat Gondi V, Cui Y, Mehta MP, et al. Real-time pretreatment review limits unacceptable deviations on a cooperative group radiation therapy technique trial: quality assurance results of RTOG 0933. Int J Radiat Oncol Biol Phys. 2015;91:564–70.CrossRefPubMedPubMedCentral Gondi V, Cui Y, Mehta MP, et al. Real-time pretreatment review limits unacceptable deviations on a cooperative group radiation therapy technique trial: quality assurance results of RTOG 0933. Int J Radiat Oncol Biol Phys. 2015;91:564–70.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Habraken SM, Sharfo AW, Buijsen J, et al. The TRENDY multi-center randomized trial on hepatocellular carcinoma – trial QA including automated treatment planning and benchmark-case results. Radiother Oncol. 2017;125:507–13.CrossRefPubMed Habraken SM, Sharfo AW, Buijsen J, et al. The TRENDY multi-center randomized trial on hepatocellular carcinoma – trial QA including automated treatment planning and benchmark-case results. Radiother Oncol. 2017;125:507–13.CrossRefPubMed
41.
Zurück zum Zitat Hussein M, South CP, Barry MA, et al. Clinical validation and benchmarking of knowledge-based IMRT and VMAT treatment planning in pelvic anatomy. Radiother Oncol. 2016;120:473–9.CrossRefPubMed Hussein M, South CP, Barry MA, et al. Clinical validation and benchmarking of knowledge-based IMRT and VMAT treatment planning in pelvic anatomy. Radiother Oncol. 2016;120:473–9.CrossRefPubMed
42.
Zurück zum Zitat Fogliata A, Belosi F, Clivio A, et al. On the pre-clinical validation of a commercial model-based optimization engine: application to volumetric modulated arc therapy for patients with lung or prostate cancer. Radiother Oncol. 2014;113:385–91.CrossRefPubMed Fogliata A, Belosi F, Clivio A, et al. On the pre-clinical validation of a commercial model-based optimization engine: application to volumetric modulated arc therapy for patients with lung or prostate cancer. Radiother Oncol. 2014;113:385–91.CrossRefPubMed
43.
Zurück zum Zitat Fogliata A, Nicolini G, Clivio A, et al. A broad scope knowledge based model for optimization of VMAT in esophageal cancer: validation and assessment of plan quality among different treatment centers. Radiat Oncol. 2015;10:220.CrossRefPubMedPubMedCentral Fogliata A, Nicolini G, Clivio A, et al. A broad scope knowledge based model for optimization of VMAT in esophageal cancer: validation and assessment of plan quality among different treatment centers. Radiat Oncol. 2015;10:220.CrossRefPubMedPubMedCentral
Metadaten
Titel
Automated VMAT planning for postoperative adjuvant treatment of advanced gastric cancer
verfasst von
Abdul Wahab M. Sharfo
Florian Stieler
Oskar Kupfer
Ben J. M. Heijmen
Maarten L. P. Dirkx
Sebastiaan Breedveld
Frederik Wenz
Frank Lohr
Judit Boda-Heggemann
Daniel Buergy
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2018
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-018-1032-z

Weitere Artikel der Ausgabe 1/2018

Radiation Oncology 1/2018 Zur Ausgabe

Update Onkologie

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