• No results found

Towards an optimal clinical protocol for the treatment of moving targets with pencil beam scanned proton therapy

N/A
N/A
Protected

Academic year: 2021

Share "Towards an optimal clinical protocol for the treatment of moving targets with pencil beam scanned proton therapy"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Towards an optimal clinical protocol for the treatment of moving targets with pencil beam scanned proton therapy

Ribeiro, Cássia O.

DOI:

10.33612/diss.126443635

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Ribeiro, C. O. (2020). Towards an optimal clinical protocol for the treatment of moving targets with pencil beam scanned proton therapy. University of Groningen. https://doi.org/10.33612/diss.126443635

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)
(3)
(4)
(5)

INTRODUCTION

In the 21

st

century, cancer is estimated to rank as the leading cause of death globally [1]. Radiotherapy, next to surgery and chemotherapy, is one of the most common treatment mo- dalities for cancer [2]. The principle of radiotherapy is to use ionising radiation to kill the cancer cells, while minimising the dose exposure to healthy tissue adjacent to the tumour.

The focus in this thesis is on moving targets, such as tu- mours in the thorax (e.g. lung and oesophagus) and tumours in the abdomen region (e.g. liver) [3,4]. Small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) are the two main types of lung cancer. The most frequent histological type in NSCLC is adenocarcinoma [5]. SCLC encompasses only about 20 % of all lung cancer cases, and this type of lung cancer shows a decrease in the incidence rates over time [6]. Worldwide, lung cancer is the leading cause of cancer incidence and death, with 2.1 million new lung cancer cases in 2018 and representing 18.4 % of the cancer deaths [1]. According to global statistics from 2018, oesophageal cancer ranks seventh in terms of incidence (572 000 new cases) and sixth in mortality overall (509 000 deaths) [1].

Liver cancer is predicted to be the sixth most commonly diagnosed cancer and the fourth leading cause of cancer death worldwide in 2018, with about 841 000 new cases and 782 000 deaths annually [1].

Within photon radiotherapy, intensity-modulated radia- tion therapy (IMRT) can deliver highly conformal dose dis- tributions, while sparing neighbouring critical organs-at-risk (OARs) [7]. Volumetric modulated arc therapy (VMAT) can be regarded as an extension of IMRT, which while providing at least similar target coverage and OARs dose sparing than IMRT, is capable of reducing the beam-on time and the required monitor units [8–12]. In terms of treatment related toxicities, even more clinical benefits are anticipated with proton radiotherapy for many cancer indications [13–16].

Treatment delivery strategies within proton therapy, such

as pencil beam scanned proton therapy (PBS-PT), with

(6)

General introduction

optimisation strategies such as intensity-modulated proton therapy (IMPT), are able to improve even more the dose conformity to the target, while reducing the dose to the OARs [17–20].

The continuous technological developments and emer- gence of highly conformal radiotherapy techniques are substantial for the improvement of thoracic and abdominal cancer treatments, due to their anatomic location and the critical OARs surrounding the tumour (lungs, heart, and liver). However, this increase in precision of radiotherapy techniques becomes a challenge due to the need of deal- ing with the inter- and intra- fractional motion for these patients (mainly due to respiration), and its associated potential disturbing effects on the planned dose distri- bution [21]. To guarantee target coverage for radiotherapy of moving targets, expansions in planning margins are then required, inevitably compromising healthy tissue dose sparing. Particularly for PBS-PT for moving targets, the current concern of plan robustness as a result of motion within the treatment delivery, hampers its wide clinical implementation worldwide.

The ultimate aim of this thesis is to overcome the current challenges and move forward to the development of an optimal clinical protocol, so that thoracic and abdominal indications would be able to benefit from the advantages of PBS-PT. For this purpose, an assessment of the possible dosimetric impacts of PBS-PT for an extended number of lung, oesophagus, and liver cancer patients is conducted.

PROTON THERAPY

Instead of using X-rays (photons) as in conventional radio- therapy, proton therapy uses charged particles (protons).

As can be seen by the shape of the beam depth-dose data

(Fig. 1), protons have a low entrance dose and release most

of their energy at the so-called Bragg peak (maximum dose

point), having then a finite range (sharp distal dose fall-off)

(7)

that depends on their initial energy and the density of the traversed material [22]. Due to these physical properties, the potential advantages of proton vs. photon dose distributions become clear. In proton therapy technique is possible to deliver a high tumour dose, and at the same time, superior healthy tissue sparing than with conventional radiotherapy can be achieved [23].

PBS-PT is the proton therapy modality we have available at our proton facility in the University Medical Center Gron- ingen (UMCG) (the UMCG Proton Therapy Center [GPTC]), which is a Proteus

®

Plus (PPlus) machine (IBA, Louvain-la- Neuve, Belgium) (Fig. 2). In this technique, thin monoen- ergetic proton beams are deflected by scanning magnets to create individually weighted pencil beams with various energies to scan (or ‘paint’) the target volume in depth.

When prescribing and delivering dose, we rely on the dose distributions calculated and optimised by a treatment planning system (TPS) [23]. In order to combine fields in a PBS-PT treatment plan, two main approaches can be used:

single-field uniform dose (SFUD) and IMPT. In SFUD, since

Fig. 1. Photon vs. proton beam depth-dose profiles (adapted from Mohan et al. [24]). The green curve depicts a typical 15 MV photon beam. The red curve shows a monoenergetic proton beam. The blue curve is obtained by electromechanically spreading the monoener- getic proton beam laterally and longitudinally. The top flat portion of the blue curve is called the spread-out Bragg peak.

(8)

General introduction Fig. 2. Schematic representation of the components of the beam delivery system equipment in the PBS-PT treatment mode for an IBA PPlus gantry based system (adapted from [25–27]).

(9)

each field is optimised independently, there is the assurance of homogeneous dose distributions delivered by the individ- ual fields. Conversely, in IMPT (proton equivalent of IMRT), all fields are simultaneously optimised [28]. For IMPT, the planning target volume (PTV) concept of photon therapy can be replaced by a clinical target volume (CTV) robust optimisation technique, which incorporates setup and range uncertainties into the planning (nominal) dose distribution (i.e. error-free dose distribution). This CTV based optimisa- tion process is known by 3D robust optimisation [29].

PBS-PT has the advantage, when compared to passively scattered proton therapy, of being able to deliver a more conformal dose distribution. However, this greater precision of PBS-PT (due to the sharp gradients of pencil beams), and especially IMPT, comes with the cost of higher sensitivity to any variations from the planning situation [30,31].

3D medical imaging for treatment preparation

As for photon therapy, the treatment preparation in proton therapy is based on acquired computed tomography (CT) images of each patient in treatment position. The CT is used to delineate the planning volumes and calculate the nominal dose distribution in the TPS. For patient alignment at the treatment table, on-board cone beam computed tomography (CBCT) systems are now integrated in many new proton therapy gantries [32]. While rotating around the patient, the CBCT acquires planar images, which can be reconstructed into volumetric images [33].

Deformable image registration (DIR)

DIR is required for several processes in radiotherapy [34,35].

DIR is the process of finding a point-to-point correspon-

dence map between positions in one medical scan and posi-

tions in another scan from the same patient. In other words,

DIR is used to estimate the patient’s deformable motion

present between two images. There is always a fixed image

and a moving image in a DIR process. To perform a DIR,

the fixed and the moving images are pre-defined and then

(10)

General introduction

the physical result is an estimated deformation vector field (DVF) containing vectors for each voxel, pointing from the fixed image towards the moving image. DIR is required for different purposes throughout the treatment planning work- flow, such as contour propagation, or dose warping, from the fixed (reference) image to the moving image.

It is well-known that DIR is an ill-posed problem intrinsi- cally. When applying DIR, the resulted estimated DVFs can be inaccurate. Additionally, another problem is that differ- ent DIR algorithms applied to the same image pair provide estimated DVFs that can remarkably differ from each other.

Therefore, the quality of different DIR algorithms needs to be carefully assessed [36].

3D dose evaluation

To ensure that the CTV receives adequate dose and to limit the dose to critical organs-at-risk (OARs) despite treatment uncertainties, robust optimisation [29] and robustness eval- uation [37] are used in this work. Both robust optimisation and robustness evaluation follow a probabilistic treatment planning approach (scenario based). In photon therapy in- stead, a PTV margin approach is standardly used [38,39].

In the PTV method, the treatment uncertainties for each indication are quantified to create margin sizes, ensuring that for 90 % of the patient population, the CTV receives at least 95 % of the prescribed dose [38]. The probabilistic approach used here incorporates the dosimetric uncertainties of a certain scenario into the dose, using directly the CTV for plan optimisation and evaluation purposes.

The PTV method is not adequate for proton therapy due to the need to consider range errors. The robustly opti- mised plans generated in our commercial TPS RayStation (RaySearch Laboratories, Stockholm, Sweden) are afterwards robustly evaluated. This is necessary to guarantee adequate target coverage and minimal OARs dose under all uncer- tainty scenarios [37].

The increased sensitivity of proton therapy to geomet-

ric accuracy requires, besides accurate patient positioning,

(11)

recurrent volumetric imaging throughout the course of treat- ment [32]. To take into account patient inter-fractional an- atomic changes, verification (repeated) CTs can be acquired during treatment. As an alternative to routine CT imaging, on-board CBCT images can also be used. In comparison with repeated CT, CBCT provides insufficient image quality for direct and accurate dose calculations [40,41]. However, CBCT has the advantage of enabling daily monitoring of the patient’s anatomy prior to the treatment fraction.

PROTON THERAPY FOR MOVING TARGETS

When using a highly precise treatment technique such as PBS-PT for moving targets, several disturbing effects can occur within the treatment delivery, deteriorating the actual delivered dose distribution. To be able to evaluate the possi- ble uncertainties in the PBS-PT planned dose distributions for moving targets, extensive 4D robustness evaluations, including 4D dose calculations, are compulsory.

4D medical imaging

For the treatment preparation of moving targets with PBS-PT, time-resolved 3D (4D) imaging is necessary for quantifying the motion characteristics. The 4DCT data set contains sev- eral 3DCTs (or phases), each giving information of the patient anatomy at a certain moment during an averaged breathing cycle. A 4DCT consists of snapshot representations of the breathing motion of the patient. For the reconstruction of a 4DCT, it is assumed that the breathing pattern is regular, which in reality is rarely true.

4DCBCT reconstruction [42,43] is used for 4D image guid-

ance for both thoracic and abdominal cancer patients treated

with photon therapy [44], and its feasibility has also been

investigated for proton therapy [45]. Furthermore, 4DMRI

arises as a viable non-additional imaging dose solution, since

it provides better soft tissue contrast, while limiting dose

to the patient [46].

(12)

General introduction

Interplay effects

When employing PBS-PT for the treatment of moving tar- gets, interplay effects can occur. Interplay effects happen due to the relative motion between the target and the scanned proton beam delivery. The CTV motion will result in a mis- positioning of the planned pencil beams. This will result in dose in-homogeneities, producing cold or hot spots within the target (Fig. 3). Although interplay effects may lead to pro- nounced deteriorations of the dose distribution for a single fraction, these are potentially smoothed out over a fraction- ated treatment course [47,48].

4D dose evaluation

For the treatment of moving targets in free breathing with PBS-PT, dosimetric disturbances caused by motion (target miss, dose blurring, and the interplay effects) need to be con- sidered by a 4D dose calculation (accumulation) algorithm [47–49]. A 4D dose calculation simulates the dynamics of the pencil beam scanning, thus being crucial to evaluate the deterioration of the delivered dose distribution due to the interplay effects. Both a 4DCT and DIR application in 4D images are necessary to perform a 4D dose calculation for a moving indication (Fig. 4).

There are at least two different ways of performing a 4D

dose calculation. The most standard one (and usually the

one available in most TPSs) consists in performing multiple

calculations of the corresponding dose on the particular

phase of the 4DCT in which was delivered [50–53]. Then

these doses are subsequently warped back to a reference

phase where they will be accumulated (or summed). Another

developed and experimentally validated 4D dose calculation

engine for PBS-PT involves deforming the dose grid (using

DIR) as a function of time [54]. After performing a 4D dose

calculation, we obtain a 4D dose distribution in a reference

CT, which represents the actual delivered dose considering

possible interplay effects. Within any 4D dose calculation,

the intrinsic inaccuracies of DIR can directly prompt dis-

tinguishable differences in dose distribution, which would

(13)

consequently influence any further dosimetric analysis and clinical decision [55].

Motion mitigation techniques

There are several ways to reduce the impact of motion effects in the delivered dose distributions: beam gating or tracking, rescanning, breath-hold, abdominal compression, increase in

Fig. 3. A: Nominal dose distribution and B: fraction dose distribu- tion affected by interplay effects for a sample lung cancer patient planned with PBS-PT.

Fig. 4. Workflow for a 4D dose calculation algorithm.

(14)

General introduction

spot size, 4D robust optimisation, among others [56]. Motion mitigation techniques explored in this thesis are rescanning, breath-hold, and 4D robust optimisation.

Rescanning consists in delivering the planned proton spots multiple times within each field [57]. There are sev- eral flavours of rescanning. The rescanning type that can be applied in a PBS-PT PPlus machine, for instance, is called scaled (controlled) [58]. Scaled rescanning can be done using either a layered or a volumetric approach [59]. 4D robust optimisation has been introduced and suggested to moderate respiratory-induced dosimetric perturbations [60–63]. This IMPT treatment planning approach includes several 4DCT phases during the optimisation process. To mitigate inter- and intra-fractional motion for PBS-PT of moving targets within the treatment delivery, deep inspiration breath-hold (DIBH) gating is also currently clinically implemented [64].

THESIS OUTLINE

Towards an optimal clinical PBS-PT planning protocol for moving targets, the possible dosimetric uncertainties (with and without the application of motion mitigation techniques) are thoroughly investigated for thoracic and abdominal tu- mours in the six remaining chapters of this thesis.

Chapter 2 focuses on incorporating all the possible disturb- ing effects in a treatment course of PBS-PT for moving targets in a dedicated tool for 4D evaluation (the 4D robustness eval- uation method [4DREM]). The 4DREM was implemented using the scripting capabilities of our TPS (RayStation) and was applied to exemplary IMPT plans of two thoracic indi- cations to illustrate its capability.

Chapter 3 presents an exhaustive evaluation of uncertain-

ties occurring when using different DIR algorithms. The

geometric error as well as the dosimetric error (the latter

one provided by 4D dose calculations), were analysed and

quantified for a comprehensive 4DCT data set of liver cancer

patients treated with SFUD.

(15)

In Chapter 4, the 4DREM (Chapter 2) is used to evaluate thoroughly designed planning protocols (using 3D or 4D robust optimisation approaches and also the DIR error infor- mation from Chapter 3) for a representative number of lung and oesophagus indications. Extensive patient 4DCT images (data acquired as part of a clinical trial) and treatment plan delivery information were used in this chapter. As a result of this study, an optimal and efficient clinical protocol for the first thoracic patients treated at our proton facility was established.

In Chapter 5, the 4DREM (Chapter 2) assesses two dif- ferent beam delivery configurations, provided by different PBS-PT implementations at Proteus

®

One and Proteus

®

Plus, respectively. This chapter uses NSCLC patient information and machine specific data.

Chapter 6 arouse as a follow-up of Chapter 3. This chapter consists in an evaluation of the provided DIR ambiguity for inter-fractional variations (i.e. differences in dose warping and accumulation provided by several different DIR algo- rithms). NSCLC patients under DIBH conditions planned with IMPT are included in this study.

In the last chapter of this thesis (Chapter 7), all the findings

and conclusions are summarised and future perspectives

are discussed.

(16)

General introduction

[1] Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A.

Global Cancer Statistics 2018:

GLOBOCAN Estimates of Inci- dence and Mortality Worldwide for 36 Cancers in 185 Countries.

CA Cancer J Clin 2018;68:394–424.

[2] Baskar R, Lee KA, Yeo R, Yeoh K-W. Cancer and Radiation Therapy: Current Advances and Future Directions. Int J Med Sci 2012;9:193–9.

[3] Torshabi AE, Dastyar SAR. Motion Challenge of Thoracic Tumors at Radiotherapy by Introducing an Available Compensation Strategy.

Radiotherapy, 2017, p. 263–80.

[4] Maingon P, Nouhaud É, Mornex F, Créhange G. Stereotactic body radiation therapy for liver tu- mours. Cancer/Radiothérapie 2014;18:313–9.

[5] Couraud S, Zalcman G, Milleron B, Morin F, Souquet P-J. Lung can- cer in never smokers – A review.

Eur J Cancer 2012;48:1299–311.

[6] Riaz SP, Lüchtenborg M, Cou- pland VH, Spicer J, Peake MD, Møller H. Trends in incidence of small cell lung cancer and all lung cancer. Lung Cancer 2012;75:280–4.

[7] Oh Y-K, Baek J-G, Kim O-B, Kim J-H. Assessment of setup uncertainties for various tumor sites when using daily CBCT for more than 2200 VMAT treat- ments. J Appl Clin Med Phys 2014;15:85–99.

[8] Van Benthuysen L, Hales L, Pod- gorsak MB. Volumetric Modulated Arc Therapy vs. IMRT for the Treatment of Distal Esophageal Cancer. Med Dosim 2011;36:404–9.

[9] Wijsman R, Dankers F, Troost EGC, Hoffmann AL, van der Heijden EHFM, de Geus-Oei L-F, et al. Comparison of toxicity and outcome in advanced stage non- small cell lung cancer patients treated with intensity-modulated (chemo-) radiotherapy using IMRT or VMAT. Radiother Oncol 2017;122:295–9.

[10] Wang P-M, Hsu W-C, Chung N-N, Chang F-L, Fogliata A, Cozzi L.

Radiation treatment with vol- umetric modulated arc therapy of hepatocellular carcinoma patients. Early clinical outcome and toxicity profile from a retro- spective analysis of 138 patients.

Radiat Oncol 2012;7:1–8.

[11] Tsai Y-C, Tsai C-L, Hsu F-M, Wu J-K, Wu C-J, Cheng JC-H. Supe- rior liver sparing by combined coplanar/noncoplanar volu- metric-modulated arc therapy for hepatocellular carcinoma: A planning and feasibility study.

Med Dosim 2013;38:366–71.

[12] Bae SH, Jang W Il, Park HC. In- tensity-modulated radiotherapy for hepatocellular carcinoma:

Dosimetric and clinical results.

Oncotarget 2017;8:59965–76.

[13] Ask A, Johansson B, Glimelius B. The potential of proton beam

REFERENCES

(17)

radiation therapy in gastrointes- tinal cancer. Acta Oncol (Madr) 2005;44:896–903.

[14] Dionisi F, Ben-Josef E. The Use of Proton Therapy in the Treatment of Gastrointestinal Cancers: Liver.

Cancer J 2014;20:371–7.

[15] Chuong MD, Hallemeier CL, Jab- bour SK, Yu J, Badiyan S, Merrell KW, et al. Improving Outcomes for Esophageal Cancer using Pro- ton Beam Therapy. Int J Radiat Oncol Biol Phys 2016;95:488–97.

[16] Chang JY, Jabbour SK, De Ruysscher D, Schild SE, Simone CB, Rengan R, et al. Consensus Statement on Proton Therapy in Early-Stage and Locally Advanced Non-Small Cell Lung Cancer.

Int J Radiat Oncol Biol Phys 2016;95:505–16.

[17] Welsh J, Gomez D, Palmer MB, Riley BA, Mayankkumar A V, Komaki R, et al. Intensity-Mod- ulated Proton Therapy Further Reduces Normal Tissue Exposure During Definitive Therapy for Locally Advanced Distal Esopha- geal Tumors: A Dosimetric Study.

Int J Radiat Oncol Biol Phys 2011;81:1336–1342.

[18] Stuschke M, Kaiser A, Pöttgen C, Lübcke W, Farr J. Potentials of robust intensity modulated scanning proton plans for locally advanced lung cancer in com- parison to intensity modulated photon plans. Radiother Oncol 2012;104:45–51.

[19] Zhang Y, Huth I, Wegner M, Weber DC, Lomax AJ. An evaluation of

rescanning technique for liver tu- mour treatments using a commer- cial PBS proton therapy system.

Radiother Oncol 2016;121:281–7.

[20] Chang JY, Zhang X, Knopf A, Li H, Mori S, Dong L, et al. Consensus Guidelines for Implementing Pencil-Beam Scanning Proton Therapy for Thoracic Malignan- cies on Behalf of the PTCOG Thoracic and Lymphoma Sub- committee. Int J Radiat Oncol Biol Phys 2017;99:41–50.

[21] Jiang SB. Radiotherapy of Mobile Tumors. Semin Radiat Oncol 2006;16:239–48.

[22] Paganetti H, Bortfeld T. Proton beam radiotherapy - The state of the art. New Technol. Radiat.

Oncol., 2005, p. 3–540.

[23] Paganetti H. Proton beam therapy.

Prot. beam Ther., 2017, p. 1–23.

[24] Mohan R, Mahajan A, Minsky BD. New strategies in radiation therapy: Exploiting the full po- tential of protons. Clin Cancer Res 2013;19:6338–43.

[25] Marchand B, Prieels D, Bauvir B, Sépulchre R, Gérard M. IBA proton pencil beam scanning: an innovative solution for cancer treatment. Proc. EPAC, 2000, p.

2539–41.

[26] Rana S, Bennouna J, Samuel EJJ, Gutierrez AN. Development and long-term stability of a compre- hensive daily QA program for a modern pencil beam scanning (PBS) proton therapy delivery system. J Appl Clin Med Phys 2019;20:29–44.

(18)

General introduction

[27] Wang P, Zheng J, Song Y, Zhang W, Wang M. Analysis and design of an energy verification system for SC200 proton therapy facility.

Electron 2019;8:1–9.

[28] Lomax A. SFUD, IMPT, and Plan Robustness. Part. Radiother.

Emerg. Technol. Treat. Cancer, 2016, p. 169–94.

[29] Fredriksson A, Forsgren A, Hår- demark B. Minimax optimization for handling range and setup uncertainties in proton therapy.

Med Phys 2011;38:1672–84.

[30] Lomax AJ. Intensity modulated proton therapy and its sensitivity to treatment uncertainties 1: the potential effects of calculational uncertainties. Phys Med Biol 2008;53:1027–42.

[31] Lomax AJ. Intensity modulated proton therapy and its sensitivity to treatment uncertainties 2: The potential effects of inter-fraction and inter-field motions. Phys Med Biol 2008;53:1043–56.

[32] Veiga C, Janssens G, Teng C-L, Bau- dier T, Hotoiu L, McClelland JR, et al. First Clinical Investigation of Cone Beam Computed Tomogra- phy and Deformable Registration for Adaptive Proton Therapy for Lung Cancer. Int J Radiat Oncol Biol Phys 2016;95:549–59.

[33] Molitoris JK, Diwanji T, Snider JW, Mossahebi S, Samanta S, Badiyan SN, et al. Advances in the use of motion management and image guidance in radiation therapy treatment for lung cancer.

J Thorac Dis 2018;10:S2437–50.

[34] K. Ø. Noe. Deformable Image Reg- istration for Use in Radiotherapy.

2009.

[35] Kadoya N. Use of deformable im- age registration for radiotherapy applications. J Radiol Radiat Ther 2014;2:1–7.

[36] Kierkels RGJ, den Otter LA, Ko- revaar EW, Langendijk JA, van der Schaaf A, Knopf AC, et al.

An automated, quantitative, and case-specific evaluation of de- formable image registration in computed tomography images.

Phys Med Biol 2018;63:045026.

[37] Korevaar EW, Habraken SJM, Scandurra D, Kierkels RGJ, Unipan M, Eenink MGC, et al.

Practical robustness evaluation in radiotherapy – A photon and proton-proof alternative to PTV- based plan evaluation. Radiother Oncol 2019.

[38] Van Herk M, Remeijer P, Rasch C, Lebesque J V. The probability of correct target dosage: Dose-pop- ulation histograms for deriving treatment margins in radiother- apy. Int J Radiat Oncol Biol Phys 2000;47:1121–35.

[39] Tilly D, Ahnesjö A. Fast dose al- gorithm for generation of dose coverage probability for robust- ness analysis of fractionated radiotherapy. Phys Med Biol 2015;60:5439–54.

[40] Kurz C, Kamp F, Park Y-K, Zöllner C, Rit S, Hansen D, et al. Inves- tigating deformable image reg- istration and scatter correction for CBCT-based dose calculation

(19)

in adaptive IMPT. Med Phys 2016;43:5635–46.

[41] Veiga C, Janssens G, Baudier T, Hotoiu L, Brousmiche S, Mc- Clelland J, et al. A comprehen- sive evaluation of the accuracy of CBCT and deformable regis- tration based dose calculation in lung proton therapy. Biomed Phys Eng Express 2017;3:015003.

[42] Jin P, Hulshof MCC., van Wi- eringen N, Bel A, Alderliesten T.

Interfractional variability of respi- ration-induced esophageal tumor motion quantified using fiducial markers and four-dimensional cone-beam computed tomography.

Radiother Oncol 2017;124:147–54.

[43] Bryce-Atkinson A, Marchant T, Rodgers J, Budgell G, McWilliam A, Faivre-Finn C, et al. Quantita- tive evaluation of 4D Cone beam CT scans with reduced scan time in lung cancer patients. Radiother Oncol 2019;136:64–70.

[44] Velec M, Moseley JL, Craig T, Dawson LA, Brock KK. Accumu- lated Dose in Liver Stereotactic Body Radiotherapy: Positioning, Breathing, and Deformation Ef- fects. Int J Radiat Oncol Biol Phys 2012;83:1132–40.

[45] Niepel K, Kamp F, Kurz C, Hansen D, Rit S, Neppl S, et al. Feasibility of 4DCBCT-based proton dose calculation: An ex vivo porcine lung phantom study. Z Med Phys 2019;29:249–61.

[46] von Siebenthal M. Analysis and Modelling of Respiratory Liver Motion using 4DMRI. 2008.

[47] Bert C, Grözinger SO, Rietzel E.

Quantification of interplay effects of scanned particle beams and moving targets. Phys Med Biol 2008;53:2253–65.

[48] Grassberger C, Dowdell S, Lomax A, Sharp G, Shackleford J, Choi N, et al. Motion Interplay as a Function of Patient Parameters and Spot Size in Spot Scanning Proton Therapy for Lung Cancer.

Int J Radiat Oncol 2013;86:380–6.

[49] Dowdell S, Grassberger C, Sharp G, Paganetti H. Interplay effects in proton scanning for lung: a 4D Monte Carlo study assessing the impact of tumor and beam delivery parameters. Phys Med Biol 2013;58:4137–4156.

[50] Rietzel E, Chen GTY, Choi NC, Willet CG. Four-dimensional image-based treatment planning:

Target volume segmentation and dose calculation in the presence of respiratory motion. Int J Radiat Oncol Biol Phys 2005;61:1535–50.

[51] Lin L, Kang M, Huang S, Mayer R, Thomas A, Solberg TD, et al. Beam specific planning target volumes incorporating 4DCT for pencil beam scanning proton therapy of thoracic tumors. J Appl Clin Med Phys 2015;16:281–92.

[52] Inoue T, Widder J, van Dijk L V., Takegawa H, Koizumi M, Takash- ina M, et al. Limited Impact of Setup and Range Uncertainties, Breathing Motion, and Interplay Effects in Robustly Optimized Intensity Modulated Proton Therapy for Stage III Non-small

(20)

General introduction

Cell Lung Cancer. Int J Radiat Oncol 2016;96:661–9.

[53] Pfeiler T, Bäumer C, Engwall E, Geismar D, Spaan B, Timmermann B. Experimental validation of a 4D dose calculation routine for pencil beam scanning proton therapy. Z Med Phys 2018;28:121–33.

[54] Krieger M, Klimpki G, Fattori G, Hrbacek J, Oxley D, Safai S, et al.

Experimental validation of a de- forming grid 4D dose calculation for PBS proton therapy. Phys Med Biol 2018;63:11pp.

[55] Zhang Y, Boye D, Tanner C, Lomax AJ, Knopf A. Respiratory liver motion estimation and its effect on scanned proton beam therapy.

Phys Med Biol 2012;57:1779–95.

[56] Grassberger C, Dowdell S, Sharp G, Paganetti H. Motion mitigation for lung cancer patients treated with active scanning proton ther- apy. Med Phys 2015;42:2462–9.

[57] Knopf AC, Hong TS, Lomax A.

Scanned proton radiotherapy for mobile targets - The effectiveness of re-scanning in the context of different treatment planning ap- proaches and for different motion characteristics. Phys Med Biol 2011;56:7257–71.

[58] Zenklusen SM, Pedroni E, Meer D. A study on repainting strat- egies for treating moderately moving targets with proton pencil beam scanning at the new Gantry 2 at PSI. Phys Med Biol 2010;55:5103–21.

[59] Bernatowicz K, Lomax AJ, Knopf A. Comparative study of layered

and volumetric rescanning for different scanning speeds of pro- ton beam in liver patients. Phys Med Biol 2013;58:7905–20.

[60] Graeff C. Motion mitigation in scanned ion beam therapy through 4D-optimization. Phys Medica 2014;30:570–7.

[61] Liu W, Schild SE, Chang JY, Liao Z, Chang Y-H, Wen Z, et al. Ex- ploratory Study of 4D versus 3D Robust Optimization in Intensity Modulated Proton Therapy for Lung Cancer. Int J Radiat Oncol 2016;95:523–33.

[62] Yu J, Zhang X, Liao L, Li H, Zhu R, Park PC, et al. Motion-robust intensity-modulated proton ther- apy for distal esophageal cancer.

Med Phys 2016;43:1111–8.

[63] Ge S, Wang X, Liao Z, Zhang L, Sahoo N, Yang J, et al. Potential for improvements in robustness and optimality of intensity-mod- ulated proton therapy for lung cancer with 4-Dimensional robust optimization. Cancers (Basel) 2019;11:1–15.

[64] Boda-Heggemann J, Knopf A-C, Simeonova-Chergou A, Wertz H, Stieler F, Jahnke A, et al. Deep Inspiration Breath Hold-Based Radiation Therapy: A Clinical Review. Int J Radiat Oncol Biol Phys 2016;94:478–92.

(21)

Referenties

GERELATEERDE DOCUMENTEN

Chapter 5 Evaluation of intrinsic ‘rescanning’ (pulsed beam) versus scaled rescanning (continuous beam) for the treatment of moving targets with pencil beam scanned proton

The voxel-wise worst-case dose distributions obtained from the 4DREM were used to assess the robustness of 3D and 4D robust optimised IMPT treatment plans of a lung and

Towards an optimal clinical protocol for the treatment of moving targets with pencil beam scanned proton therapy.. Ribeiro,

Planning target volume (PTV)-based VMAT plans and internal clinical target volume (ICTV)- based robust IMPT plans were optimized in 3D on the av_p4DCT and re-calculated on

For the challenging issue to correct the measured drag values, at least the missing integration surface due to the opening for the sting seems to be correctable: Since

Het gerechtshof is van mening dat een beding waarbij de bevoegdheid de door de verzekeringsnemer verschuldigde kosten te berekenen en vast te stellen wordt

Naar de mening van De Vries blijkt uit jurisprudentie, met betrekking tot reguliere bedrijfsmiddelen, dat sommige kosten in een te ver verwijderd causaal verband staan met de

Hoewel de mate van centralisatie, advertorial of reguliere advertentie, geen invloed blijkt te hebben op de betrokkenheid bij Instagram, blijkt deze wel invloed uit te oefenen op