Download as pdf or txt
Download as pdf or txt
You are on page 1of 32

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/269465283

Image-Guided Radiation Therapy IntroductIon

Chapter · October 2012

CITATIONS READS
0 3,144

4 authors, including:

Guang George Li Lei Dong


Memorial Sloan Kettering Cancer Center University of Pennsylvania
122 PUBLICATIONS   1,557 CITATIONS    504 PUBLICATIONS   16,376 CITATIONS   

SEE PROFILE SEE PROFILE

Radhe Mohan
University of Texas MD Anderson Cancer Center
423 PUBLICATIONS   25,258 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Patient setup accuracy of SGRT-only for conventional radiotherapy View project

Biological response theories and radiation therapy View project

All content following this page was uploaded by Guang George Li on 13 December 2014.

The user has requested enhancement of the downloaded file.


[AQ1] CHAPTER
14 Image-Guided Radiation Therapy

[AQ2] Guang Li  ■  Gig S. Mageras  ■  Lei Dong  ■  Radhe Mohan

Introduction inter-fractional setup (i.e., variation between treatment


fractions) and intra-fractional organ motion (i.e., motion
Most modern radiation therapy of cancer involves nonin- occurring within a treatment fraction) could be corrected
vasive targeting of localized disease and conformal avoid- for more accurate delivery of the planned treatment dose
ance of nontarget tissues. Radiation dose is delivered to to the target. As the variation of normal tissues is usually
the volume of intended treatment without the benefit of ignored in the current image-guided approach, dosimetric
physically seeing the regions being irradiated. Therefore, and clinical consequences must be assessed.
imaging guidance is crucial in every step of the radiation Increasing evidence shows that there are substantial
therapy, including cancer diagnosis, staging, and delinea- inter- and intra-fractional variations in the shapes, vol-
tion; treatment simulation and planning; patient setup, umes, and positions of treatment targets and the interven-
tumor localization and motion monitoring; and treatment ing and surrounding normal tissues, in contrast to the
response assessment, efficacy evaluation and strategy “snapshot” planning anatomy of a patient. The causes of
refinement. In fact, most of the significant advances in such variations include body shift, rotation and deforma-
radiation oncology over the last three decades have been tion, respiration-induced organ motion, weight loss, and
made possible by preceding advances in medical imaging, radiation-induced changes such as tumor shrinkage.
including computed tomography (CT), magnetic reso- These variations could have a significant impact on the
nance imaging (MRI), magnetic resonance spectroscopic outcome of treatments, as they may result in underdosing
imaging (MRSI), positron emission tomography (PET), the target or overdosing the organs at risk (1–3). In the
single photon emission computed tomography (SPECT), current state of the art of the planning and delivery of
and ultrasound (US). With the aid of CT and MRI, three- radiation therapy, based on the use of CT, PET, and MRI
dimensional (3D) shapes of treatment regions and critical images acquired prior to the course of treatments, it is
normal structures are delineated with great precision, thus assumed implicitly that patient’s anatomy discerned from
reducing the chance for marginal misses of the tumor and initial imaging remains static throughout the course of the
minimizing the exposure of normal tissues to high radia- radiation therapy. In the current practice, wide treatment
tion dose. MRI, MRSI, PET, and SPECT images provide margins, derived from population-based studies, are used
additional functional and metabolic information to define to ensure coverage of the disease, exposing considerable
and delineate the extent of the disease. Examples of image volumes of normal tissues to unwanted radiation. The use
guidance in various stages of radiation therapy are illus- of large margins limits the ability to safely deliver higher
trated in Figure 14.1. tumor doses because of increased risk of normal tissue
Image-guided radiation therapy (IGRT) is composed toxicity, especially for hypo-fractioned stereotactic body
of a multitude of major innovations to address some of radiotherapy (SBRT), in which the high dose per fraction
the problems arising from inter- and intra-fractional ana- exceeds the normal tissue’s capacity for sublethal repair.
tomic variations. IGRT aims to deliver a radiation treat- Furthermore, the margin needed for some patients exhib-
ment as it is planned based on an image acquired at iting large target variations may exceed the population-
simulated treatment conditions. This simulation image based margin, potentially leading to marginal misses,
establishes a reference of 3D patient anatomy (with pos- especially with the use of highly conformal modalities,
sible inclusion of tumor respiratory motion information) such as 3D conformal radiotherapy (3DCRT), intensity-
for both image-based treatment planning and image- modulated radiotherapy (IMRT), volumetric-modulated
guided treatment delivery. The former follows the exact arc therapy (VMAT), and proton therapy (4–6). Treat-
3D anatomy (both the tumor and normal tissues) for ment planning and delivery techniques that do not correct
dosimetric planning, while the latter focuses mostly on for such daily volumetric variations adequately may lead
tumor alignment between the reference image and daily to suboptimal treatments. These factors may, in part, be
images at the treatment unit prior to and during treat- responsible for the poor outcome and high toxicity in
ment, in reference to the treatment beams. Therefore, radiation therapy for some cancers. IGRT has the potential
the variations of tumor position in the image-guided to target gross and microscopic diseases accurately, to

229

LWBK933_Ch14_p229-258.indd 229 6/1/11 6:43 PM


230 n  Treatment Planning in Radiation Oncology

2D Radiographic Imaging
Two-dimensional (2D) radiographic (projection) imaging
is typically used in treatment rooms to align the patient
relative to the radiation beams. Megavoltage (MV) elec-
tronic portal imaging (EPI) is the most commonly used
form of radiographic imaging (9,10). MV imaging uses
therapy x-ray beams and an amorphous-silicon (a-Si) flat-
panel imager to verify patient’s setup, defined as the posi-
tion of the skeletal anatomy. Other uses of MV imaging
are to verify treatment beam apertures prior to treatment
(11,12) and in vivo dosimetry during treatment (13).
Because the same therapy MV x-ray beam is used for ver-
ification, it provides direct in-field verification of treat-
ment delivery, and therefore serves as a “gold standard” for
validating new IGRT techniques. Disadvantages of MV
imaging include higher radiation dose to the patient from
the procedure (typically 1–5 cGy) and poorer image qual-
Figure 14.1.  Image guidance at various stages of the radiotherapy ity due to a large Compton scattering contribution from
process. the higher x-ray energies. Recently, in-line low-energy
(∼4 MV), with a low atomic number (Z), target (e.g., car-
bon) without beam flattening filter has become available,
individualize treatments to reduce margins, and to allow which produces portal images of improved soft-tissue
radiation dose escalation to higher levels with the expecta- contrast with lower imaging dose (14).
tion of improving local control and reducing toxicity Two general categories of 2D kilovoltage (kV) x-ray
(7,8). Although this rationale is supported by increasing imaging are frequently used for IGRT. One is a gantry-
evidences, continuing research to further develop IGRT mounted kV imaging system on a linear accelerator (linac)
methods for clinical trials is required to determine the true that is orthogonal to the therapy MV x-ray beam. The
clinical promise of IGRT. x-ray source and flat-panel imager are mounted on
[AQ3] This chapter focuses on IGRT technologies related to retractable arms. Kilovoltage x-ray imaging provides near-
treatment planning as well as treatment delivery. The diagnostic quality images. With either kV or MV imaging,
related topics are covered in the following sections: second determination of the correction to patient position com-
and third sections introduce various forms of IGRT tech- monly uses orthogonal image pairs that are matched to
nologies and their commercial implementations for inter- digitally reconstructed radiographs (DRRs) derived from
fractional and intra-fractional imaging, respectively. the planning CT as a reference. The second category of kV
Fourth section reviews requirements and considerations imaging is ceiling-mounted systems (15). These systems
for IGRT, including quality assurance (QA). Various pos- provide an oblique orthogonal image pair for stereoscopic
sible IGRT strategies, margin assessment and reduction, imaging at a wide range of treatment couch angles. Most
and clinical implications are described in fifth section. kV x-ray radiographic imaging systems have a companion
Finally, sixth section attempts to look into the future and fluoroscopic imaging mode, which is useful for observing
speculate on new processes coming into this field. motion of the internal anatomy or implanted fiducial
markers.
Kilovoltage radiographs are often not sufficient for
Inter-Fractional Igrt detecting soft-tissue targets but are more successful in
Imaging Modalities aligning skeletal landmarks or implanted radio-opaque
fiducials as target surrogates. In-room kV x-ray imaging
In this section, we focus on in-room IGRT imaging represents a major improvement over MV imaging due to
modalities for daily patient setup. Images acquired imme- its superior image quality and its low imaging dose. The
diately prior to treatment are used to reposition the different appearance of kV and MV images, as shown in
patient so as to align the target or its surrogate (such as Figure 14.2 (thorax), results from the higher proportion of
implanted radio-opaque fiducial markers in or near the Compton scattering and high-energy electrons in MV
tumor) with the planned radiation beams. This is the x-ray beams relative to kV x-rays.
simplest form of image-guided radiotherapy without As the kV imaging beam lines are not aligned with MV
modification of the original treatment design. A couch beam line, the kV-MV isocenter discrepancy must be
positional adjustment is typically used to realign the established within a clinical tolerance through initial and
patient. The inter-fractional imaging modalities may be periodic QA processes. The radiation dose from kV imag-
classified as follows. ing is low, typically in the range of 0.01 to 0.1  cGy per

LWBK933_Ch14_p229-258.indd 230 6/1/11 6:43 PM


Image-Guided Radiation Therapy  n 231

[Au:
Please
check
figures of
this chapter
for better
quality.]

Figure 14.2.  The appearance of anatomy kV (top row) and MV (bottom row) radiographs can be quite different. At kV
x-ray energies, the bony structures are enhanced; at the therapeutic (MV) energies, the air cavity is enhanced. Direct
comparison of kV DRRs with MV portal images can be difficult.

image, which facilitates its use for daily image-guided stereotactic radiotherapy of brain and lung cancers, to
patient setup. reduce the uncertainty in multi-fractional frame-based
stereotactic radiotherapy. A combination of single-slice
3D Tomographic Imaging Philips CT scanner and Varian Clinac 2100EX, which used
a rail system to transport the patient between treatment
CT imaging inside the treatment room provides 3D ana- and CT couches, was assembled at the Memorial Sloan-
tomical information and improved soft-tissue visibility, Kettering Cancer Center. The system was used for treat-
thus providing advantages over radiographic imaging. In- ment of paraspinal lesions and prostate cancer (17,18).
room CT images may potentially be used to reconstruct The first commercial CT-linac system in the United
dose distributions based on anatomy captured at treat- States was installed in 2000 in Morristown Memorial
ment; its application to adaptive radiation therapy, in Hospital, New Jersey (19). The system consists of a Siemens
which a patient’s treatment plan is modified in response to medical linac and a moveable Siemens CT scanner that
changes in anatomy, is an active area of investigation. In slides along a pair of rails (“CT-on-Rails”). A picture of
the following sections, we describe various forms of in- the system is shown in Figure 14.3A. The CT scan is per-
room CT-based on x-ray systems. formed with the patient rotated 180 degrees from the
treatment position. The initial clinical experience with this
kV Helical CT system has been reported by Wong et al. and Fung et al.
Helical single- or multiple-slice CT systems have been (19,20). A similar “CT-on-Rails” commercial system is
widely used in diagnostic imaging and radiation treatment installed at the University of Texas M. D. Anderson Cancer
planning for many years. The first integrated clinical sys- Center (EXaCT™, Varian Oncology Systems, Palo Alto,
tem, which combined a linac unit and conventional CT CA). The mechanical accuracy of the system is found to be
unit in the same room was developed by Uematsu et al. at within 0.5 mm (21). The system, as shown in Figure 14.3B,
the National Defense Medical College, Saitama, Japan has been recently upgraded with a 6-degree-of-freedom
(16). The system was designed for noninvasive, frameless couch (HexaPOD™, Medical Intelligence, Germany),

LWBK933_Ch14_p229-258.indd 231 6/1/11 6:43 PM


232 n  Treatment Planning in Radiation Oncology

A B
Figure 14.3.  A: A Siemens Primatom™ CT-on-rails contains a Primus™ linear accelerator and a Somatom™ sliding-gantry CT scanner. The
first Primatom™ was installed at the Morristown Memorial Hospital, New Jersey (Photograph courtesy of Lisa Grimm, PhD). B: A CT-on-rails
system combining a General Electric Smart Gantry™ CT scanner and a Varian 2100EX linear accelerator. A 6-degree-of-freedom couch
(HexaPOD™, Medical Intelligence, Germany) is installed on top of the standard Varian ExaCT™ couch. CT scan of the patient in immobilized
treatment position is acquired after rotating the couch 180 degrees.

which allows for more precise couch translations and periodically to maintain image quality and geometric accu-
small rotations. The system has been routinely used for racy (27). Corrections on the order of 0.2 cm are required
fractionated SBRT, treatment of spinal metastases (22), to compensate for the gravity-induced flex in the support
and lung cancers. The biggest advantage of an in-room CT arms of the source, detector, and gantry. Sub-millimeter
scanner for IGRT is the similarity of the image quality and spatial resolution has been demonstrated in phantom. It is
fields of view with planning CT images. possible to reconstruct volumetric images with nearly iso-
tropic spatial resolution, which is useful in cases requiring
kV Cone-Beam CT high resolution, such as stereotactic radiosurgery. Kilovoltage
Gantry-mounted kV imaging systems are capable of radiog- CBCT images demonstrate acceptable soft-tissue contrast for
raphy, fluoroscopy, and cone-beam CT (CBCT), providing a target and organ-at-risk localization.
versatile solution for IGRT applications (23–25). CBCT Since October 2005, all three major manufacturers have
imaging involves acquisition of projection images of the offered CBCT capabilities (Elekta Synergy™, Elekta Inc.,
patient as the gantry rotates through an arc of at least Sweden; Varian On-board Imager (OBI), Palo Alto, CA;
180 degrees plus the “cone-beam angle” subtended by the and Siemens Artiste™, Germany). A picture of the com-
imaging panel (∼200 degrees total). A filtered back-projection mercial implementation of the kV-CBCT system by Elekta
algorithm is used to reconstruct the volumetric images (Synergy™) and by Varian (TrueBeam™) is shown in
(26). Geometric calibration of the CBCT system is needed Figure 14.4A and B, respectively. Elekta’s system uses a

A B
Figure 14.4.  A: An Elekta Synergy™ unit (Elekta Inc., Sweden). B: A Varian TrueBeam™ unit (Varian Oncology Systems, Palo Alto, CA). Both
linear accelerators have a kV imaging system orthogonal to the therapy beam direction. Both systems provide 2D radiographic, fluoroscopic,
and CBCT modes.

LWBK933_Ch14_p229-258.indd 232 6/1/11 6:43 PM


Image-Guided Radiation Therapy  n 233

slightly larger flat panel detector (41 by 41 cm), compared 4 MV. The CT detector uses an array of 738 channel xenon
to Varian’s detector (40 cm wide by 30 cm long), which limits ion chambers and an FOV of 40 cm can be reconstructed.
the scan length to 15 cm when using the full-fan scanning There is no limitation in the superior–inferior direction.
mode. A “shifted detector” (or half-fan) technique can extend Owing to the use of MV beam for imaging, MVCT
the axial field of view (FOV) to at least 40 cm (28). To further image quality is not as high as that from a diagnostic CT
increase the FOV with the existing imager size, Li et al. has image: the noise level is higher and low-contrast resolution
recently proposed an “off-axis” ellipse cone-beam scanning is less. Nevertheless, the MVCT images provide sufficient
method, which shifts the center of rotation during CBCT to contrast to verify patient position (37) and to delineate
increase the sampling outside regular FOV, while sacrificing many anatomic structures (38,39). It is interesting to note
the sampling in the center (29). that the MVCT numbers are linear with respect to the elec-
Limitations of CBCT image quality include elevated tron density of material imaged, which yields reliable and
x-ray scatter, which reduces image contrast and introduces accurate dose calculations (40). This capability, together
cupping artifacts. Scatter can be reduced by using post- with the strong research effort in treatment plan optimiza-
processing methods (Elekta’s solution) (30) or anti-scatter tion and image registration by the TomoTherapy group
grids (Varian’s solution). Because of regulations on gantry (41–43), makes tomotherapy appealing as an integrated
rotation speed (maximum 1 rpm), CBCT image quality is solution for image-guided adaptive radiotherapy.
adversely affected by the breathing motion. The IGRT
setup process may add 5 minutes (1–2 min scan time and MV Cone-Beam CT
1–3 min for image registration and approval) to the regu- Megavoltage cone-beam CT (MV CBCT) uses the elec-
lar treatment schedule. tronic portal imaging device (EPID) mounted on a linac
gantry and the therapy MV x-ray as a basic configuration
MV Helical CT (Tomotherapy) for CT imaging. The initial attempt was performed by
Tomotherapy (TomoTherapy Inc., Madison, WI) is an inte- Swindell et al. (44) and subsequently implemented by
grated technology that combines a helical megavoltage CT Mosleh-Shirazi et al. (45). With recent advance in highly
(MVCT) with a linear accelerator, which is specially sensitive EPIDs, particularly a-Si flat panel detectors (46–48),
designed for delivering intensity-modulated radiation in a it has become possible to rapidly acquire multiple, low-
slit geometry. The concept was originally proposed by dose 2D projection images with current treatment
Mackie et al. (31,32) and substantially developed by the machines. Similar to kV CBCT described in previous sec-
University of Wisconsin group (33–37). Helical tomother- tions, a CBCT imaging system uses a large image detector
apy refers to the continuous gantry and couch motion, and a single rotation of the x-ray source to complete the
which, as shown in Figure 14.5A, resembles the motion image acquisition. In addition to the MV x-ray source, the
from a conventional helical CT scanner. Low-dose, typically use of a large-area receptor (EPID) with no effective MV
1 to 2 cGy, pretreatment MVCT images are obtained from scatter-reduction mechanism limits image quality. The
the same treatment beam line but with a nominal energy of amount of scatter reaching the detector depends on the

A B
Figure 14.5.  A: A picture of tomotherapy unit (TomoTherapy Inc., Madison, WI) (Photograph courtesy of H. Ning, PhD). Tomotherapy is an
integrated IGRT system, which combines a linear accelerator with an MVCT image guidance system. B: A Siemens MV CBCT imaging system
using a conventional linac and a flat-panel EPID. Source: Reprinted from Morin O, Gillis A, Chen J, et al. Megavoltage cone-beam CT: system
description and clinical applications. Med Dosim 2006;31:9.

LWBK933_Ch14_p229-258.indd 233 6/1/11 6:43 PM


234 n  Treatment Planning in Radiation Oncology

photon energy, field size, and the thickness of the imaged CBCT is that the physics of absorbed dose from a thera-
object; however, the imaging system can be optimized by peutic MV beam is well understood, thus enabling accu-
calibrating the system using standard site-specific calibra- rate calculation of dose from an MV CBCT scan that could
tion phantoms (49,50). be included in the treatment plan (51). Soft-tissue contrast
Figure 14.5B shows a prototype MV CBCT system from is the principal limiting factor of MV CBCT systems.
Siemens. The system consists of a standard treatment unit Investigators at the University of California San Francisco
(Primus™, Siemens Medical Solutions, Concord, CA) and have used a lower-Z target material, which generates more
an a-Si flat-panel adapted for MV photons on a retractable low-energy photons and enhances image contrast (49).
support. The 41 × 41 cm flat-panel x-ray detector (AG9-ES, One study showed that removal of the flattening filter
PerkinElmer, Optoelectronics) consists of a 1 mm copper improved contrast by 200% (52).
plate and a Kodak Lanex fast scintillation plate (Gd2O2
S:Tb) overlaid on a light-sensitive and charge-integrating Digital Tomosynthesis
thin-film transistor (TFT) array. Due to a more stringent Digital tomosynthesis (DTS) is a special situation of
requirement for geometric integrity, a calibration proce- tomographic reconstruction, which uses limited arc (20 to
dure is required to correct for mechanical support sag 40 degrees) of projection images (53–55). The reconstruc-
(49,50). The image acquisition lasts ∼45 seconds with gan- tion planes are approximately perpendicular to the direc-
try rotation over a 200-degrees arc, at a rate of one image tion of the x-ray beam at the arc center. They do not have
per degree. Image reconstruction time is less than 2 min- to go through the isocenter, but can be at different depths
utes for a 256 × 256 × 270 image volume (0.7 mm3 voxel in the patient. In some methods, reconstruction is fol-
size). Imaging dose ranges from 2 to 8 cGy (51). lowed by a procedure to suppress out-of-plane objects
The MV CBCT system has good image quality for bony (56,57). The principal advantage of DTS over CBCT is the
structures and acceptable quality for soft-tissue targets, shorter scan time, which is typically less than 10 seconds.
due to the higher imaging dose. An advantage of both MV DTS provides soft-tissue image visualization, which can be
CBCT and MVCT systems is the reduced influence of improved in respiratory disease sites by combining with
implanted metal objects on image quality, in contrast to breath-hold or respiration correlation of the projection
kV CT, which exhibits strong artifacts when high-Z mate- images (58).
rial is present. MVCT images thus provide complementary Depending on the acquisition arc length, spatial resolu-
information, which cannot be discerned in conventional tion is less in the direction perpendicular to the recon-
CT images (Fig. 14.6). An additional advantage of MV structed planes, which can be addressed by acquisition of
a second DTS from a quasi-orthogonal direction to the
first. Reference DTS images can be calculated from the
planning CT, similar to the calculation of DRRs (59). A
comparison of the conventional CT, CBCT, and DTS
images of the same patient is shown in Figure 14.7.

Hybrid Cone-Beam CT
Combining kV and MV projection images for CBCT recon-
struction has been reported (60). A hybrid CBCT can be
achieved by combining orthogonal kV and MV x-ray pro-
jection images with a partial arc gantry rotation as little as
90 degrees (61). The resultant projection images span an arc
length of 180 degrees. Acquisition requires only 15 seconds,
making it optimal for breath-hold imaging.

4D Computed Tomography Imaging


Respiratory motion is one of the important factors in the
management of radiation therapy for moving tumors,
which can have displacements up to 3 to 4 cm (62). CT
scans acquired synchronously with the respiratory signal
can be used to reconstruct a set of (3D) CT scans, repre-
Figure 14.6.  Images showing the artifacts due to the presence of metal senting the 3D anatomy at different times (or respiratory
objects in the conventional kV CT images (left panels). Artifact-free
images were obtained with a MV CBCT (right panels). Source:
phases). This collection of 3D CT data sets is called respi-
Reprinted from Morin O, Gillis A, Chen J, et al. Megavoltage cone-beam ration-correlated CT (RCCT), or 4D CT, which describes
CT: system description and clinical applications. Med Dosim 2006; the snapshots of patient’s 3D anatomy over a periodic
31:11. respiratory signal. Typically, the breathing cycle is divided

LWBK933_Ch14_p229-258.indd 234 6/1/11 6:43 PM


Image-Guided Radiation Therapy  n 235

Figure 14.7.  A coronal planar image reconstruction based on conventional kV CT (left), CBCT (middle), and digital tomosynthesis (DTS, right)
(Photograph courtesy of Fang-Fang Yin, PhD).

into 10 respiratory phases. Four-dimensional medical CT images are sorted based on a respiration phase
imaging techniques has been reviewed recently (63). angle assigned by an algorithm that determines the peri-
odic behavior of the respiratory signal. Phase-based sort-
Respiration-Correlated CT ing assumes repeatable breathing cycles, that is, that the
The approach common to RCCT methods is to acquire anatomy is at same position for a given phase in every
sufficient data for generating CT images at all phases of cycle, which is usually not the case for normal breathing
the respiratory cycle while simultaneously recording res- with its varying cycle-to-cycle amplitude variations, and
piration, then retrospectively correlating the CT images has led to investigations into sorting based on the dis-
with phase. CT acquisition uses either a cine or helical placement of the respiratory signal (71,74–76).
mode. In cine mode, repeat CT slices are acquired over
Respiration-Correlated CBCT
slightly more than one respiratory cycle with the couch
stationary while recording patient respiration; the couch A limitation of CBCT systems is image degradation caused
is then incremented and the process repeated. Following by patient motion artifacts, which is a consequence of the
acquisition, the images are sorted with respect to the respi- limited gantry speed and the resultant long (∼1  min)
ratory signal, leading to a set of volume images at different acquisition times. Respiratory motion in particular
respiration points in the cycle (64,65). Helical acquisition degrades images in the thorax and abdomen. Different
uses a low pitch and adjusting the gantry rotation period methods are under active investigation to reduce motion
such that all voxels are viewed by the CT detectors for at in CBCT. One approach is respiration-correlated CBCT
least one respiratory cycle (66,67). RCCT images have (RC-CBCT) using retrospective sorting of projection
been widely characterized and applied clinically for esti- images into different breathing phases (77,78). A slower
mating the extent of a moving tumor in lung (68–70). gantry rotation is required to acquire sufficient projec-
The selection of the type of respiratory signal can vary, tions in each phase bin, resulting in scan times of 3 to
which may change the interpretation of the RCCT images; 6 minutes. The limited number of projections per phase
however, a periodic motion is always implied. Deviations reduces the contrast resolution and introduces image view
from the regular periodic motion will have an impact on artifacts; thus, the method is more suited to detecting
the quality of RCCT images and on the accuracy of anat- high-contrast objects such as tumor in parenchymal lung
omy discerned from these images. Commercial systems (77–79).
commonly use one of the two types of respiratory moni- An alternative approach is to process the CBCT images
tors. One such monitor (Real-time Position Management, to correct for motion, using a motion model of the patient
RPMTM, Varian Oncology Systems, Palo Alto, CA) cap- (80–84). Most of the methods make use of deformable
tures the up/down motion of a box with infrared reflectors image registration (DIR) to deform the images to a com-
placed on the patient’s abdomen or chest using an infrared mon motion state.
camera. The other is a “pneumo bellows” system (Phillips
Medical Systems, Milpitas, CA) that records the digital Non-radiographic Imaging
voltage signal from a differential pressure sensor wrapped
around the patient’s abdomen. RCCT scans can be used in Ultrasound Imaging
so-called 4D treatment planning to explicitly account for Ultrasound is a noninvasive, non-radiographic, and real-time
tumor motion (63,68,70,72,73). imaging technique for soft-tissue targeting in radiotherapy,

LWBK933_Ch14_p229-258.indd 235 6/1/11 6:44 PM


236 n  Treatment Planning in Radiation Oncology

particularly for prostate cancer, but also for upper abdom- and corresponding CT images (bottom row) for a prostate
inal malignancies (85–89). Fung et al. have recently cancer patient undergoing ultrasound-guided setup (91).
reviewed ultrasound guidance for IMRT (90). In this example, the agreement between ultrasound and CT
The ultrasound transducer, which is made of the piezo- alignments is within 2  mm. It has been shown that the
electric crystal, is both a sound source and a sound detec- inter- and intra-user variability is large for ultrasound-
tor. An ultrasound transducer transmits brief pulses of guided setup, mainly because of the poor image quality,
ultrasound that propagate into the tissues. Whenever there anatomic distortion due to pressure variation and inade-
is a change in acoustic impedance, for example, owing to quate user training (86,92). When compared with
the tissue density or elasticity changes at the interface of implanted fiducial markers for prostate localization, the
two organs, some of the ultrasound will be reflected back ultrasound alignment displayed even more variations (93)
to the transducer as echoes. The round-trip time from the and a larger planning margin is recommended (94). Dobler
pulse transmission to reception of an echo is used to deter- et al. studied the displacement of prostate when acquiring
mine the transducer-to-object distances. A scan line con- transabdominal ultrasound images (95). X-ray simulations
verter will construct a 2D image of the patient, in which the were performed before and during ultrasound image
amplitude of echoes varies along depth direction. When acquisition for 10 patients who had undergone iodine-125
sweeping the ultrasound through a volume, a 3D ultra- seed implantation. The seeds, which were visible in x-ray
sound image can be constructed. The basic principles of images, were used to represent prostate position. A maxi-
this imaging technique limit its use to soft-tissue structures mum displacement of the prostate of 2.3 mm in anteropos-
and tumors in pelvic, abdominal, and breast locations. terior and 1.9  mm in craniocaudal direction and a
Although ultrasound has been widely used in patient rotational change of up to 2.5 degrees were observed. If the
setup, inherently poor image quality and the unfamiliar system was not handled correctly and too much pressure
appearance of ultrasound images for radiation therapists was applied, a shift of the prostate of up to 10 mm could be
have limited its potential for more precise image guidance. induced. Tome et al. has reported on ultrasound commis-
Figure 14.8 shows a pair of ultrasound images (top row) sioning and QA procedure for radiation therapy (96).

Figure 14.8.  A pair of ultrasound images is shown in the top row. The images were acquired from an ultrasound-guided alignment system
(BAT™, NOMOS, Chatsworth, CA). A pair of CT images of the same patient is shown in the bottom row. The CT images were acquired using
an in-room CT-on-rails scanner within 5 minutes of the ultrasound image acquisition. The alignment relative to the planning CT by these two
independent systems was within 2 mm.

LWBK933_Ch14_p229-258.indd 236 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 237

Stereoscopic Optical Surface Imaging in the solid phase. A limitation for radiotherapy applica-
Stereoscopic optical surface imaging (OSI) provides real- tions is that an MR image cannot be easily converted to
time imaging capability, primarily used in patient setup electron density, which is essential for dosimetry calcula-
for superficial tumors, such as the breasts, or immobile tion in a treatment planning system. Recently, MRI-based
tumors such as in the head. One currently available OSI treatment planning has been reported (102–104).
device (AlignRTTM, VisionRT, Ltd., London, UK) is com- Several MRI-guided radiotherapy systems are currently
posed of two to three ceiling-mounted stereo-camera under development. Such systems offer not only in-room
pods, each contains two cameras and a speckle pattern soft-tissue-based target alignment, but also near real-time
projector. The triangulation between the fixed stereo cam- MRI volumetric imaging for tumor motion monitoring.
eras and a skin point identified by the speckle pattern The section “MRI Real-Time Volumetric Imaging” discusses
serves to calculate their distances and the location of the these systems in more detail.
point in space. A patient skin surface image is recon-
structed from the visible surface points. The accuracy of
the surface imaging is within 1.0 mm. Intra-Fractional Real-Time Imaging
Because OSI does not visualize the tumor, it requires and Motion Compensation
substantial validation against another established image
modality, such as radiographic imaging. Early studies of The main goal of real-time tracking is to minimize the
surface imaging in radiotherapy have been reported by effect of target motion not only between treatments, but
Massachusetts General Hospital (97) and Johns Hopkins also during a treatment fraction. Real-time tracking usu-
University (98) in 2005. Validated with x-ray imaging, OSI ally requires real-time motion monitoring (detection) and
provides a quick and non-radiologic means for image- the execution of correction with the shortest time delay. In
guided setup. It has been applied in breast, lung, brain, and almost all cases, implanted markers are used as surrogates
head and neck. The common procedure for patient setup for target position. Therefore, the proximity of the mark-
is to register the surface image to a reference region of ers to the target and their motion relative to the target are
interest (ROI) defined on the delineated patient surface in important factors in their reliability for a real-time track-
a simulation CT. ing system. In the following sections, we review different
approaches for real-time monitoring and tracking.
Magnetic Resonance Imaging
Fluoroscopic Imaging with Implant Fiducials
MRI is based on the radiofrequency signal from the relax-
ation process of the dipole moment of an atom with Commercial fluoroscopy-based tracking systems can be
unpaired proton, such as 1H in tissue, after it is excited categorized into (1) room-mounted and (2) linac gantry-
with a radiofrequency pulse sequence in the presence of an mounted systems. The primary reason for using fluoro-
external magnetic field. The field strength is ∼0.2 Tesla (T) scopic imaging is to detect metal fiducials, which are
for open-field MRI and 1.5 T or 3 T for a regular MRI implanted in or near the tumor. The fluoroscopic images
scanner. The geometric location of the signal is deter- can be matched to a reference DRR.
mined using the field gradient, phase and frequency A very interesting, room-mounted system for real-time
encoding. The soft-tissue heterogeneity provides an envi- tracking has been developed by Shirato et al. in collabora-
ronment that alters the relaxation process, thus providing tion with Mitsubishi at the University of Hokkaido
high soft-tissue contrast. (105,106). It uses a pair of x-ray tubes that rotate on a
MRI imaging is more versatile than radiologic imaging, circular track embedded in the floor, as shown in Figure
as it provides different image appearances when different 14.9A. Each tube has a corresponding x-ray detector that
pulse sequences are applied, such as T1-weighted, rotates synchronously on a ceiling-mounted track, so as to
T2-weighted, and fast fluid-attenuation inversion recovery avoid obstruction of the patient by the treatment gantry.
(FLAIR) (99). These MRI images are often used in delin- The x-ray tubes allow pulsed imaging interlaced with linac
eating a tumor or postoperation cavity and evaluating pulses to treat the patient. The radio-opaque marker
local edema, especially for brain cancer. MRI can also pro- images are tracked with pulsed fluoroscopy prior to the
duce non-axial scans in any spatial orientation. Like radio- beginning of the treatment and patient is repositioned so
logic imaging, MRI can produce 2D planar, 3D volumetric, that the end of one of the tracks corresponding to the least
and 4D volumetric set images (63). Four-dimensional mobile portion of the motion is within a predefined gating
prospective volumetric MRI imaging has been utilized in window. During irradiation, fluoroscopy continues and the
radiotherapy (100). beam is automatically switched on when the detected
MRI may suffer from geometric distortion due to non- image of the fiducial is within the window; otherwise, it is
uniformity of the external magnetic field strength. This turned off.
scanner-specific factor can be corrected by measuring the A commercially available room-mounted system, as
field gradient with a large grid phantom (101). MRI does shown in Figure 14.9B, is developed by BrainLab (ExacTrac™,
not produce a visible image in bone due to little relaxation BrainLAB AG, Feldkirchen, Germany). The ExacTrac is an

LWBK933_Ch14_p229-258.indd 237 6/1/11 6:44 PM


238 n  Treatment Planning in Radiation Oncology

A B
Figure 14.9.  Two room-mounted kV image-guided IGRT real-time tracking systems. A: A real-time tracking radiation therapy (RTRT) system
(Photograph courtesy of Hiroki Shirato, MD, PhD). B: ExacTrac system, BrainLAB AG, Feldkirchen, Germany (ExacTrac® is a trademark of
BrainLAB AG in Germany and the United States.)

integrated IGRT system for target localization, setup the immobilization device or the couch. The kV-MV con-
correction, and the delivery of high-precision stereotactic figuration provides a possibility to acquire images during
radiotherapy and stereotactic radiosurgery. The image treatment (111). To avoid scatter interference between the
guidance utilizes two distinct imaging subsystems: a real- two x-ray beams, alternating the beam-on time may be
time infrared (IR) tracking and a kV stereoscopic x-ray used (113).
imaging subsystem. Two ceiling-mounted IR cameras are A prototype kV-MV dual in-line imaging system is
used to monitor the movement of infrared-reflecting mark- commercially available (ArtisteTM, Siemens Medical Sys-
ers placed on patient’s skin or on the reference frame tems, Germany) (113,114). This is achieved by placing the
mounted on the treatment couch. The marker images are kV x-ray tube in a retractable lower shelf. The kV image
automatically compared to stored reference, generating ini- detector is mounted just below the MV collimator. The
tial couch shift instruction to set up the patient. The x-ray MV therapy beam must penetrate the kV image detector
imaging system performs further internal target alignment to treat the patient. This design allows for simultaneous
based on either bony landmarks or implanted fiducial imaging of the beam shaping device (MLC-collimated
markers. The reference DRRs are provided by the BrainLab beams) and patient’s anatomy for beam-by-beam verifica-
treatment planning system. During treatment delivery, the tion of treatment delivery or motion compensation.
IR tracking system and the fluoroscopic x-ray imaging sys-
tem work together to monitor target position and to per- Optical Fiducial Motion Surrogates
form treatment interventions. The external fiducial markers
can be “tuned” to the internal fiducial markers during a pre- Optical tracking determines an object’s position by mea-
treatment verification procedure. Two types of treatment suring light either emitted or reflected from the object.
interventions can be performed: adaptive gating of the The hallmark of optical tracking systems is their high spa-
treatment beam or real-time correction of target offset by tial and temporal resolutions (0.1 mm and <0.1 s, respec-
using a 6D robotic couch (ExacTrac™, BrainLAB AG, Feld- tively) in tracking infrared markers attached to the
kirchen, Germany). CyberKnifeTM system (Accuray Inc., patient’s external surface. The positions of the optical
Sunnyvale, CA) has also its own room-mounted stereo- markers relative to the target volume, together with the
scopic x-ray imaging system and 6D robotic couch. Research desired marker positions relative to the treatment isocen-
and clinical experience of using these systems have been ter, are determined during CT simulation. In the treat-
reported by various groups (107–110). ment room, the real marker positions are measured
Gantry-mounted kV imaging systems usually have only relative to isocenter; a rigid-body transformation deter-
one kV x-ray imager and achieve the orthogonal pair by mines marker displacements from their desired positions
rotating the gantry. These systems include Varian’s On- and hence target displacement from the isocenter. Real-
board Imager (OBI) and Elekta’s Synergy. These kV imag- time feedback allows one to correct the patient’s position
ing beam lines are orthogonal to the MV treatment beam continuously during treatment. Meeks et al. have reviewed
line (EPID), sharing the same isocenter of gantry rotation, the technology in several implementations for intracranial
as shown in Figure 14.4. The gantry-mounted kV imaging and extracranial stereotaxis radiotherapy (115). The first
system is not blocked by the treatment unit, but it may systems used rigid arrays of optical markers that were
have limited clearance or the potential for collision with attached to the patient via a biteplate linkage. Subsequent

LWBK933_Ch14_p229-258.indd 238 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 239

systems for extracranial radiotherapy tracked external


markers to determine the patient position and/or gate the
radiation beam based on patient motion.

Video-Based 3D Optical Surface Imaging


Real-time 3D-surface matching has been characterized for
clinical use (97,98,116). The reference surface image is
either a 3D optical surface image acquired at simulation or
a CT/MRI-surface rendering from the treatment plan. The
real-time 3D surface images are captured by stereoscopic
video cameras mounted on the ceiling of the treatment
vault. A fast automatic alignment between the real-time
surface and the reference surface using a modified iterative-
closest-point method leads to an efficient and robust sur-
face-guided target refixation. Experimental and clinical
results demonstrate the excellent efficacy of <2 minutes
set-up time and high accuracy and precision of <1 mm in
isocenter shifts and <1 degree in rotation (116).
The AlignRT system has been described in a previous
section for patient alignment. In addition, it offers capa-
bilities for real-time motion monitoring. Due to pro-
cessing time required for image reconstruction, it only
provides prospective imaging at one to five frames per
second (fps), far below its maximum capture rate
Figure 14.10.  A diagram showing the prototype AC electromagnetic
(∼15 fps), (117) depending upon the size and resolution field tracking system with the detector array and the infrared cameras
of ROI. Several investigators have shown that this real- (Calypso Medical Technologies, Inc., Seattle, WA) The Beacon™ tran-
time surface imaging capability can be applied to head sponder is shown in the inset.
motion monitoring during frameless stereotactic radio-
therapy or radiosurgery (118,119). Using a 3D optical
surface image acquired at treatment as reference, sys- MRI Real-Time Volumetric Imaging
tematic errors of the imaging system can be cancelled
out and achieve a sub-millimeter accuracy for motion Another innovative technology being developed for radia-
detection. tion therapy applications is an integrated MRI-guided
gamma ray IMRT system (Renaissance System™, ViewRay,
Real-Time Electromagnetic Inc., Gainesville, FL). A schematic diagram of the IG-
Localization and Tracking IMRT system is shown in Figure 14.11. The system con-
sists of a low-field open MRI system and three Cobalt
Continuous tracking of target position without ionizing irradiation sources; the latter was chosen for its compati-
radiation is possible with a novel technology utilizing AC bility with the MRI system. The computerized multileaf
electromagnetic fields to induce and detect signals from collimator system provides gamma-ray intensity modula-
implanted “wireless” transponder (Calypso Medical Tech- tion equivalent to linac-based IMRT systems. The thrust
nologies, Inc., Seattle, WA). The system consists of a console, of the technology is the MRI-guided, real-time volumetric
optical tracking system, and a tracking station. The console imaging system, which can track patient’s 3D anatomy
instrument is situated in the treatment room and includes a every 0.5 to 2.0 seconds without interrupting the treat-
display screen and the AC magnetic array (Fig. 14.10). The ment delivery. As stated earlier, MRI has superior soft-
magnetic array is lightweight and contains source coils tissue contrast, which can be used for better definition and
that generate signals to excite the transponders, and sensor tracking of target volumes. As an integrated imaging,
coils that detect the unique response signals returned by adaptive treatment planning and dynamic delivery system,
each transponder. Unlike passive fiducial markers, the this promising technology is perhaps the only “near” real-
Calypso system can actively detect the position of tran- time, volumetric soft-tissue targeting system. Because of
sponders without using the radiographic method. The the known limits of the Cobalt source, such as low energy
Calypso 4D Localization System can update target posi- and large penumbra, the plan quality is compromised.
tion 10 times per second, which is sufficiently fast to track Recently, there has been development of integrated MRI-
breathing motion of the tumor. Sub-millimeter tracking linac systems, thus combining the advantages of high soft-
accuracy and clinical experience in prostate target local- tissue discrimination for tracking and linac quality radiation
ization has been reported (120–124). treatment (125–127). There are two designs, with the

LWBK933_Ch14_p229-258.indd 239 6/1/11 6:44 PM


240 n  Treatment Planning in Radiation Oncology

Figure 14.11.  A schematic of a prototype MRI-guided real-time volumetric tracking system for IGRT (Renaissance System™, ViewRay, Inc.,
Gainesville, FL). The system is designed to have a low-field open MRI for real-time imaging and three-headed Cobalt source for intensity-
modulated gamma ray irradiation (Photograph courtesy of James Dempsey, PhD).

radiation beam either parallel or perpendicular to the mag- Mobile Treatment Couch Approach
netic field. A nontrivial technical task is to shield the mag- D’Souza et al. have proposed compensation of the tumor
netic field from the linac in transporting electrons to the motion using a mobile couch (131). Unlike the DMLC
target. In addition, the presence of magnetic field affects the approach, this method compensates for 3D tumor motion
motion of the scattered secondary electron scatters, thereby with isotropic spatial resolution; however, there may be
distorting the dose distribution. Nevertheless, integrated patient-related physical and medical concerns. For
MRI-linac systems promise high-precision radiation dose instance, couch motion could induce a counterreaction
delivery guided by high-quality soft-tissue visualization. from the patient, body shift, or tissue deformation when
changing motion directions, especially for obese patients.
Real-Time Tumor Motion Compensation Clinically, a patient may feel dizziness or nausea. Recently,
a dynamically adjustable head support was developed for
Real-time tracking refers to continuous adjustment of the repositioning and for potentially tracking head motion in
radiation beam or patient position during treatment so as to 6D (translations and rotations) with sub-millimeter accu-
follow the changing position of the tumor or its surrogate. A racy in frameless stereotactic radiotherapy (132,133). As
means of localizing the target in real time is coupled to the the head is fairly well immobilized with a thermoplastic
repositioning control system. In principle, real-time tracking mask and/or head mold, the motion is slow and the
provides more efficient treatment than gating, in that the motion range is on the order of 1 to 3 mm. Therefore, the
beam duty cycle is at or near 100%, while reducing the mar- compensating motion for repositioning would be small
gin needed for target motion. In the following sections, we and slow, posing little or no clinical concerns.
summarize three strategies in various stages of development
involving motion tracking of a linac system. Movable Gantry Approach
The CyberKnifeTM (Accuray Inc., Sunnyvale, CA) is an
Dynamic Multi-leaf Collimator Approach intelligent robotic IGRT system consisting of a small
Keall et al. have demonstrated motion tracking using X-band linear accelerator mounted on a 6D robotic arm
dynamic MLC (DMLC) (128). The Calypso electromag- and a 6D robotic couch for patient alignment. The robotic
netic system can provide a near real-time signal of prostate arm can move at a speed of several centimeters per second,
motion with <2 mm accuracy and 220 ms system latency which allows it to keep up with breathing-induced tumor
(123). One concern of this motion compensation motion. The room-mounted x-ray stereoscopic guidance
approach is the anisotropic spatial resolution. Depending system provides IGRT setup as well as real-time tracking
on whether the motion is along or perpendicular to the for image-guided radiosurgery applications, as depicted in
leaf motion, the spatial resolution is either <1 mm (the Figure 14.12A. An optical tracking system of IR reflectors
leaf motion accuracy) or 5 mm (the leaf width), respec- on a vast worn by the patient provides a real-time target
tively. The mechanical motion limitations of MLC pose motion signal that is confirmed by periodic x-ray imaging.
constraints when DMLC is used simultaneously for The CyberKnife is the first clinical radiotherapy system to
motion tracking and IMRT delivery. Investigators have use real-time motion compensation and has accumulated
examined different strategies to optimize leaf trajectories a wealth of data on the experience of tracking moving tar-
for this purpose (129,130). gets in a wide variety of treatment scenarios (134–136).

LWBK933_Ch14_p229-258.indd 240 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 241

A B
Figure 14.12.  A CyberKnife system (Accuray Inc., Sunnyvale, CA) is shown in (A). A VERO system (BrainLAB AG Feldkirchen, Germany and
Mitsubishi Heavy Industries, Tokyo, Japan) is shown in (B) (Photograph courtesy of Dirk Verellen, PhD). Both systems offer quick gantry movement
aiming to a moving tumor, guided by room-mounted or gantry-mounted stereoscopic x-ray imaging systems.

Clinical experience in frameless image-guided cranial task group (TG) reports, covering in-room kV x-ray imag-
(137,138), spinal (139–141), pancreatic (142), and lung ing for patient setup and target localization (TG #104)
radiosurgery (143,144) has been reported. (148), medical accelerator QA (TG #142) (149), stereotac-
The VEROTM system is a joint development by BrainLAB tic body radiation therapy (TG #101) (150), and manage-
AG (Feldkirchen, Germany) and Mitsubishi Heavy Indus- ment of respiratory motion (TG #76) (62). These reports
[AQ4] tries (Tokyo, Japan). It is based on a gimbal design, as shown provide guidelines for clinical use and QA of the IGRT
in Figure 14.12B, such that the radiation gantry can rotate imaging systems and procedures. In the following, we
quickly to track a moving tumor (145–147). For both pan- summarize three important aspects: geometric accuracy,
ning and tilting rotations, the maximum motion range at image quality, and motion detection.
treatment isocenter is 4.4 cm (or 2.5 degrees). The response
lag time is <50 ms for both panning and tilting rotations. Clinical “Gold Standard” for IGRT
With system lag compensated by using a predicting model,
sub-millimeter accuracy of motion tracking has been One of most important tasks in commissioning an in-room
achieved in initial tests (147). One prototype, installed in imaging modality is to compare the imaging isocenter with
UZ Brussels Hospital, the Netherlands, is equipped with an the treatment isocenter. Traditionally, it is paramount to
EPID for portal imaging and two orthogonal kV imaging check the alignment of radiation isocenter, mechanical iso-
systems that are 45 degrees from the MV beam. CBCT capa- center, and laser isocenter. When using IGRT imaging
bility is available and patient position corrections are pro- modalities, their isocenter coincidence with the treatment
vided by a BrainLAB 5DOF robotic couch that is integrated radiation isocenter must be initially and periodically
in the system. Although more characterization studies of checked to assure a clinically acceptable tolerance on their
the system are needed, initial tests have shown its potential discrepancy. The MV EPID is used as the gold standard as it
for tumor motion tracking. provides direct reference to the treatment beam line. For
stereotactic procedures, the discrepancy should be within
1.0 mm; otherwise, it should be within 2.0 mm (149). Due
Igrt Requirements and Considerations to the stringent geometric accuracy requirement for the
alignment of the OBI and EPID detectors, a calibration pro-
IGRT Commissioning and Quality Assurance cedure is required to correct the mechanical sagging
(27,49,50,151). Customized QA phantoms have been devel-
Commissioning and QA of all IGRT-enabled technologies oped for different IGRT systems, including kV and MV
are essential. In the past few years, the American Associa- imaging systems of a linac (149,152) and MVCT imaging
tion of Physicists in Medicine (AAPM) has issued several system of a helical tomotherapy (153,154).

LWBK933_Ch14_p229-258.indd 241 6/1/11 6:44 PM


242 n  Treatment Planning in Radiation Oncology

Imaging Quality QA registration of the tumor position is commonly accepted.


Bissonnette and coworkers have established QA program for Visual verification, together with manual adjustment, is
CBCT imaging quality with the Elekta Synergy and Varian necessary for automatic registration. Most visual verifica-
OBI systems. The description includes flat-panel detector tion is performed in three orthogonal planar views using
stability, performance and image quality of 10 volumetric color blending, checkerboard, split windows, and differ-
imaging systems over a period of 3 years (155). Details on ence images. A 3D volumetric visualization to seek most
correcting background (dark current) and pixel-by-pixel homogeneous color distribution of mono-color images on
gain uniformity (flood image) of the plat-panel detector are a volumetric anatomical landmark has been reported with
discussed. Similar to diagnostic imaging QA, the CatPhan greater spatial accuracy for rigid anatomy (163,164). Image
500 phantom (The Phantom Laboratory, Salem, NY) is used registration method QA should be performed using an
to quantify image quality (156,157). A comprehensive QA appropriate phantom. A QA procedure for image registra-
program by Yoo et al. covers safety and functionality, geo- tion and segmentation has been reported (165).
metric accuracy, and image quality for the Varian OBI sys-
tem (158). Image quality characterization and QA procedures Rigid Image Registration
for EPID (159), MV CBCT (160), and MVCT in helical Most registration tools, which are used for image-guided
tomotherapy (154) have also been reported. patient setup, belong to rigid image registration with up to
Non-gantry mounted x-ray imaging and non-radiolog- six degrees of freedom (6DOF or 6D). Owing to limitation
ical imaging have been reviewed by Bissonnette (155), of the standard treatment couch, three or four DOF are
including the commissioning of image-guided technolo- usually employed, unless a 6D robotic couch is available.
gies and procedures, QA of geometric accuracy and imple- As patient motion and deformation is inevitable, rigid
mentation of a clinical IGRT QA program. A stereotactic image registration has limited accuracy. In addition, inter-
head phantom (Model 605 Radiosurgery Head Phantom; observer variation for manual image registration is well
CIRS, Norfolk, VA) or equivalent is used for imaging QA known. The registration accuracy, couch adjustment accu-
of the CyberKnife system (155,161). racy, kV-MV isocenter discrepancy, and couch walk for
non-coplanar beams determine the overall setup accuracy
Motion-Related QA Processes for tumor localization.
In AAPM TG #142 (149), x-ray (MV and kV) 2D radio- Locally focused rigid image registrations are more clin-
graphic imaging, 3D tomographic (fan-beam and cone- ically relevant in the presence of tissue deformation.
beam) imaging, and 4D imaging are included as part of Zhang et al. (166), Van Kranen et al. (167), and Giske et al.
the QA process for medical accelerators. For 2D fluoro- (168) have reported using multiple ROIs for rigid registra-
scopic imaging, the temporal resolution should be 100 ms tion and evaluated the local deformation among the three
or less, which will produce a uncertainty <2  mm for a to nine different ROIs in the head and the neck region.
moving object at a speed no greater than 2 cm/s. Jiang et al. Park and coworkers have developed a spatially weighted
have laid out major clinical QA challenges in respiratory- image registration method to allow users to define the
related procedures, including respiratory gating, breath structure of interest, such that a better registration result
holding, and 4D CT imaging (162). As external surrogates is achieved with respect to the structure of interest (169).
are used in most respiratory gating and breath holding, Manual intervention of the semiautomatic registration
the authors conclude that the biggest challenge is how to using visual guidance is always required.
ensure treatment accuracy in these cases. Indeed, many
reports have discussed the unreliability of the internal– Deformable Image Registration
external correlation using external fiducials, such as IR DIR is not suitable for setup correction with simple couch
reflectors for motion monitoring. When external respira- shifts, as rigid transformation cannot compensate for tis-
tory fiducial markers are used, a pretreatment calibration sue deformation. Nevertheless, DIR is essential for evalu-
and during-treatment verification against x-ray radio- ating cumulative delivered dose distribution, and therefore
graphic imaging is needed. Clinical motion management necessary for adaptive actions to compensate for potential
guideline has been published in AAPM TG #76 (62), dosimetric deviation from what was planned in the
covering respiration-induced motions of the target and remaining treatment fractions. DIR has been intensively
normal tissue. studied (42,170–173) and is an important tool in present-
ing developments of high-precision radiotherapy. Indeed,
Image Registration many clinical studies using DIR have been reported since
2007, including automatic segmentation and dose distri-
Image registration quantitatively aligns the daily 3D or 2D bution mapping for adaptive radiotherapy (174–179).
setup images with the planning CT volume or DRR images, In the IGRT context, DIR plays a significant role in the
respectively. Such image registrations can be suboptimal as following three areas. First, it can track a deformed anat-
the underlying anatomy changes, including daily motion, omy voxel by voxel from deformed images to a reference
deformation, or physical changes. Clinically, the focus on image, producing a deformation transformation matrix,

LWBK933_Ch14_p229-258.indd 242 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 243

Figure 14.13.  Automated image segmentation of multiple repeat CT data sets. In this head-and-neck example, contours drawn manually on the
planning CT were deformed to obtain contours for repeat CT scans obtained during the course of radiotherapy. Deformations were carried out
using transformation matrices based on deforming planning CT image to match each of the repeat CT images. Such automatic segmentation
tools, once validated by clinical studies, would make adaptive replanning practical.

which is useful in studying tumor and organ motion. ated treatment, which is a basis for image-guided adaptive
Zhang et al. have illustrated that the combination of DIR radiotherapy (184–186).
with principal component analysis (PCA) provides a
patient-specific motion model (180). Such a DIR-PCA
approach has been further developed to predict lung tumor Information Technology Infrastructure for IGRT
location based on single x-ray projection image (181).
With the increasing use of highly conformal treatment
Second, the deformation transformation matrix can be
techniques, such as IMRT, VMAT, and SBRT, the need for
used for mapping segmented anatomic ROIs, such as
image guidance is mounting at every stage of the radio-
delineated tumor and organs at risk, from a reference
therapy process. Implementation of IGRT into routine
image to other deformed images. Such an auto-contour-
clinical workflow requires tighter integration of imaging
ing technique is essential for 4D and adaptive radiother-
and treatment systems and more efficient information
apy, as illustrated in Figure 14.13. Wijesooriya et al. have
flow. IGRT represents a shift from a traditionally static
studied the accuracy of the automated segmentation
treatment planning process to a more dynamic, close-loop
among different phase CT images in 4D CT by comparing
practice with multiple feedback check/control points. To
692 pairs of automated and physician-drawn contours.
meet the technical and logistical needs for this dynamic
The surface congruence of the gross tumor volume (GTV)
treatment process, the following infrastructure and soft-
and all organs was within 5 mm in >90% of all cases (178).
ware tools are considered important to IGRT applications:
Wang et al. have studied the use of deformable registration
to propagate contours in daily CBCT for both lung and ■ IGRT Data Management
head-and-neck cancer patients, and found the volume ■ Radiotherapy Picture Archival and Communication
overlap index to be 83% between the deformed and the System (RT-PACS): Further development is needed
physician-drawn contours (175). Physician evaluation of to meet the evolving needs of new IGRT workflows
these automated contours is highly recommended, espe- and extra data requirements using efficient central-
cially in the presence of motion and metal artifacts. ized communication among different imaging and
Third, the deformation transformation matrix can be treatment procedures.
applied to map a dose distribution that is calculated based ■ Digital Imaging and Communications in Medicine
on static volumetric CT images to a reference volumetric (DICOM) and its radiotherapy (RT) extension are
image (128,182). At the end, a cumulative dose distribu- industrial standards for facilitating connectivity and
tion can be determined for evaluating delivered radiation interoperability of medical image data. Advance-
dose using daily setup images and for 4D planning based ments in IGRT have revealed shortcomings in the
on all phase CTs within a 4D CT image set (183). In IGRT, DICOM-RT standard and the need to address them.
the principal advantage of DIR is to track radiation doses The development of new-generation objects is being
deposited in a deformed organ over the course of fraction- addressed by a DICOM working group for radiation

LWBK933_Ch14_p229-258.indd 243 6/1/11 6:44 PM


244 n  Treatment Planning in Radiation Oncology

therapy objects (187). Compatibility of DICOM and v iii. Magnitude of dose gradients
DICOM-RT files among different vendors may need ix. Available treatment capacity and patient load (treat-
further attention. ments/hour)
■ Record and Verify (R&V) server: With the more fre- x. Identification of individuals responsible for mainte-
quent use of volumetric image acquisition and pos- nance and development
sible adaptive replanning during the course of
treatment, data storage requirements can increase 1
to 2 orders of magnitude. It is also essential to have Igrt Correction Strategies
mirrored servers, robust data backup mechanism, and Applications
and a disaster recovery strategy for routine, continu-
ous clinical operations (188). Online Versus Off-line Corrections
■ IGRT Facilitating Tools:
■ DIR: Both rigid and DIR tools are necessary for The development of various mechanisms for measuring
implementing various IGRT approaches at different patient position has created a wealth of valuable data for
levels to correct for geometric and dosimetric varia- correction of patient position. The use of these data to
tions (171,176,189,190). stratify treatment decision and to modify the treatment
■ Fast treatment planning and optimization algo- process is referred to as a correction strategy. Strategies are
rithms: With the increasing need for treatment plan broadly divided into online and off-line approaches. The
adaptation to the changed anatomy or altered target online approach makes adjustment to the treatment
volumes, it becomes necessary to develop fast treat- parameters or patient position based upon data acquired
ment planning and optimization strategies (191). during the current treatment session. This may be as sim-
■ Cross-platform planning comparison tools: The ple as adjusting the couch position or as complex as full
computational environment for radiotherapy reoptimization of the treatment parameters based on
research (CERR), developed by Deasy and cowork- changes in the shape and relative position of target and
ers, provides a common platform for creating multi- normal structures. The off-line approach is one in which
institutional IMRT treatment plan database, the intervention is determined from an accumulation of
including adaptive plans. This also provides a plat- information that may be drawn from previous treatment
form for data mining in the planning database for sessions or other times of measurement. The online
clinical outcome research and analysis (192–194). approach is generally categorized as having a greater
capacity to increase precision with an associated increase
in effort for the same level of accuracy as can be achieved
Selection of IGRT Technology with off-line strategies, but at the cost of a higher work-
load. In general, clinical implementations typically oper-
The selection of an appropriate image-guidance solution
ate with a hybrid of online and off-line approaches that
is a complex process that is a compromise of clinical
are invoked under different error thresholds. A familiar
objective, product availability, existing infrastructure, and
example is seen in conventional portal film practice in
manpower (195,196). The deployment of a new technol-
which the first treatment session is adjusted “online” (at
ogy requires a thorough understanding of the complete
the time of therapy), while subsequent corrections are
clinical process and the necessary infrastructure to sup-
applied off-line (physician review of portal images). For a
port data collection, analysis, and intervention. The AAPM
detailed review of the numerous strategies in clinical use,
TG #104 report (148) suggests four aspects of consider-
the reader is referred to recent reviews in the literature
ations, clinical, technical, resource, and administration,
(6,7,146,191,197).
when choosing an IGRT solution. We believe that these
More complex IGRT procedures, which employ increased
considerations will evolve, depending on industry trend
frequency of imaging, alignment tools, and decision rules,
and economics. Ten key points are as follows:
offer potentially increased accuracy compared to conven-
i. Clinical objectives (dose escalation/normal tissue tional practice. The overhead associated with the align-
sparing) ment tools and decision rules can be prohibitive unless
ii. Structures of interest (target and normal structures) properly integrated. The adaptive radiotherapy program
iii. Strength of surrogates (skin markers, bony land- at William Beaumont Hospital (184–186,198,199) was
marks, implanted fiducials, etc.) made possible only through in-house software integration
iv. Desired level of geometric precision (radiosurgery/ efforts. Similarly, effort was also required from the proce-
hypofractionation) dure and policy perspective to coordinate the actions of
v. Uncertainties to be managed through the use of the off-line efforts with the radiation therapists operating
margins the machine. Online approaches require elevated levels of
vi. Method of intervention/correction (degrees of software and hardware integration for operation as the
freedom) analysis and interpretation are performed at the time of
vii. Techniques of managing tumor motion therapy.

LWBK933_Ch14_p229-258.indd 244 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 245

Figure 14.14.  An in-room volumetric CT-


guided radiotherapy process. CT images of
patient’s setup and anatomy information are
acquired and sent to an alignment workstation
where the images are compared and aligned to
match with the planning CT. An interventional
decision is made based on the magnitude of ana-
tomic variations to assess the need for an online
or off-line correction. If necessary, dose tracking
may be enabled and used for replanning.

Correction for Inter-fractional Setup Error lung and liver cancers (63,146,198). To setup a patient
with a moving tumor, marker imaging may be used by
Inter-fractional variations are usually larger than intra- aligning the track of the implanted marker, or a portion
fractional ones; therefore, various techniques have been thereof, with the track discerned from the reference pre-
developed for pretreatment setup corrections. Without treatment 4D CT. Alternatively, bony landmarks may be
loss of generality, we consider an in-room CT-guided used for setup alignment. However, the tumor motion tra-
IGRT system (Fig. 14.14). In the diagram, 3D volumetric jectory relative to the bony landmark may change, and
CT (CT-on-rails or CBCT) images are acquired after the recent studies have shown that the inter-fractional tumor
patient is immobilized and the skin marks aligned with location has significant variation (205,206).
room lasers, then sent to an alignment workstation where Real-time monitoring of implanted markers, or of the
the images are registered with the planning CT. The first tumor directly, is needed for accurate “gating” of radiation
level of intervention is always the correction of transla- treatments. Respiratory monitoring and control devices
tional shifts. Usually, rotation has smaller effect than the used for imaging may also be used to trigger the beam and
translational shifts of the target, and can be corrected the MLC motion on and off during the delivery of gated
using a 6D couch with isocentric rotation. Depending on radiation treatments. The radiation beam is automatically
the magnitude of the differences observed, an off-line cor- switched on only during a small, generally most reproduc-
rection protocol may be used to reduce systematic uncer- ible portion of the breathing cycle. In gated IMRT, leaf
tainties while improving operation efficiency (200–202). motion is also correspondingly stopped and resumed,
The second level of intervention may be dose based. In thereby avoiding an interplay effect (207). Similar to imag-
theory, the ultimate treatment goal should be based on the ing, video feedback may be used to improve breathing or
final delivered dose distributions, instead of what was breath-hold regularity and reproducibility (208). The
originally planned using the initial planning CT. In-room same methodology may also be used for “gated breath-
CT images of the patient’s treatment anatomy allows hold” treatments in which the patient voluntarily or invol-
reconstruction of the delivered dose distributions and untarily holds his or her breath at the same point in the
therefore dose tracking. Cumulative dose distributions breathing cycle. In voluntary breath-hold, the beam is
can be corrected infrequently using an off-line adaptive turned on only if the breath is held at the same point.
correction scheme (191,199,203,204). Reproducible involuntary breath-hold may be achieved
using technology such as active breathing control (ABC)
Management of Intra-fractional Tumor Motion developed by Wong et al. (209,210).
There are considerations regarding implantation of fidu-
In the presence of significant intra-fractional motion, cial markers for lung cancers. Implantation is invasive and
additional geometric and dosimetric variations should may cause clinical complications. Marker images do not pro-
also be included. This will set the complexity to another vide 3D anatomic information on tumor shape changes.
level. There are many different approaches in the manage- Variations of the normal anatomy are ignored altogether. In
ment of intra-fractional tumor motion. Not surprisingly, addition, experience to date shows that markers may get lost
most of these applications are related to the treatment of and drift appreciably over the course of radiotherapy. Ideally,

LWBK933_Ch14_p229-258.indd 245 6/1/11 6:44 PM


246 n  Treatment Planning in Radiation Oncology

prospective volumetric imaging would be desirable for both (217). A proper patient immobilization improves setup
geometric guidance and dosimetric assessment, such as 4D reproducibility and reduces voluntary patient motion,
volumetric MRI integrated with a linac (125–127). Never- therefore decreases systematic and random errors. Margin
theless, implanted markers may be ideal for tracking targets is also dependent on the motion-management technique
during treatments, and often serve as a gold standard for employed. For involuntary respiratory motion, breath-
verification of the target location. hold, motion compression, motion gating, or tracking will
Real-time tumor motion tracking has been implemented compensate for tumor motion at different levels. As a con-
with the CyberKnife unit with guidance of both x-ray imag- sequence, the margin will be reduced, such that it is
ing of implanted fiducial markers and an optical external smaller than a motion-encompassing internal tumor vol-
motion surrogate (211–213). For other types of linacs, such ume (ITV) (184,218–220). The ability to reduce margin by
capability is still at the development stage. Zimmerman using image guidance will therefore decrease the normal
et al. have demonstrated motion tracking with intensity- tissue toxicity and increase dose acceleration, particularly
modulated arc therapy (214). Depuydt et al. have demon- important for SBRT.
strated sub-millimeter beam tracking accuracy in a In prostate cases, image-guided margin reduction is
phantom study using a gimbals-based linac unit (147). one of most dramatic examples in all anatomic sites. With
in-room CT guidance, a 3-mm margin was reported ade-
Population-Based and Individualized Margins quate for prostate dose coverage, but may lose some of the
seminal vesicles coverage due to daily variation in rectal
The relative importance of systematic and random errors filling that causes local deformation (221). A comparative
in the determination of PTV margins should be considered study using four different IGRT setup methods, skin
in the design of the clinical strategy. Geometric errors in marks, 3D bony landmarks, 3D fiducial markers, and
radiation field placement are typically characterized by dis- Calypso transponders, has shown that the last two meth-
tributions of nonzero mean and variance. The mean ods can achieve 4  mm and 3  mm margin requirement,
describes the systematic discrepancy for an individual respectively (222). Another study has compared four setup
patient and the variance of the random component. Several techniques using skin marks, 2D bony registration, ultra-
authors have highlighted the relative importance of these sound guidance, and in-room 3D CT (223). It has pro-
two categories of errors in determining appropriate PTV posed a correction for patient-specific systematic shifts
margins (215,216). These conclusions are not completely after the bony landmark alignment, and provides equiva-
general, as the number of fractions is an important factor in lent dose coverage to ultrasound guidance. A recent
determining the relative importance of these two categories Calypso study has shown that a margin of 2 mm would
of error, especially for SBRT cases with less than five fractions. produce sufficient CTV dose coverage based on 1,267
A treatment margin depends not only on the imaging tracking sessions of 35 patients (224). Figure 14.15A
modality chosen and tumor surrogate used, but also on demonstrates that the alignment accuracy increases along
the patient immobilization dedicated for a treatment with the complexity of the alignment technology: from

A B
Figure 14.15.  A: Patient setup accuracy of prostate cancer using different in-room imaging modalities. Generally, the accuracy increases with imaging
frequency, dimension, and use of fiducial. The skin mark and ultrasound setup have largest variation, as indicated by the error bars. The margin could
[AQ8] be reduced from 8 to 2 mm based on this finding. Source: Reprinted with permission from Semin Radiat Oncol 2007;17:268–77. B: Target coverage
based on various types of image-guided setups for treatment. In this example, 24 treatment-time CT scans of a prostate cancer patient were used to
compare the effectiveness of four alignment techniques for patient setup using a fixed-margin IMRT plan. The minimum target dose is lowest
(59.3 Gy) for skin marks-based setup and highest (76.0 Gy) for the CT-guided setup. The day-to-day variations in the minimum dose (represented
by the error bars) are smallest for CT-guided technique and largest for skin-mark and ultrasound-guided techniques.

LWBK933_Ch14_p229-258.indd 246 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 247

skin to bone to ultrasound and to CT (6). The dosimetric Anatomic Variations and
result (prostate underdose) for one patient exhibiting rel- Dosimetric Consequences
atively large organ motion is shown in Figure 14.15B.
In head and neck cases, an IGRT assessment of setup Inter- and Intra-fractional Variations in Anatomy
error for 225 patients suggests that 3 mm margin is appro- Typical inter- and intra-fractional organ variations and
priate (225). The major variation for this anatomical site setup uncertainties of gynecological tumors, liver, dia-
comes from deformation. Depending on tumor location, phragm, lung tumors, prostate, seminal vesicles, bladder,
locally focused image registration is needed to minimize and rectum have been reviewed by Langen et al. (228) and
the setup uncertainty due to deformation (166–168). Booth et al. (229). Both publications have reported sig-
In lung cases, both inter- and intra-fractional motion nificant variations for all organs studied. Even with careful
affect setup accuracy, and the overall margin would be immobilization and alignment of the patient, significant
the sum of the two terms: regular setup margin plus the changes occur because of the nonrigidity of anatomy,
ITV. Yan and coworkers proposed a margin formula that bowel gas movement, and the variable filling of the blad-
contains both components of population-based and der (230–233). Li et al. have reported inter-factional ana-
individualized systematic and random errors for lung tomic variations for all major sites based on daily CT
cancer (217,226). Such a margin formula was applied in assessment (234). Target and normal structure variations
clinical cases and 3D CT-guided setup resulted in a 65% can also follow certain trends, including volume shrinkage
to 75% margin reduction. To compensate for the base- up to 12 months after the initial hormone treatment (235),
line drift, Pepin et al. have reported using a dynamic gat- and soft-tissue changes such as tumor shrinkage and
ing window (228). As the tumor motion is location and weight loss. Figure 14.16A shows a side-by-side compari-
patient specific and 4D CT imaging provides the means son of a head and neck target volume that has shrunk sig-
to measure the patient-specific ITV, individualized nificantly during the course of treatment. The skin
tumor motion margin is often applied in lung cancer contour no longer matches well with the immobilization
treatment (217). mask. Changes in target volume and position of normal

Figure 14.16.  A: An example of setup error


for a patient immobilized with a thermo-
plastic facemask due to tumor shrinkage as
treatment progresses. Approximately half
way through the treatment course (right
panel), the lower neck was not centered on
the headrest, presumably due to the rela-
tively “roomier” mask. B: Dosimetric
impact of inter-fractional variations in
head and neck anatomy. The solid lines
show the volumes of the parotid glands (left
and right) decreased as the treatment pro-
gressed. At the same time, the centers of
both parotid glands also moved medially
due to tumor shrinkage and weight loss. As
a result, the percent of parotid volume
exceeding 26 Gy increased by least 10% over
the course of radiotherapy. B

LWBK933_Ch14_p229-258.indd 247 6/1/11 6:44 PM


248 n  Treatment Planning in Radiation Oncology

Figure 14.17.  Intra-fractional varia-


tions of anatomy observed in a pros-
tate patient in the span of 20 minutes.
CT images were acquired just prior
and immediately after an IMRT
treatment fraction. The contours of
pelvic anatomy before treatment
(left) are overlaid on the CT image of
the patient acquired immediately
after the treatment (right). Prostate
target (red) was displaced anteriorly
for >5 mm.

critical organs (such as the spinal cord) could have sig- finds that the mean change in target D95% is 1 ± 4% and
nificant clinical consequences (166,167). In a prostate the average cumulative effect is smeared out after five frac-
IMRT treatment, changes in bladder filling can cause tions. In individual fractions, the D95% may be off by as
prostate and surrounding organs to move away from the high as 20%. For normal tissues, such as the rectum, Chen
planning position, as demonstrated in Figure 14.17. Dur- et al. have reported that dose variation is significant due to
ing a 20- to 30-minute IMRT treatment delivery, intra- daily variation of the rectal volume and 27% of actual
fractional motion may cause significant dose deviation for treatment would benefit from adaptive replanning. The
a session. authors recommend an empty rectum for both planning
and treatment to avoid such adverse effects (239). Any
Dosimetric Effects due to Inter-fractional Motion effort to make patient conditions reproducible is worth-
The common approach to evaluating the dosimetric effect while so as to treat as planned.
of a delivered dose is by applying the treatment plan to the In breast cases, Goddu et al. have demonstrated the
daily setup 3D CT images with the corresponding setup potential benefit of daily MVCT setup by simulating the
isocenter shift. The actual dynamic leaf sequence can also integrated dose distribution without image-guided setup
be retrieved from a treatment log file for synchronized correction. Significant dose reduction in the PTV is
dose reconstruction (236). To generate a cumulative dose observed mostly at the lung interface where steep dose
distribution over multiple fractions, DIR is applied to map falloff is expected (240). The possible adverse dosimetric
the daily dose distribution to a reference image for final effect is anisotropic with regard to the steep dose falloff.
dose evaluation (178). For highly conformal head and neck IMRT treatments,
In prostate cases, it is reported that 25% (8/33) of it is desirable to reduce the dose to the parotid glands in
patients would have geometric or dosimetric miss without order to minimize the incidence of late xerostomia (241).
daily MVCT improve tumor localization (237). Obese Unfortunately, the parotid glands can decrease in volume
patients and patients with large daily rectal motion would and move medially during the course of treatment (242).
be most subject to such marginal miss. van Herk has As a result, parotid dose can increase by 10% and exceed
pointed out that the systematic uncertainty is more 26 Gy, as illustrated in Figure 14.16B, in which one patient
important and should be minimized as much as possible received repeat CT imaging during his course of IMRT
(215). Langen et al. (238) have investigated the dosimetry treatment. A single mid-course correction to adapt the
consequences of prostate motion during helical tomother- treatment plan to the anatomical change could help reduce
apy for 16 patients with 515 daily MVCT scans. The study the dose for both parotid glands (243).

LWBK933_Ch14_p229-258.indd 248 6/1/11 6:44 PM


Image-Guided Radiation Therapy  n 249

B
Figure 14.18.  A: Potential consequence of respiratory motion on target coverage. An IMRT plan, developed using conventional
CT, was applied to the patient’s 4D CT. Dose distributions were calculated in each of the 10 phases of the breathing cycle. A
portion of the CTV, shown in thick yellow line, was not covered by the 70 Gy prescription dose line (red) in phases 1 through
4. B: Comparison of a treatment plan as perceived on a free-breathing CT (top row) and as realized after accounting for breath-
ing motion in all 10 phases (bottom row). The latter was obtained by summing dose distributions computed on individual
phases of the 4D CT image (A), and mapped to a reference CT image using deformable image registration (DIR).

Dosimetric Effects of Intra-fractional Motion the entire breathing cycle shows a dose deficiency in the
The dosimetric consequence of intra-fractional breathing CTV (red arrow), as illustrated in the bottom row of
motion for lung tumors can be demonstrated by 4D CT Figure 14.18B. In this case, the cumulative dose distribution
images. Figure 14.18A shows a case study that used a free- when using the internal target volume (ITV) derived from
breathing CT image to design a treatment plan with an the 10-phase 4D CT does not underdose the target but
inadequate 8-mm margin to cover the CTV (shown in yel- results in treatment to a larger volume. The ITV method
low). The actual dose distribution does not cover the is an example of using an individualized margin to account
entire target volume in some of the breathing phases due for target motion in treatment planning. The dosimetric
to respiratory motion, which is not detected in the free- impact of motion in free breathing and gated radiotherapy
breathing CT. Using DIR, the cumulative dose distribution of lung cancer has been studied using Monte Carlo dose
from the 10 individual phases is calculated and mapped to calculation (244). The doses are similar between free
a free-breathing fast CT scan (near full exhalation, #7). breathing and all phases, except for the end-inspiration
The resultant cumulative dose distribution summed from phase in which the tumor is underdosed by ∼10%.

LWBK933_Ch14_p229-258.indd 249 6/1/11 6:45 PM


250 n  Treatment Planning in Radiation Oncology

Wu et al. have compared three delivery techniques, high-grade toxicity with organ motion (1–3). Well-
3DCRT, IMAT, and IMRT, in five treatment cases in liver, designed clinical trials are needed to demonstrate its
with tumor motions ranging from 0.5 to 1.75  cm. The advantages fully. We believe that further advance in IGRT
variation in D95% is largest (−8.3%) for IMRT and smallest relies on the innovation and integration of multiple auto-
(<2%) for 3DCRT, with negligible dose–volume histo- mated technologies to facilitate extra evaluations and
gram variations for normal tissues (178). Kuo et al. have additional processes. Parallel computing technologies,
found that with an adequate margin for motion, D95% such as graphics processing unit (GPU) technology (181),
variation is <2% in the CTV (72). is a solution for increasing real-time performance, espe-
cially for intra-fractional motion management and online
Adaptive Approaches for Correcting dosimetric replanning. In motion monitoring, non-
Dosimetric Deviations radiographic alternatives may be more clinically desir-
William Beaumont Hospital has pioneered the adaptive able, such as in-room MRI imaging with near real-time
radiotherapy strategy by using a purpose-built treatment volumetric information (62). In off-line and online dosi-
planning system to facilitate off-line dosimetric evaluation metric replanning, automated and integrated systems are
and replanning (185,199). Without the support from a necessary to make such actions clinically feasible. The
more automated planning system, routine replanning is treatment efficacy and benefit of real-time tracking and
not feasible under current clinical conditions. Special staff online adaptation must be evaluated with clinical evi-
effort may only be reserved for those patients with poten- dences, such as improved potency of hypo- or single-
tially severe dosimetric consequence (245). Mageras and fractioned SBRT.
Mechalakos have discussed treatment planning in the In image-guided treatment planning, biological imag-
IGRT context and the various challenges to treatment- ing with different molecular probes will play an increasing
plan adaptation strategies in various disease sites (6). An role in reducing the uncertainties in delineation of gross
alternative treatment planning approach to evaluating a and clinical tumor volumes. Different molecular probes
PTV is to simulate motion and other uncertainties directly for detecting different biological attributes of cancer may
in the dose calculation. A benefit of this approach is that prove useful in accurately defining the biological tumor
the resultant dose distributions of not only the CTV, but volume, as shown in Figure 14.19. Various molecular trac-
also organs at risk can be examined. ers are available for probe tumor metabolism, prolifera-
Recent articles have reviewed clinical applications of tion, hypoxia, and angiogenesis. The concept of
adaptive radiotherapy for various treatments (191). The multiplexed imaging used in diagnosis to simultaneously
adaptive concept as applied to radiotherapy practice probe different molecular targets may be adopted by the
derives from modern informatics and control theory. Off- therapeutic clinic as theragnostic imaging (250) to pre-
line adaptation has been implemented in various institu- scribe the distribution of therapeutic dose (8). Micro-
tions (246–248), and online replanning has been reported, scopic cancerous cells spreading beyond the GTV requires
with computation time within 5 to 8 minutes (249). The histopathological assessment and accurate registration
knowledge gained from geometric and dosimetric varia- with 3D anatomic image.
tions via clinical IGRT research will be useful for guiding In image-guided treatment evaluation, multi-modality
the treatment planning process to be more cautious in imaging must be applied to assess the complex radiation
certain clinical scenarios. response. This will remain an intense clinical research arena
to improve our fundamental understanding of the radiation
response at molecular, cellular, organ functional, and phys-
Future Directions iological levels. Proper biological attributes, such as DNA
double-strand breaks, need to be identified and probed.
Image-guided radiotherapy is commonly considered in Ideally, response assessment within the treatment course
the context of treatment delivery verification, but it is would be most beneficial for individualized treatments,
more appropriate to broaden its scope to include imaging while the reality is lack of an effective assessment index even
at other stages of the radiotherapy process (8). We, there- after treatment. A response-driven, biologically adaptive
fore, briefly discuss future directions as they apply to this radiotherapy is still distant from clinical practice. The dosi-
broader definition. metric feedback loop, which is within the reach to ensure
In the application of IGRT to treatment delivery, we that the treatment process goes along the intended course
have better understanding of various uncertainties, cor- (6,191), can provide more reliable clinical data to tune pre-
rection strategies, and technical limitations. Geometri- diction models of treatment outcome.
cally, a large body of evidence has shown the improved Radiation therapy has gone through a series of techno-
accuracy in patient setup and motion management. Dosi- logical revolutions following several breakthroughs in
metrically, IGRT improves treatment delivery in certain imaging in the past three decades. We have witnessed the
plans that involve sharp dose gradients or a moving growth of IGRT, which has provided improved geometric
target. Clinically, increasing evidence has unveiled the and dosimetric accuracy in radiation therapy of localized
connections of local failure with marginal miss and cancers. We believe that there are more technologic

LWBK933_Ch14_p229-258.indd 250 6/1/11 6:45 PM


Image-Guided Radiation Therapy  n 251

Figure 14.19.  Imaging paradigm in radiation oncology hypothetically illustrating the possibility of integrating different
functional imaging modalities (panels A–D) with CT-defined gross tumor volume (GTV) (panel E) to obtain a combined
biological target volume (BTV) (panel F). Panel A: Metabolism (i.e., FDG); panel B: proliferation (i.e., FLT); panel C: hypoxia
(i.e., Cu-ATSM); and panel D: angiogenesis (i.e., MMP). Improved tumor target coverage and/or dose escalation to these
physiological sub-regions may be achievable. A region of increased angiogenesis can be seen in panel F that lies outside other
biological regions. This may represent a potential area of recurrence that should be included into the overall BTV. Lines
(yellow) outlining the anatomical GTV (panel E) and BTV (panel F) accentuate the change in target volume delineation.

advances to come at all levels of IGRT, closing the physical, 5. Li JG, Xing L. Inverse planning incorporating organ
biological, and clinical feedback loop for radiation motion. Med Phys 2000;27:1573–1578.
therapy. 6. Mageras GS, Mechalakos J. Planning in the IGRT
context: closing the loop. Semin Radiat Oncol 2007;17:
268–277.
References 7. van Herk M. Different styles of image-guided radio-
therapy. Semin Radiat Oncol 2007;17:258–267.
1. Eisbruch A, Harris J, Garden AS, et al. Multi-institutional 8. Greco C, Clifton Ling C. Broadening the scope of image-
trial of accelerated hypofractionated intensity-modulat- guided radiotherapy (IGRT). Acta Oncol 2008;47(7):
ed radiation therapy for early-stage oropharyngeal cancer 1193–1200.
(RTOG 00–22). Int J Radiat Oncol Biol Phys 2010;76(5): 9. van Herk M, Meertens H. A matrix ionisation chamber
1333–1338. imaging device for on-line patient setup verification
2. de Crevoisier R, Tucker SL, Dong L, et al. Increased risk of during radiotherapy. Radiother Oncol 1988;11:369–378.
biochemical and local failure in patients with distended 10. Boyer AL, Antonuk L, Fenster A, et al. A review of elec-
rectum on the planning CT for prostate cancer radiother- tronic portal imaging devices (EPIDs). Med Phys 1992;19:
apy. Int J Radiat Oncol Biol Phys 2005;62:965–973. 1–16.
3. Tucker SL, Jin H, Wei X, et al. Impact of toxicity grade 11. Bijhold J, Gilhuijs KG, van Herk M. Automatic verifica-
and scoring system on the relationship between mean tion of radiation field shape using digital portal images.
lung dose and risk of radiation pneumonitis in a large Med Phys 1992;19:1007–1014.
cohort of patients with non-small cell lung cancer. Int J 12. Dong L, Boyer AL. A portal image alignment and patient
Radiat Oncol Biol Phys 2010;77:691–698. setup verification procedure using moments and corre-
4. Mageras GS, Kutcher GJ, Leibel SA, et al. A method of lation techniques. Phys Med Biol 1996;41:697–723.
incorporating organ motion uncertainties into three- 13. Van Elmpt W, McDermott, L, Nijsten S, et al. A literature
dimensional conformal treatment plans. Int J Radiat review of electronic portal imaging for radiotherapy
Oncol Biol Phys 1996;35:333–342. dosimetry. Radiother Oncol 2008;88:289–309.

LWBK933_Ch14_p229-258.indd 251 6/1/11 6:45 PM


252 n  Treatment Planning in Radiation Oncology

14. Faddegon BA, Aubin M, Bani-Hashemi A, et al. Com- 30. Siewerdsen JH, Daly MJ, Bakhtiar B, et al. A simple,
parison of patient megavoltage cone beam CT images direct method for x-ray scatter estimation and correc-
acquired with an unflattened beam from a carbon target tion in digital radiography and cone-beam CT. Med
and a flattened treatment beam. Med Phys 2010;37(4): Phys 2006;33:187–197.
1737–1741. 31. Mackie TR, Holmes T, Swerdloff S, et al. Tomotherapy:
15. Shirato H, Shimizu S, Kunieda T, et al. Physical aspects of a new concept for the delivery of dynamic conformal
a real-time tumor-tracking system for gated radiotherapy. radiotherapy. Med Phys 1993;20:1709–1719.
Int J Radiat Oncol Biol Phys 2000;48:1187–1195. 32. Mackie TR, Balog J, Ruchala K, et al. Tomotherapy.
16. Uematsu M, Fukui T, Shioda A, et al. A dual comput- Semin Radiat Oncol 1999;9:108–117.
ed tomography linear accelerator unit for stereotactic 33. Olivera GH, Shepard DM, Reckwerdt PJ, et al. Maximum
radiation therapy: a new approach without cranially likelihood as a common computational framework in
fixated stereotactic frames. Int J Radiat Oncol Biol Phys tomotherapy. Phys Med Biol 1998;43:3277–3294.
1996;35:587–592. 34. Lu W, Fitchard EE, Olivera GH, et al. Image/patient regis-
17. Yenice KM, Lovelock DM, Hunt MA, et al. CT image- tration from (partial) projection data by the Fourier phase
guided intensity-modulated therapy for paraspinal matching method. Phys Med Biol 1999;44:2029–2048.
tumors using stereotactic immobilization. Int J Radiat 35. Ruchala KJ, Olivera GH, Schloesser EA, et al. Megavolt-
Oncol Biol Phys 2003;55:583–593. age CT on a tomotherapy system. Phys Med Biol 1999;44:
18. Hua CH, Lovelock DM, Mageras GS, et al. Development 2597–2621.
of a semi-automatic alignment tool for accelerated local- 36. Ruchala KJ, Olivera GH, Schloesser EA, et al. Calibra-
ization of the prostate. Int J Radiat Oncol Biol Phys 2003; tion of a tomotherapeutic MVCT system. Phys Med Biol
55:811–824. 2000;45:N27–N36.
19. Wong JR, Cheng CW, Grimm L, et al. Clinical imple- 37. Mackie TR, Jaradat H, Hui S, et al. The utility of mega-
mentation of the world’s first Primatom, a combination voltage computed tomography images from a helical
of CT scanner and linear accelerator, for precise tumor tomotherapy system for setup verification purposes. Int
targeting and treatment. Phys Med 2001;17:271–276. J Radiat Oncol Biol Phys 2004;60:1639–1644.
20. Fung AY, Grimm SY, Wong JR, et al. Computed tomog- 38. Meeks SL, Langen KM, Willoughby TR, et al. Perform-
raphy localization of radiation treatment delivery versus ance characterization of megavoltage computed tomog-
conventional localization with bony landmarks. J Appl raphy imaging on a helical tomotherapy unit. Med Phys
Clin Med Phys 2003;4:112–119. 2005;32:2673–2681.
21. Court L, Rosen I, Mohan R, et al. Evaluation of mechanical 39. Kupelian PA, Meeks SL, Willoughby TR, et al. Serial
precision and alignment uncertainties for an integrated megavoltage CT imaging during external beam radio-
CT/LINAC system. Med Phys 2003;30:1198–1210. therapy for non-small-cell lung cancer: observations on
22. Shiu AS, Chang EL, Ye JS, et al. Near simultaneous tumor regression during treatment. Int J Radiat Oncol
computed tomography image-guided stereotactic spi- Biol Phys 2005;63:1024–1028.
nal radiotherapy: an emerging paradigm for achieving 40. Langen KM, Meeks SL, Poole DO, et al. The use of meg-
true stereotaxy. Int J Radiat Oncol Biol Phys 2003;57: avoltage CT (MVCT) images for dose recomputations.
605–613. Phys Med Biol 2005;50:4259–4276.
23. Jaffray DA, Drake DG, Moreau M, et al. A radiograph- 41. Shepard DM, Olivera GH, Reckwerdt PJ, et al. Iterative
ic and tomographic imaging system integrated into a approaches to dose optimization in tomotherapy. Phys
medical linear accelerator for localization of bone and Med Biol 2000;45:69–90.
soft-tissue targets. Int J Radiat Oncol Biol Phys 1999;45: 42. Lu W, Chen ML, Olivera GH, et al. Fast free-form
773–789. deformable registration via calculus of variations. Phys
24. Jaffray DA, Siewerdsen JH, Wong JW, et al. Flat-panel Med Biol 2004;49:3067–3087.
cone-beam computed tomography for image-guid- 43. Zhang T, Jeraj R, Keller H, et al. Treatment plan opti-
ed radiation therapy. Int J Radiat Oncol Biol Phys mization incorporating respiratory motion. Med Phys
2002;53:1337–1349. 2004;31:1576–1586.
25. Siewerdsen JH, Moseley DJ, Jaffray DA, et al. The influ- 44. Swindell W, Simpson RG, Oleson JR, et al. Computed
ence of antiscatter grids on soft-tissue detectability tomography with a linear accelerator with radiotherapy
in cone-beam computed tomography with flat-panel applications. Med Phys 1983;10:416–420.
detectors. Med Phys 2004;31:3506–3520. 45. Mosleh-Shirazi MA, Evans PM, Swindell W, et al. A cone-
26. Feldkamp IA, Davis LC, Kress JW. Practical cone-beam beam megavoltage CT scanner for treatment verification
algorithm. J Opt Soc Am A 1984;1:612–619. in conformal radiotherapy. Radiother Oncol 1998;48:
27. Fahrig R, Holdsworth DW. Three-dimensional computed 319–328.
tomographic reconstruction using a C-arm mounted 46. Groh BA, Siewerdsen JH, Drake DG, et al. A perform-
XRII: image-based correction of gantry motion nonide- ance comparison of flat-panel imager-based MV and
alities. Med Phys 2000;27:30–38. kV cone-beam CT. Med Phys 2002;29:967–975.
28. Cho PS, Rudd AD, Johnson RH. Cone-beam CT from 47. Ford EC, Chang J, Mueller K, et al. Cone-beam CT with
width-truncated projections. Comput Med Imaging Graph megavoltage beams and an amorphous silicon electronic
1996;20:49–57. portal imaging device: potential for verification of radio-
29. Li T, Li X, Yang Y, et al. A novel off-axis scanning method therapy of lung cancer. Med Phys 2002;29:2913–2924.
for an enlarged ellipse cone-beam computed tomography 48. Sillanpaa J, Chang J, Mageras G, et al. Developments in
field of view. Med Phys 2010;37:6233–6239. megavoltage cone beam CT with an amorphous silicon

LWBK933_Ch14_p229-258.indd 252 6/1/11 6:45 PM


Image-Guided Radiation Therapy  n 253

EPID: reduction of exposure and synchronization with 67. Vedam SS, Keall PJ, Kini VR, et al. Acquiring a four-
respiratory gating. Med Phys 2005;32:819–829. dimensional computed tomography dataset using an
49. Morin O, Gillis A, Chen J, et al. Megavoltage cone-beam external respiratory signal. Phys Med Biol 2003;48:45–62.
CT: System description and clinical applications. Med 68. Keall P. 4-dimensional computed tomography imaging
Dosim 2006;31:51–61. and treatment planning. Semin Radiat Oncol 2004;14:
50. Pouliot J, Bani-Hashemi A, Chen J, et al. Low-dose 81–90.
megavoltage cone-beam CT for radiation therapy. Int J 69. Mageras GS, Pevsner A, Yorke ED, et al. Measurement
Radiat Oncol Biol Phys 2005;61:552–560. of lung tumor motion using respiration-correlated CT.
51. Morin O, Gillis A, Descovich M, et al. Patient dose con- Int J Radiat Oncol Biol Phys 2004;60:933–941.
siderations for routine megavoltage cone-beam CT 70. Rietzel E, Chen GT, Choi NC, et al. Four-dimensional
imaging. Med Phys 2007;34:1819–1827. image-based treatment planning: target volume seg-
52. Faddegon BA, Aubin M, Bani-Hashemi A, et al. Com- mentation and dose calculation in the presence of
parison of patient megavoltage cone beam CT images respiratory motion. Int J Radiat Oncol Biol Phys 2005;
acquired with an unflattened beam from a carbon tar- 61:1535–1550.
get and a flattened treatment beam. Med Phys 2010;37: 71. Lu W, Parikh PJ, Hubenschmidt JP, et al. A comparison
1737–1741. between amplitude sorting and phase-angle sorting
53. Kolitsi Z, Panayiotakis G, Anastassopoulos V, et al. A using external respiratory measurement for 4D CT. Med
multiple projection method for digital tomosynthesis. Phys 2006;33:2964–2974.
Med Phys 1992;19:1045–1050. 72. Kuo HC, Mah D, Chuang KS, et al. A method incorpo-
54. Zwicker RD, Atari NA. Transverse tomosynthesis on a rating 4DCT data for evaluating the dosimetric effects of
digital simulator. Med Phys 1997;24:867–871. respiratory motion in single-arc IMAT. Phys Med Biol
55. Godfrey DJ, Yin FF, Oldham M, et al. Digital tomosyn- 2010;55:3479–3497.
thesis with an on-board kilovoltage imaging device. Int 73. Franks KN, Purdie TG, Dawson LA, et al. Incorporating
J Radiat Oncol Biol Phys 2006;65:8–15. heterogeneity correction and 4DCT in lung stereotactic
56. Kolitsi Z, Panayiotakis G, Pallikarakis N. A method for body radiation therapy (SBRT): the effect on target cov-
selective removal of out-of-plane structures in digital erage, organ-at-risk doses, and dose conformity. Med
tomosynthesis. Med Phys 1992;20:47–50. Dosim 2010;35:101–107.
57. Badea C, Kolitsi Z, Pallikarakis N. A wavelet-based 74. Rietzel E, Pan T, Chen GTY. Four-dimensional computed
method for removal of out-of-plane structures in digital tomography: Image formation and clinical protocol.
tomosynthesis. Comp Med Img Graph 1998;22:309–315. Med Phys 2005;32:874.
58. Santoro J, Kriminski S, Lovelock DM, et al. Evaluation 75. Fitzpatrick MJ, Starkschall G, Antolak JA, et al.
of respiration-correlated digital tomosynthesis in lung. Displacement-based binning of time-dependent com-
Med Phys 2010;37:1237–1245. puted tomography image data sets. Med Phys 2006;33:
59. Oelfke U, Tucking T, Nill S, et al. Linac-integrated kV- 235–246.
cone beam CT: technical features and first applications. 76. Abdelnour AF, Nehmeh SA, Pan T, et al. Phase and ampli-
Med Dosim 2006;31:62–70. tude binning for 4D-CT imaging. Phys Med Biol 2007;52:
60. Yin FF, Guan H, Lu W. A technique for on-board CT 3515–3529.
reconstruction using both kilovoltage and megavoltage 77. Purdie TG, Moseley DJ, Bissonnette JP, et al. Respiration
beam projections for 3D treatment verification. Med correlated cone-beam computed tomography and 4DCT
Phys 2005;32:2819–2826. for evaluating target motion in stereotactic lung radia-
61. Blessing M, Stsepankou D, Wertz H, et al. Breath-hold tion therapy. Acta Oncol 2006;45:915–922.
target localization with simultaneous kilovoltage/ 78. Sonke JJ, Zijp L, Remeijer P, et al. Respiratory correlated
megavoltage cone-beam computed tomography and cone beam CT. Med Phys 2005;32:1176–1186.
fast reconstruction. Int J Radiat Oncol Biol Phys 2010;78: 79. Li T, Xing L, Munro P, et al. Four-dimensional cone-beam
1219–1226. computed tomography using an on-board imager. Med
62. Keall PJ, Mageras GS, Balter JM, et al. The management Phys 2006;33:3825–3833.
of respiratory motion in radiation oncology report of 80. Chang J, Sillanpaa J, Ling CC, et al. Integrating respiratory
AAPM Task Group 76. Med Phys 2006;33:3874–900. gating into a megavoltage cone-beam CT system. Med
63. Li G, Citrin D, Camphausen K, et al. Advances in 4D Phys 2006;33:2354–2361.
medical imaging and 4D radiation therapy. Technol 81. Li T, Koong A, Xing L. Enhanced 4D cone-beam CT
Cancer Res Treat 2008;7:67–81. with inter-phase motion model. Med Phys 2007;34:
64. Low DA, Nystrom M, Kalinin E, et al. A method for the 3688–3695.
reconstruction of four-dimensional synchronized CT 82. Leng S, Zambelli J, Tolakanahalli R, et al. Streaking
scans acquired during free breathing. Med Phy 2003;30: artifacts reduction in four-dimensional cone-beam
1254–1263. computed tomography. Med Phys 2008;35:4649–4659.
65. Pan T, Lee TY, Rietzel E, et al. 4D-CT imaging of a vol- 83. Rit S, Wolthaus J, Van Herk M, et al. On-the-fly motion-
ume influenced by respiratory motion on multi-slice compensated cone-beam CT using an a priori model
CT. Med Phys 2004;31:333–340. of the respiratory motion. Med Phys 2009;36:2283–
66. Ford EC, Mageras GS, Yorke E, et al. Respiration-correlated 2296.
spiral CT: a method of measuring respiratory-induced 84. Zhang Q, Hu YC, Liu F, et al. Correction of motion arti-
anatomic motion for radiation treatment planning. Med facts in cone-beam CT using a patient-specific respiratory
Phys 2003;30:88–97. motion model. Med Phys 2010;37:2901–2909.

LWBK933_Ch14_p229-258.indd 253 6/1/11 6:45 PM


254 n  Treatment Planning in Radiation Oncology

85. Huang E, Dong L, Chandra A, et al. Intrafraction pros- for prostate IMRT. Int J Radiat Oncol Biol Phys 2004;60:
tate motion during IMRT for prostate cancer. Int J Radiat 636–647.
Oncol Biol Phys 2002;53:261–268. 103. Wang C, Chao M, Lee L, et al. MRI-based treatment
86. Langen KM, Pouliot J, Anezinos C, et al. Evaluation of planning with electron density information mapped
ultrasound-based prostate localization for image-guid- from CT images: a preliminary study. Technol Cancer
ed radiotherapy. Int J Radiat Oncol Biol Phys 2003;57: Res Treat 2008;7:341–348.
635–644. 104. Karlsson M, Karlsson MG, Nyholm T, et al. Dedicated
87. Kuban DA, Dong L, Cheung R, et al. Ultrasound-based magnetic resonance imaging in the radiotherapy clinic.
localization. Semin Radiat Oncol 2005;15:180–191. Int J Radiat Oncol Biol Phys 2009;74:644–651.
88. Fuss M, Salter BJ, Cavanaugh SX, et al. Daily ultrasound- 105. Shirato H, Suzuki K, Sharp GC, et al. Speed and ampli-
based image-guided targeting for radiotherapy of upper tude of lung tumor motion precisely detected in four-
abdominal malignancies. Int J Radiat Oncol Biol Phys dimensional setup and in real-time tumor-tracking
2004;59:1245–1256. radiotherapy. Int J Radiat Oncol Biol Phys 2006;64:
89. Peng C, Kainz K, Lawton C, et al. A comparison of daily 1229–1236.
megavoltage CT and ultrasound image guided radiation 106. Onimaru R, Shirato H, Fujino M, et al. The effect of tumor
therapy for prostate cancer. Med Phys 2008;35:5619–5628. location and respiratory function on tumor movement
90. Fung AY, Ayyangar KM, Djajaputra D, et al. Ultrasound- estimated by real-time tracking radiotherapy (RTRT) sys-
based guidance of intensity-modulated radiation therapy. tem. Int J Radiat Oncol Biol Phys 2005;63:164–169.
Med Dosim 2006;31:20–29. 107. Ding GX, Duggan DM, Coffey CW. Commissioning ster-
91. Dong L, de Crevoisier R, Bonnen M, et al. Evaluation of eotactic radiosurgery beams using both experimental and
an ultrasound-based prostate target localization tech- theoretical methods. Phys Med Biol 2006;51:2549–2566.
nique with an in-room CT-on-rails. Int J Radiat Oncol 108. Verellen D, Soete G, Linthout N, et al. Quality assur-
Biol Phys 2004;60:S332–S333. ance of a system for improved target localization and
92. Fuss M, Cavanaugh SX, Fuss C, et al. Daily stereotactic patient set-up that combines real-time infrared track-
ultrasound prostate targeting: Inter-user variability. ing and stereoscopic X-ray imaging. Radiother Oncol
Technol Cancer Res Treat 2003;2:161–169. 2003;67:129–141.
93. Van Den Heuvel F, Powell T, Seppi E, et al. Independ- 109. Linthout N, Verellen D, Tournel K, et al. Six dimen-
ent verification of ultrasound based image-guided sional analysis with daily stereoscopic x-ray imag-
radiation treatment, using electronic portal imag- ing of intrafraction patient motion in head and neck
ing and implanted gold markers. Med Phys 2003;30: treatments using five points fixation masks. Med Phys
2878–2887. 2006;33:504–513.
94. Scarbrough TJ, Golden NM, Ting JY, et al. Compari- 110. Jin JY, Yin FF, Tenn SE, et al. Use of the BrainLAB
son of ultrasound and implanted seed marker prostate ExacTrac X-Ray 6D system in image-guided radiotherapy.
localization methods: implications for image-guided Med Dosim 2008;33:124–134.
radiotherapy. Int J Radiat Oncol Biol Phys 2006;65: 111. Nakagawa K, Haga A, Shiraishi K, et al. First clinical
378–387. cone-beam CT imaging during volumetric modulated
95. Dobler B, Mai S, Ross C, et al. Evaluation of possible arc therapy. Radiother Oncol 2009;90:422–423.
prostate displacement induced by pressure applied 112. Wiersma RD, Mao W, Xing L. Combined kV and MV
during transabdominal ultrasound image acquisition. imaging for real-time tracking of implanted fiducial
Strahlenther Onkol 2006;182:240–246. markers. Med Phys 2008;35:1191–1198.
96. Tomé WA, Meeks SL, Orton NP, et al. Commission- 113. Amies C, Bani-Hashemi A, Celi JC, et al. A multi-
ing and quality assurance of an optically guided three- platform approach to image guided radiation therapy
dimensional ultrasound target localization system for (IGRT). Med Dosim 2006;31:12–19.
radiotherapy. Med Phys 2002;29:1781–1788. 114. Stutzel J, Oelfke U, Nill S. A quantitative image quality
97. Bert C, Metheany KG, Doppke K, et al. A phantom comparison of four different image guided radiotherapy
evaluation of a stereo-vision surface imaging system for devices. Radiother Oncol 2008;86:20–24.
radiotherapy patient setup. Med Phys 2005;32:2753– 115. Meeks SL, Tome WA, Willoughby TR, et al. Optically
2762. guided patient positioning techniques. Semin Radiat
98. Djajaputra D, Li S. Real-time 3D surface-image-guided Oncol 2005;15:192–201.
beam setup in radiotherapy of breast cancer. Med Phys 116. Li S, Liu D, Yin G, et al. Real-time 3D-surface-guided
2005;32:65–75. head refixation useful for fractionated stereotactic radi-
99. Hendee WR, Ritenour ER. Medical imaging physics, otherapy. Med Phys 2006;33:492–503.
4th ed. New York, NY: Wiley-Liss, 2002. 117. Schoffel PJ, Harms W, Sroka-Perez G, et al. Accuracy
100. Dinkel J, Hintze C, Tetzlaff R, et al. 4D-MRI analysis of a commercial optical 3D surface imaging system for
of lung tumor motion in patients with hemidiaphrag- realignment of patients for radiotherapy of the thorax.
matic paralysis. Radiother Oncol 2009;91:449–454. Phys Med Biol 2007;52:3949–3963.
101. Wang D, Doddrell DM. A proposed scheme for com- 118. Peng JL, Kahler D, Li JG, et al. Characterization of a
prehensive characterization of the measured geometric real-time surface image-guided stereotactic positioning
distortion in magnetic resonance imaging using a three- system. Med Phys 2010;37:5421–5433.
dimensional phantom. Med Phys 2004;31:2212–2218. 119. Cervino LI, Pawlicki T, Lawson JD, et al. Frame-less and
102. Chen L, Price RA Jr, Wang L, et al. MRI-based treat- mask-less cranial stereotactic radiosurgery: a feasibility
ment planning for radiotherapy: dosimetric verification study. Phys Med Biol 2010;55:1863–1873.

LWBK933_Ch14_p229-258.indd 254 6/1/11 6:45 PM


Image-Guided Radiation Therapy  n 255

120. Balter JM, Wright JN, Newell LJ, et al. Accuracy of a 138. Chamberlain MC, Blumenthal DT. Intracranial menin-
wireless localization system for radiotherapy. Int J Radiat giomas: diagnosis and treatment. Expert Rev Neurother
Oncol Biol Phys 2005;61:933–937. 2004;4:641–648.
121. Santanam L, Malinowski K, Hubenshmidt J, et al. 139. Ryu SI, Chang SD, Kim DH, et al. Image-guided hypo-
Fiducial-based translational localization accuracy of fractionated stereotactic radiosurgery to spinal lesions.
electromagnetic tracking system and on-board kilo- Neurosurgery 2001;49:838–846.
voltage imaging system. Int J Radiat Oncol Biol Phys 140. Gerszten PC, Ozhasoglu C, Burton SA, et al. Evaluation
2008;70:892–899. of CyberKnife frameless real-time image-guided stere-
122. Santanam L, Noel C, Willoughby TR, et al. Quality otactic radiosurgery for spinal lesions. Stereotact Funct
assurance for clinical implementation of an electromag- Neurosurg 2003;81:84–89.
netic tracking system. Med Phys 2009;36:3477–3486. 141. Yu C, Main W, Taylor D, et al. An anthropomorphic
123. Sawant A, Smith RL, Venkat RB, et al. Toward submil- phantom study of the accuracy of Cyberknife spinal
limeter accuracy in the management of intrafraction radiosurgery. Neurosurgery 2004;55:1138–1149.
motion: the integration of real-time internal position 142. Murphy MJ, Adler JR Jr, Bodduluri M, et al. Image-
monitoring and multileaf collimator target tracking. guided radiosurgery for the spine and pancreas. Comput
Int J Radiat Oncol Biol Phys 2009;74:575–582. Aided Surg 2000;5:278–288.
124. Willoughby TR, Kupelian PA, Pouliot J, et al. Target 143. Whyte RI, Crownover R, Murphy MJ, et al. Stereotactic
localization and real-time tracking using the Calypso radiosurgery for lung tumors: preliminary report of a
4D localization system in patients with localized prostate phase I trial. Ann Thorac Surg 2003;75:1097–1101.
cancer. Int J Radiat Oncol Biol Phys 2006;65:528–534. 144. Silvano G. New radiation techniques for treatment of
125. Raaijmakers AJ, Raaymakers BW, Lagendijk JJ. Integrat- locally advanced non-small cell lung cancer (NSCLC).
ing a MRI scanner with a 6 MV radiotherapy accelerator: Ann Oncol 2006;17:34–35.
dose increase at tissue-air interfaces in a lateral magnet- 145. Kamino Y, Takayama K, Kokubo M, et al. Development
ic field due to returning electrons. Phys Med Biol 2005; of a four-dimensional image-guided radiotherapy system
50:1363–1376. with a gimbaled X-ray head. Int J Radiat Oncol Biol Phys
126. Fallone BG, Murray B, Rathee S, et al. First MR images 2006;66:271–278.
obtained during megavoltage photon irradiation from a 146. Verellen D, Depuydt T, Gevaert T, et al. Gating and track-
prototype integrated linac-MR system. Med Phys 2009; ing, 4D in thoracic tumours. Cancer Radiother 2010;14:
36:2084–2088. 446–454.
127. Kirkby C, Murray B, Rathee S, et al. Lung dosimetry in a 147. Depuydt T, Verellen D, Haas O, et al. Geometric accuracy
linac-MRI radiotherapy unit with a longitudinal mag- of a novel gimbals based radiation therapy tumor tracking
netic field. Med Phys 2010;37:4722–4732. system. Radiother Oncol 2011;98:365–372.
128. Keall PJ, Joshi S, Vedam SS, et al. Four-dimensional 148. Yin FF, Wong J, Balter J, et al. The role of in-room kV [AQ5]
radiotherapy planning for DMLC-based respiratory x-ray imaging for patient setup and target localization.
motion tracking. Med Phys 2005;32:942–951. December 2009. http://www.aapm.org/pubs/reports/
129. McQuaid D, Webb S. IMRT delivery to a moving target RPT_104.pdf
by dynamic MLC tracking: delivery for targets moving 149. Klein EE, Hanley J, Bayouth J, et al. Task Group 142
in two dimensions in the beam’s eye view. Phys Med report: quality assurance of medical accelerators. Med
Biol 2006;51:4819–4839. Phys 2009;36:4197–4212.
130. McMahon R, Papiez L, Rangaraj D. Dynamic-MLC 150. Benedict SH, Yenice KM, Followill D, et al. Stereotactic
leaf control utilizing on-flight intensity calculations: a body radiation therapy: the report of AAPM Task Group
robust method for real-time IMRT delivery over moving 101. Med Phys 2010;37:4078–4101.
rigid targets. Med Phys 2007;34:3211–3223. 151. Sharpe MB, Moseley DJ, Purdie TG, et al. The stability
131. D’Souza WD, Naqvi SA, Yu CX. Real-time intra-fraction- of mechanical calibration for a kV cone beam computed
motion tracking using the treatment couch: a feasibility tomography system integrated with linear accelerator.
study. Phys Med Biol 2005;50:4021–4033. Med Phys 2006;33:136–144.
132. Wiersma RD, Wen Z, Sadinski M, et al. Development of 152. Mao W, Lee L, Xing L. Development of a QA phan-
a frameless stereotactic radiosurgery system based on tom and automated analysis tool for geometric quality
real-time 6D position monitoring and adaptive head assurance of on-board MV and kV x-ray imaging sys-
motion compensation. Phys Med Biol 2010;55:389–401. tems. Med Phys 2008;35:1497–1506.
133. Takakura T, Mizowaki T, Nakata M, et al. The geometric 153. Goddu SM, Mutic S, Pechenaya OL, et al. Enhanced effi-
accuracy of frameless stereotactic radiosurgery using a ciency in helical tomotherapy quality assurance using a
6D robotic couch system. Phys Med Biol 2010;55:1–10. custom-designed water-equivalent phantom. Phys Med
134. Adler JR Jr, Chang SD, Murphy MJ, et al. The Cyber- Biol 2009;54:5663–5674.
knife: a frameless robotic system for radiosurgery. Ster- 154. Langen KM, Papanikolaou N, Balog J, et al. QA for heli-
eotact Funct Neurosurg 1997;69:124–128. cal tomotherapy: report of the AAPM Task Group 148.
135. Quinn AM. CyberKnife: a robotic radiosurgery system. Med Phys 2010;37:4817–4853.
Clin J Oncol Nurs 2002;6:149, 156. 155. Bissonnette JP. Quality assurance of image-guidance
136. Murphy MJ. Tracking moving organs in real time. Semin technologies. Semin Radiat Oncol 2007;17:278–286.
Radiat Oncol 2004;14:91–100. 156. Bissonnette JP, Moseley DJ, Jaffray DA. A quality assurance
137. Chang SD, Adler JR. Robotics and radiosurgery–the program for image quality of cone-beam CT guidance in
cyberknife. Stereotact Funct Neurosurg 2001;76:204–208. radiation therapy. Med Phys 2008;35:1807–1815.

LWBK933_Ch14_p229-258.indd 255 6/1/11 6:45 PM


256 n  Treatment Planning in Radiation Oncology

157. Bissonnette JP, Moseley D, White E, et al. Quality 174. Zhong H, Kim J, Chetty IJ. Analysis of deformable
assurance for the geometric accuracy of cone-beam CT image registration accuracy using computational mod-
guidance in radiation therapy. Int J Radiat Oncol Biol eling. Med Phys 2010;37:970–979.
Phys 2008;71:S57–S61. 175. Wang H, Garden AS, Zhang L, et al. Performance evalu-
158. Yoo S, Yin FF. Dosimetric feasibility of cone-beam CT- ation of automatic anatomy segmentation algorithm on
based treatment planning compared to CT-based treat- repeat or four-dimensional computed tomography
ment planning. Int J Radiat Oncol Biol Phys 2006;66: images using deformable image registration method.
1553–1561. Int J Radiat Oncol Biol Phys 2008;72:210–219.
159. Gopal A, Samant SS. Use of a line-pair resolution phan- 176. Yue NJ, Kim S, Lewis BE, et al. Optimization of couch
tom for comprehensive quality assurance of electronic translational corrections to compensate for rotational
portal imaging devices based on fundamental imaging and deformable target deviations in image guided radi-
metrics. Med Phys 2009;36:2006–2015. otherapy. Med Phys 2008;35:4375–4385.
160. Gayou O, Miften M. Commissioning and clinical imple- 177. Wu QJ, Thongphiew D, Wang Z, et al. The impact of
mentation of a mega-voltage cone beam CT system for respiratory motion and treatment technique on stereo-
treatment localization. Med Phys 2007;34:3183–3192. tactic body radiation therapy for liver cancer. Med Phys
161. Antypas C, Pantelis E. Performance evaluation of a 2008;35:1440–1451.
CyberKnife G4 image-guided robotic stereotactic radi- 178. Wijesooriya K, Weiss E, Dill V, et al. Quantifying the
osurgery system. Phys Med Biol 2008;53:4697–4718. accuracy of automated structure segmentation in 4D
162. Jiang SB, Wolfgang J, Mageras GS. Quality assurance CT images using a deformable image registration algo-
challenges for motion-adaptive radiation therapy: gat- rithm. Med Phys 2008;35:1251–1260.
ing, breath holding, and four-dimensional computed 179. Thongphiew D, Wu QJ, Lee WR, et al. Comparison of
tomography. Int J Radiat Oncol Biol Phys 2008;71: online IGRT techniques for prostate IMRT treatment:
S103–S107. adaptive vs repositioning correction. Med Phys 2009;36:
163. Li G, Xie H, Ning H, et al. A novel 3D volumetric voxel 1651–1662.
registration technique for volume-view-guided image 180. Zhang Q, Pevsner A, Hertanto A, et al. A patient-
registration of multiple imaging modalities. Int J Radiat specific respiratory model of anatomical motion for
Oncol Biol Phys 2005;63:261–273. radiation treatment planning. Med Phys 2007;34:4772–
164. Li G, Xie H, Ning H, et al. Accuracy of 3D volumet- 4781.
ric image registration based on CT, MR and PET/CT 181. Li R, Jia X, Lewis JH, et al. Real-time volumetric image
phantom experiments. J Appl Clin Med Phys 2008;9: reconstruction and 3D tumor localization based on a
2781. single x-ray projection image for lung cancer radiother-
165. Sharpe M, Brock KK. Quality assurance of serial 3D apy. Med Phys 2010;37:2822–2826.
image registration, fusion, and segmentation. Int J Radiat 182. Rietzel E, Chen GT, Choi NC, et al. Four-dimensional
Oncol Biol Phys 2008;71:S33–S37. image-based treatment planning: target volume seg-
166. Zhang L, Garden AS, Lo J, et al. Multiple regions-of- mentation and dose calculation in the presence of
interest analysis of setup uncertainties for head-and- respiratory motion. Int J Radiat Oncol Biol Phys 2005;
neck cancer radiotherapy. Int J Radiat Oncol Biol Phys 61:1535–1550.
2006;64:1559–1569. 183. Hugo GD, Campbell J, Zhang T, et al. Cumulative
167. van Kranen S, van Beek S, Rasch C, et al. Setup uncer- lung dose for several motion management strategies
tainties of anatomical sub-regions in head-and-neck as a function of pretreatment patient parameters. Int J
cancer patients after offline CBCT guidance. Int J Radiat Radiat Oncol Biol Phys 2009;74:593–601.
Oncol Biol Phys 2009;73:1566–1573. 184. Yan D, Vicini F, Wong J, et al. Adaptive radiation therapy.
[AQ6] 168. Giske K, Stoiber EM, Schwarz M, et al. Local setup errors Phys Med Biol 1997;42:123–132.
in image-guided radiotherapy for head and neck cancer 185. Yan D, Jaffray DA, Wong JW. A model to accumulate
patients immobilized with a custom-made device. Int J fractionated dose in a deforming organ. Int J Radiat
Radiat Oncol Biol Phys 2010;1–8. Oncol Biol Phys 1999;44:665–675.
169. Park SB, Rhee FC, Monroe JI, et al. Spatially weighted 186. Yan D, Lockman D. Organ/patient geometric variation
mutual information image registration for image guided in external beam radiotherapy and its effects. Med Phys
radiation therapy. Med Phys 2010;37:4590–4601. 2001;28:593–602.
170. Pluim JP, Maintz JB, Viergever MA. Mutual-information- 187. DICOM: Digital Imaging and Communications in
based registration of medical images: a survey. IEEE Trans Medicine, PS3 Part 3, 2008. NEMA: National Electri-
Med Imaging 2003;22:986–1004. cal Manufacturers Association. Available from ftp://
171. Brock KK, Sharpe MB, Dawson LA, et al. Accuracy of medical.nema.org/medical/dicom/2008/08_03pu.pdf.
finite element model-based multi-organ deformable Accessed Mar. 25, 2011.
image registration. Med Phys 2005;32:1647–1659. 188. Mageras GS, Hu YC, McNamara S, et al. Imaging for
172. Wang H, Dong L, O’Daniel J, et al. Validation of an radiation treatment planning. In: Curran BH, Starkschall
accelerated “demons” algorithm for deformable image G, eds. Informatics in radiation oncology. Boca Raton,
registration in radiation therapy. Phys Med Biol 2005;50: FL: CRC Press, 2011 (in press).
2887–2905. 189. Lu W, Olivera GH, Chen Q, et al. Automatic re-contouring
173. Liang J, Yan D. Reducing uncertainties in volumetric in 4D radiotherapy. Phys Med Biol 2006;51:1077–1099.
image based deformable organ registration. Med Phys 190. Söhn M, Birkner M, Alber M, et al. Modelling individual
2003;30:2116–2122. geometric variation based on dominant eigenmodes of

LWBK933_Ch14_p229-258.indd 256 6/1/11 6:45 PM


Image-Guided Radiation Therapy  n 257

organ deformation: Implementation and evaluation. 210. Remouchamps VM, Letts N, Vicini FA, et al. Initial
Phys Med Biol 2005;50:5893–5908. clinical experience with moderate deep-inspiration
191. Yan D. Adaptive radiotherapy: merging principle into breath hold using an active breathing control device
clinical practice. Semin Radiat Oncol 2010;20:79–83. in the treatment of patients with left-sided breast can-
192. Deasy JO, Blanco AI, Clark VH. CERR: a computational cer using external beam radiation therapy. Int J Radiat
environment for radiotherapy research. Med Phys 2003; Oncol Biol Phys 2003;56:704–715.
30:979–985. 211. Schweikard A, Shiomi H, Adler J. Respiration tracking
193. El Naqa I, Suneja G, Lindsay PE, et al. Dose response in radiosurgery. Med Phys 2004;31:2738–41.
explorer: an integrated open-source tool for exploring 212. Muacevic A, Drexler C, Wowra B et al. Technical
and modelling radiotherapy dose-volume outcome description, phantom accuracy, and clinical feasibility for
relationships. Phys Med Biol 2006;51:5719–5735. single-session lung radiosurgery using robotic image-
194. Chow JC, Leung MK, Islam MK, et al. Evaluation of the guided real-time respiratory tumor tracking. Technol
effect of patient dose from cone beam computed tomog- Cancer Res Treat 2007;6:321–328.
raphy on prostate IMRT using Monte Carlo simulation. 213. Stintzing S, Hoffmann RT, Heinemann V, et al. Frame-
Med Phys 2008;35:52–60. less single-session robotic radiosurgery of liver metas-
195. Jaffray D. Image-guided radiation therapy: from concept tases in colorectal cancer patients. Eur J Cancer 2010;46:
to practice. Semin Radiat Oncol 2007;17(4):243–244. 1026–1032.
196. Mageras GS. Introduction to management of target 214. Zimmerman J, Korreman S, Persson G, et al. DMLC
localization uncertainties in external beam therapy. motion tracking of moving targets for intensity mod-
Semin Radiat Oncol 2005;15(3):133–135. ulated arc therapy treatment: a feasibility study. Acta
197. Jaffray D, Kupelian P, Djemil T, et al. Review of image- Oncol 2009;48:245–250.
guided radiation therapy. Expert Rev Anticancer Ther 215. van Herk M. Errors and margins in radiotherapy. Semin
2007;7:89–103. Radiat Oncol 2004;14:52–64.
198. Yan D, Ziaja E, Jaffray D, et al. The use of adaptive radia- 216. van Herk M, Remeijer P, Rasch C, et al. The probability
tion therapy to reduce setup error: a prospective clinical of correct target dosage: dose-population histograms
study. Int J Radiat Oncol Biol Phys 1998;41:715–720. for deriving treatment margins in radiotherapy. Int J
199. Yan D, Lockman D, Brabbins D, et al. An off-line strategy Radiat Oncol Biol Phys 2000;47:1121–1135.
for constructing a patient-specific planning target vol- 217. Grills IS, Hugo G, Kestin LL, et al. Image-guided radio-
ume in adaptive treatment process for prostate cancer. therapy via daily online cone-beam CT substantially
Int J Radiat Oncol Biol Phys 2000;48:289–302. reduces margin requirements for stereotactic lung
200. Keller H, Ritter MA, Mackie TR. Optimal stochastic radiotherapy. Int J Radiat Oncol Biol Phys 2008;70:
correction strategies for rigid-body target motion. Int 1045–1056.
J Radiat Oncol Biol Phys 2003;55:261–270. 218. Liu HH, Balter P, Tutt T, et al. Assessing respiration-
201. Bortfeld T, Jiang SB, van Herk M. When should systemat- induced tumor motion and internal target volume
ic patient positioning errors in radiotherapy be corrected? using four-dimensional computed tomography for
Phys Med Biol 2002;47:(23):N297–N302. radiotherapy of lung cancer. Int J Radiat Oncol Biol
202. van Lin EN, Van Der Vight L, Huizenga H, et al. Set-up Phys 2007;68:531–540.
improvement in head and neck radiotherapy using a 3D 219. Panakis N, McNair HA, Christian JA, et al. Defining
off-line EPID-based correction protocol and a custom- the margins in the radical radiotherapy of non-small
ised head and neck support. Radiother Oncol 2003;68: cell lung cancer (NSCLC) with active breathing con-
137–148. trol (ABC) and the effect on physical lung parameters.
203. Wu Q, Liang J, Yan D. Application of dose compensation Radiother Oncol 2008;87:65–73.
in image-guided radiotherapy of prostate cancer. Phys 220. Nelson C, Balter P, Morice RC, et al. Evaluation of
Med Biol 2006;51:1405–1419. tumor position and PTV margins using image guid-
204. Song W, Schaly B, Bauman G, et al. Image-guided adaptive ance and respiratory gating. Int J Radiat Oncol Biol Phys
radiation therapy (IGART): Radiobiological and dose 2010;76:1578–1585.
escalation considerations for localized carcinoma of the 221. Melancon AD, O’Daniel JC, Zhang L, et al. Is a 3-mm
prostate. Med Phys 2005;32:2193–2203. intrafractional margin sufficient for daily image-guided
205. Pepin EW, Wu H, Sandison GA, et al. Site-specific volu- intensity-modulated radiation therapy of prostate cancer?
metric analysis of lung tumour motion. Phys Med Biol Radiother Oncol 2007;85:251–259.
2010;55(12):3325–3337. 222. Tanyi JA, He T, Summers PA, et al. Assessment of plan-
206. Riboldi M, Sharp GC, Baroni G, et al. Four-dimensional ning target volume margins for intensity-modulated
targeting error analysis in image-guided radiotherapy. radiotherapy of the prostate gland: role of daily inter- and
Phys Med Biol 2009;54(19):5995–6008. intrafraction motion. Int J Radiat Oncol Biol Phys 2010;78:
207. Chen H, Wu A, Brandner ED, et al. Dosimetric evalua- 1579–1585.
tions of the interplay effect in respiratory-gated intensity- 223. O’Daniel JC, Dong L, Zhang L, et al. Daily bone alignment
modulated radiation therapy. Med Phys 2009;36:893–903. with limited repeat CT correction rivals daily ultrasound
208. Kini VR, Vedam SS, Keall PJ, et al. Patient training in res- alignment for prostate radiotherapy. Int J Radiat Oncol
piratory-gated radiotherapy. Med Dosim 2003;28:7–11. Biol Phys 2008;71:274–280.
209. Wong JW, Sharpe MB, Jaffray DA, et al. The use of active 224. Li HS, Chetty IJ, Enke CA, et al. Dosimetric conse-
breathing control (ABC) to reduce margin for breathing quences of intrafraction prostate motion. Int J Radiat
motion. Int J Radiat Oncol Biol Phys 1999;44:911–919. Oncol Biol Phys 2008;71:801–812.

LWBK933_Ch14_p229-258.indd 257 6/1/11 6:45 PM


258 n  Treatment Planning in Radiation Oncology

225. Chen AM, Farwell DG, Luu Q, et al. Evaluation of the 238. Langen KM, Lu W, Willoughby TR, et al. Dosimetric
planning target volume in the treatment of head and neck effect of prostate motion during helical tomotherapy.
cancer with intensity-modulated radiotherapy: what is Int J Radiat Oncol Biol Phys 2009;74:1134–1142.
the appropriate expansion margin in the setting of daily 239. Chen L, Paskalev K, Xu X, et al. Rectal dose variation
[AQ7] image guidance? Int J Radiat Oncol Biol Phys 2010. during the course of image-guided radiation therapy of
226. Hugo GD, Yan D, Liang J. Population and patient-specific prostate cancer. Radiother Oncol 2010;95:198–202.
target margins for 4D adaptive radiotherapy to account 240. Goddu SM, Yaddanapudi S, Pechenaya OL, et al. Dosi-
for intra- and inter-fraction variation in lung tumour metric consequences of uncorrected setup errors in hel-
position. Phys Med Biol 2007;52:257–274. ical tomotherapy treatments of breast-cancer patients.
227. Pepin EW, Wu H, Shirato H. Dynamic gating window Radiother Oncol 2009;93:64–70.
for compensation of baseline shift in respiratory-gated 241. Chao KS, Majhail N, Huang CJ, et al. Intensity-modulated
radiation therapy. Med Phys 2011;38:1912–1918. radiation therapy reduces late salivary toxicity without
228. Langen KM, Jones DT. Organ motion and its manage- compromising tumor control in patients with oropha-
ment. Int J Radiat Oncol Biol Phys 2001;50:265–278. ryngeal carcinoma: a comparison with conventional
229. Booth JT, Zavgorodni SF. Set-up error and organ techniques. Radiother Oncol 2001;61:275–280.
motion uncertainty: a review. Australas Phys Eng Sci 242. Barker JL Jr, Garden AS, Ang KK, et al. Quantification
Med 1999;22:29–47. of volumetric and geometric changes occurring during
230. Yorke E, Rosenzweig KE, Wagman R, et al. Inter- fractionated radiotherapy for head-and-neck cancer
fractional anatomic variation in patients treated with using an integrated CT/linear accelerator system. Int J
respiration-gated radiotherapy. J Appl Clin Med Phys Radiat Oncol Biol Phys 2004;59:960–970.
2005;6:19–32. 243. O’Daniel JC, Garden AS, Schwartz DL, et al. Parotid
231. Nuyttens JJ, Robertson JM, Yan D, et al. The variability gland dose in intensity-modulated radiotherapy for
of the clinical target volume for rectal cancer due to head and neck cancer: is what you plan what you get?
internal organ motion during adjuvant treatment. Int Int J Radiat Oncol Biol Phys 2007;69:1290–1296.
J Radiat Oncol Biol Phys 2002;53:497–503. 244. Seco J, Sharp GC, Wu Z, et al. Dosimetric impact of
232. Pos FJ, Koedooder K, Hulshof M, et al. Influence of motion in free-breathing and gated lung radiotherapy:
bladder and rectal volume on spatial variability of a a 4D Monte Carlo study of intrafraction and interfraction
bladder tumor during radical radiotherapy. Int J Radiat effects. Med Phys 2008;35:356–366.
Oncol Biol Phys 2003;55:835–841. 245. Wu QJ, Thongphiew D, Wang Z, et al. On-line re-
233. Fokdal L, Honore H, Hoyer M, et al. Impact of changes optimization of prostate IMRT plans for adaptive radi-
in bladder and rectal filling volume on organ motion ation therapy. Phys Med Biol 2008;53:673–691.
and dose distribution of the bladder in radiotherapy 246. Sonke JJ, Belderbos J. Adaptive radiotherapy for lung
for urinary bladder cancer. Int J Radiat Oncol Biol Phys cancer. Semin Radiat Oncol 2010;20:94–106.
2004;59:436–444. 247. Yang D, Chaudhari SR, Goddu SM, et al. Deformable
234. Li XA, Qi XS, Pitterle M, et al. Interfractional variations registration of abdominal kilovoltage treatment plan-
in patient setup and anatomic change assessed by daily ning CT and tomotherapy daily megavoltage CT for
computed tomography. Int J Radiat Oncol Biol Phys treatment adaptation. Med Phys 2009;36:329–338.
2007;68:581–591. 248. Nijkamp J, Pos FJ, Nuver TT, et al. Adaptive radiotherapy
235. Sanguineti G, Marcenaro M, Franzone P, et al. Neoadju- for prostate cancer using kilovoltage cone-beam com-
vant androgen deprivation and prostate gland shrinkage puted tomography: first clinical results. Int J Radiat
during conformal radiotherapy. Radiother Oncol 2003; Oncol Biol Phys 2008;70:75–82.
66:151–157. 249. Ahunbay EE, Peng C, Godley A, et al. An on-line replan-
236. Litzenberg DW, Hadley SW, Tyagi N, et al. Synchronized ning method for head and neck adaptive radiotherapy.
dynamic dose reconstruction. Med Phys 2007;34:91–102. Med Phys 2009;36:4776–4790.
237. Ramsey CR, Langen KM, Kupelian PA, et al. A technique 250. Kobayashi H, Longmire MR, Ogawa M, et al. Multi-
for adaptive image-guided helical tomotherapy for lung plexed imaging in cancer diagnosis: applications and
cancer. Int J Radiat Oncol Biol Phys 2006;64:1237–1244. future advances. Lancet Oncol 2010;11:589–595.

LWBK933_Ch14_p229-258.indd 258 6/1/11 6:45 PM


[AQ 1]  In TOC the chapter title was “Image-Guided Radiotherapy” whereas in chapter opening page “Image-Guided
Radiation Therapy” is given. Please check and confirm the correct chapter title.
[AQ 2] In TOC only two authors were listed, but in chapter opening page two more author names are given. Please check.
[AQ 3] Please check the edit to this paragraph.
[AQ 4]  The term “gimballed-design” has been changed to “gimbal design.” Please check.
[AQ 5]  Please provide volume number and page range for reference (148), if appropriate.
[AQ 6]  Please provide volume number for reference (168).
[AQ 7]  Please provide volume number and page range for reference (225).
[AQ 8]  Please provide complete reference.

LWBK933_Ch14_p229-258.indd
View publication stats 1 6/1/11 6:45 PM

You might also like