You are on page 1of 13

BJR 2016 The Authors.

Published by the British Institute of Radiology

Received: Revised: Accepted: https://doi.org/10.1259/bjr.20160768


23 September 2016 11 December 2016 22 December 2016

Cite this article as:


Wong KH, Panek R, Bhide SA, Nutting CM, Harrington KJ, Newbold KL. The emerging potential of magnetic resonance imaging in
personalizing radiotherapy for head and neck cancer: an oncologists perspective. Br J Radiol 2017; 90: 20160768.

REVIEW ARTICLE
The emerging potential of magnetic resonance imaging in
personalizing radiotherapy for head and neck cancer: an
oncologists perspective
1,2
KEE H WONG, FRCR, 1,2RAFAL PANEK, PhD, 1,2SHREERANG A BHIDE, PhD, 1,2
CHRISTOPHER M NUTTING, PhD,
1,2
KEVIN J HARRINGTON, PhD and 1,2KATIE L NEWBOLD, MD
1
Head and neck unit, The Royal Marsden Hospital, London, UK
2
Radiotherapy and imaging, The Institute of Cancer Research, London, UK

Address correspondence to: Dr Kee H Wong


E-mail: kee.wong@icr.ac.uk

ABSTRACT
Head and neck cancer (HNC) is a challenging tumour site for radiotherapy delivery owing to its complex anatomy and
proximity to organs at risk (OARs) such as the spinal cord and optic apparatus. Despite significant advances in
radiotherapy planning techniques, radiation-induced morbidities remain substantial. Further improvement would require
high-quality imaging and tailored radiotherapy based on intratreatment response. For these reasons, the use of MRI in
radiotherapy planning for HNC is rapidly gaining popularity. MRI provides superior soft-tissue contrast in comparison with
CT, allowing better definition of the tumour and OARs. The lack of additional radiation exposure is another attractive
feature for intratreatment monitoring. In addition, advanced MRI techniques such as diffusion-weighted, dynamic
contrast-enhanced and intrinsic susceptibility-weighted MRI techniques are capable of characterizing tumour biology
further by providing quantitative functional parameters such as tissue cellularity, vascular permeability/perfusion and
hypoxia. These functional parameters are known to have radiobiological relevance, which potentially could guide
treatment adaptation based on their changes prior to or during radiotherapy. In this article, we first present an overview of
the applications of anatomical MRI sequences in head and neck radiotherapy, followed by the potentials and limitations of
functional MRI sequences in personalizing therapy.

INTRODUCTION anatomical information by which blood vessels, masses and


Radical radiotherapy (RT) is integral to the management of adjacent soft tissues are easily distinguishable. There is no
head and neck cancer (HNC) in both the primary and increased risk of secondary malignancies with repetitive
adjuvant settings. Advances in computer-assisted radio- imaging, making MRI an attractive tool in the eld of
logical techniques over the past two decades have in turn image-guided and adaptive radiotherapy.
revolutionized radiotherapy planning. Development of
advanced radiotherapy planning techniques such as The potential advantages of using MRI as a stand-alone
intensity-modulated radiotherapy (IMRT) and volumetric- radiotherapy planning platform have led to international
modulated arc therapy have allowed for better dose con- collaboration to develop MR-Linac. MR-Linac aims to
formation to the tumour target and sparing of surrounding combine the two technologies in the MRI scanner and
normal tissues. HNC was one of the rst tumour sites linear accelerator to address the current insufciency in
where IMRT was widely implemented owing to a signi- modern image-guided radiotherapy, i.e. to accurately de-
cant reduction in radiation-induced xerostomia in com- ne the tumour and tailor radiotherapy beams in real time.
parison with three-dimensional conformal planning.1 This would help eliminate the uncertainties related to pa-
tient setup, intrafraction and interfraction movements.
CT is currently the standard platform for radiotherapy However, there are several technical challenges with MR-
planning. CT, however, provides a poor soft-tissue contrast, Linac such as the lack of electron density data and the
resulting in difculty in identifying tumour and organs at inuence of magnetic eld on radiotherapy dose distribu-
risk (OARs) in the head and neck regions. On the contrary, tion due to secondary electrons, which are currently being
MRI utilizes a strong magnetic eld to provide high-resolution addressed during its development.
BJR Wong et al

In addition, advanced MRI sequences, such as diffusion- to be an independent poor prognostic feature, even for human
weighted (DW), dynamic contrast-enhanced (DCE) and in- papilloma virus (HPV)-driven OP cancer. Figure 1 shows an
trinsic susceptibility-weighted (ISW) sequences are capable of example of a metastatic retropharyngeal LN which was readily
characterizing tumour biology further by providing quantitative detectable on MRI, but not on CT.
functional parameters that are known to have radiobiological
signicance such as tissue cellularity, vascular perfusion/ Radiotherapy planningtumour target and organ at
permeability and hypoxia. These sequences, also collectively risk delineation
referred to as functional MRI (F-MRI) in this article, are areas of Prior to the era of IMRT/volumetric-modulated arc therapy,
ongoing research; but, there is increasing evidence to support head and neck radiotherapy planning has been historically based
the role of these F-MRI parameters as predictive and prognostic on compartmental volume, dened by anatomical boundaries.
biomarkers. Consequently, radiation-induced toxicities were substantial with
signicant negative impact on patient quality of life. Advances in
In the rst section of this article, we give an overview of the both diagnostic imaging and planning techniques have permit-
applications of anatomical MRI sequences in the management of ted a shift of practice towards volumetric contouring. This
HNC with specic focus on radiotherapy. For the purpose of means that high-dose clinical target volume only includes the
this review, the term HNC refers primarily to squamous cell gross tumour volume (GTV) with a pre-determined margin for
carcinoma, which accounts for over 90% of head and neck isotropic expansion, instead of the whole anatomical subsite.
malignancies. Next, we elaborate on each F-MRI modality and Importantly, no detrimental effect on locoregional control was
discuss their potential utilities and limitations as imaging bio- observed with volumetric contouring.79
markers to guide treatment individualization for HNC.
Several planning studies have underlined the benet of in-
ANATOMICAL MRI SEQUENCES corporating MR images in head and neck radiotherapy planning
Staging with improvement of tumour delineation and reduction in in-
MRI is increasingly used for dening the extent of tumour in- terobserver variations compared with CT.1012 It also enables
vasion into adjacent structures in HNC owing to its superior a more accurate delineation of neurological OARs such as the
soft-tissue contrast. T1 weighted images are generally considered spinal cord, brain stem, optic chiasm and hippocampus, which
the best for gross structural information, whereas T2 weighted is vital to avoid irreversible neurological sequelae. Nevertheless,
images distinguish abnormal pathology from surrounding tis- the accuracy of MRI in this context is hugely dependent on the
sues. In clinical practice, MRI is the imaging modality of choice scanning position. Appropriate neck immobilization for plan-
for staging primary disease for nasopharyngeal and sinonasal ning MRI is necessary to reduce the risk of geographical miss, as
tumours. It is also increasingly used to stage oropharyngeal (OP) deformable registration is unable to fully account for the dif-
cancer and detect cartilage invasion for laryngeal/ ferences in the position of the neck.13,14 Acquiring MRI in neck
hypopharyngeal cancer.2 immobilization requires deviation from standard protocol owing
to inability to use the standard head coil. However, this can be
MRI is also more reliable than CT at detecting the presence of overcome by using a combination of ex and spine coils.15
pre-vertebral space invasion,3 perineural spread4 and retro-
pharyngeal lymph node (LN) involvement,5 which are all im- The majority of commonly used metallic implants, including
portant factors for treatment decision-making. A study by dental objects, are MRI safe or conditional and can be imaged
Samuels et al6 demonstrated retropharyngeal node involvement with MRI. However, these objects may cause local magnetic eld

Figure 1. An example of axial T2 weighted MR image showing contralateral retropharyngeal node involvement (b) (indicated by
arrow) in a patient with left tonsillar squamous cell carcinoma, which was not readily visible on CT (a).

2 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

inhomogeneity, resulting in regional signal loss and pile-ups MRI is naturally the imaging modality of choice for this ap-
depending on the implant material and geometry.16 In practice, proach owing to lack of additional radiation exposure and better
these effects are often less problematic in comparison with CT soft-tissue contrast. Current imaging modalities available in the
streak artefact caused by beam hardening and photon starvation, treatment room such as cone-beam CT or in-room CT provide
thereby providing further advantage in radiotherapy planning. limited soft-tissue contrast which precludes precise tumour
Furthermore, these artefacts can be greatly reduced for MRI by tracking. One of the projected benets with MR-Linac is that
employing dedicated metal artefact reduction sequences, albeit patients could be imaged daily to allow real-time online
at the expense of longer acquisition times.17 matching, dosimetric analysis and plan reoptimization if re-
quired. Moreover, GTV could be modied based on tumour
In addition, it is feasible to quantify the magnitude of tumour response during radiotherapy, enabling further reduction in
motion using cine-MRI. This enables an individualized internal dose to adjacent organs (Figure 2). Efforts to develop automated
margin to generate a planning target volume for each patient. Cine- OAR segmentation and rapid adaptive replanning are ongoing.
MRI is acquired through continuous real-time imaging, which
historically posed limitations with poor image quality in terms of Functional MRI sequences
spatial resolution and signal-to-noise ratio, but this has vastly im- It is now evident that HNC represents a disease spectrum, which is
proved with newer techniques.18 A study assessing deglutition- divisible into different prognostic groups based on clinical variables
induced organ motion with cine-MRI observed some tumour such as clinical/radiological staging (Tumor, Node and Metastases),
motion even in the absence of swallowing.19 The use of a custom- HPV status and smoking history. There is, therefore, a pressing need
ized intraoral immobilization device could help minimize tongue for a more personalized approach to treatment decision-making in
movement and prevent swallowing,20 but this service requires input order to optimize the balance between therapeutic efcacy and tox-
from specialized orthodontics and is not widely available. icity. However, biomarkers which could provide reliable information
on the likely impact of a specic intervention at an early time point
Radiotherapy deliveryadaptive approach are required to guide treatment adaptation. F-MRI offers an attractive,
A patient anatomy changes throughout the course of radio- non-invasive means to characterize the tumour biology by providing
therapy, which may signicantly alter the dose distribution to quantitative parameters with known radiobiological relevance. This
both planning target volume and OARs. This is a prominent has, in turn, led to a surge in the number of studies investigating
issue for HNC owing to treatment-related weight loss. For ex- the role of these parameters as imaging biomarkers for HNC over the
ample, parotids are known to shrink up to 3040% of their past decade. In this section, we review the evidence and discuss the
original volume and tend to displace medially throughout the potential roles of these F-MRI sequences in the management of HNC.
course of radiotherapy.21,22 Adaptive radiotherapy studies with
CT have already demonstrated the benet of such an approach Diffusion-weighted MRI
in reducing cumulative dose to normal tissues throughout the DW MRI utilizes the Brownian motion of water to assess tissue
radiotherapy without compromising long-term outcome.2325 cellularity without the need of any exogenous contrast agent.

Figure 2. The coronal T2 weighted MR images on the left are illustrating the early anatomical changes in primary gross tumour
volume (GTV) observed following Week-1 and Week-2 radical chemoradiotherapy (CRT) in a patient with human papilloma virus-
positive T3N0M0 left tonsillar squamous cell carcinoma. The axial image on the right is showing an overlay of co-registered MRIs
between pre-treatment and Week-2 CRT for the same patient to illustrate the potential benefit of dose reduction to the pharyngeal
constrictors and left parotid through GTV adaptation during radiotherapy with the shrinkage of the tumour away from these
structures (filled contourWeek-2 CRT; dotted contourpre-treatment).

3 of 13 birpublications.org/bjr Br J Radiol;90:20160768
BJR Wong et al

The movement of the tissue water molecules during the course by the tissueair interface. A study by Schakel et al30 found
of the diffusion-encoding gradients results in dephasing, distortions up to 26 mm in the anteroposterior axis. This,
depicted as signal loss. The signal loss is proportional to the however, can be minimized with dedicated acquisition and
amount of water molecule displacement and the duration of the post-processing methods.31,32 DW distortion in HNC was
diffusion-encoding gradients (b-value). The voxel-based signal shown to be greatly compensated using reduced-distortion
loss can be quantied to produce maps of apparent diffusion readout-segmented echoplanar imaging33 and half-Fourier
coefcient (ADC), which is inversely correlated with tumour acquisition single-shot turbo spin echo (Figure 3). The latter,
cellularity.26 however, may not be optimal for qualitative tumour assess-
ment owing to a low interobserver agreement for ADC values
DW sequences are increasingly incorporated in routine head in lesions assessed with conventional echoplanar imaging vs
and neck imaging owing to their additional values to ana- half-Fourier acquisition single-shot turbo spin echo
tomical sequences at diagnosing primary tumours (PTs), techniques.34
nodal involvement and detecting recurrences.27 Vandecaveye
et al28 further reported the ability of DW MRI to differentiate There are now much published data that support the ability of
between tumour and post-radiotherapy changes, thus DW MRI to assist in the early prediction of treatment out-
enabling identication of residual tumour immediately after comes following chemoradiotherapy in HNC. Overall, there
radiotherapy, in contrast to udeoxyglucose-positron emis- were conicting results over the value of pre-treatment ADC at
sion tomography (PET) which has a poor specicity in this predicting response to treatment (Table 1). Five studies failed
situation.29 The high contrast between tumour and to distinguish patients with unfavourable disease based on pre-
surrounding tissues in heavily DW images has also raised the treatment ADC,3539 whereas six studies found high pre-
possibility to use this technique as an adjunct to guide GTV treatment tumour ADC to be predictive of poor outcome
delineation. Nevertheless, this is hampered by geometric following RT.4045 As illustrated in Table 1, there are hetero-
distortions which are pronounced in the head and neck re- geneities between the studies in scanning protocol, analytical
gion owing to the large magnetic eld inhomogeneity caused methods and measured clinical end points, which may be

Figure 3. A comparison of geometrical fidelity in head and neck diffusion-weighted (DW) images using conventional single-shot
echoplanar imaging (SS-EPI) and the readout-segmented echoplanar imaging (RESOLVE). DW sequences were acquired in the
axial plane, reformatted to the sagittal plane and overlaid with a T2 weighted anatomical sequence. Geometrical distortion resulting
in stretching or misslocation of anatomical signal can be observed in the SS-EPI sequence (inferior part of the spinal cord,
cerebellum and position of lymph nodes). A high geometrical fidelity can be observed in the corresponding images acquired using
RESOLVE (highlighted by the arrows).

4 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

Table 1. Summary of diffusion-weighted (DW) MRI biomarker studies in head and neck cancer

Number of
patients/
Design and Scan DW End points Suggested
ROIs
Study treatment time scanning (response Results ADC
analyzed/
modalities points protocol criterion) thresholds
primary
sites
CR group has
a lower 1.11 3 102
pre-treatment 3
mm2 s21
1.5 T or 3.0 T; CR vs PR based ADC
Pre-RT, 33 patients (all
TR/TE: on clinical and
Kim Prospective, Week 1 Stage IV)/LNs CR group has
4 s/89 ms pathological
et al40 (2009) CRT and Week only 66% a larger increase in
(b 5 0, 500, evidence
2 RT oropharynx
1000 s mm22) post-CRT ADC post-Week 1
None
(,0.01) and
post-Week 2
RT (,0.05)
28 patients
Inverse correlation
Retrospective, 1.5 T; TR/TE: (Stage IIIV)/ Tumour
of pre-treatment
Kato neoadjuvant Pre-RT 5 s/7072 ms both PTs and regression rate
ADC with tumour None
et al43 (2009) CRT, IC only (b 5 0, LNs 40% (RECIST)
regression rate
and RT 1000 s mm22) larynx/ post-treatment
(r 5 20.384)
hypopharynx
PTs: 14%
Lower increase in
30 patients (Week 2)
1.5 T; TR/TE: PTs and LNs ADC
Pre-RT, (Stage IIV)/ Locoregional and 25%
7.1 s/84 ms 2 and 4 weeks
Vandecaveye Prospective, Week 2 both PTs and control (median (Week 4)
(b 5 0, 50, after treatment is
et al49 (2010) CRT and RT and Week LNs 53% follow-up LNs: 14.6%
100, 500, 750, associated with
4 RT larynx/ 2 years) (Week 2)
1000 s mm22) poor locoregional
hypopharynx and 19%
control at 2 years
(Week 4)
38 patients (17 LC vs local
1.5 T; TR/TE: LC group has
Stage $III)/ failure (median
Hatakenaka Retrospective, Pre-RT 3 s/73 ms a lower 0.88 3 1023
PTs only 60% follow-up
et al41 (2011) CRT and RT only (b 5 0, 300, pre-treatment mm2 s21
hypopharynx/ approximately
1000 s mm22) ADC
larynx 10 months)
50 patients No signal for
(Stage IIIIV)/ pre-treatment N/A
Pre-RT, 1.5 T; TR/TE: single-slice ADC
Locoregional
Week 2 2 s/75 ms ADC analysis
King Prospective, control (median
RT and (b 5 0, 100, of tumour A fall in ADC
et al36 (2010) CRT and RT follow-up
6 weeks 200, 300, 400, with largest during RT
22 months)
post-RT 500 s mm22) diameter predicts for local
None
.50% failure
hypopharynx
32 patients,
1.5 T; TR/TE: (Stage IIV)/ Low
LC (median
Ohnishi Retrospective, Pre-RT 3 s/73 ms; PTs only 53% pre-treatment 0.79 3 1023
follow-up
et al48 (2011) CRT and RT only (b 5 0, 300, hypopharynx, ADC predicts mm2 s21
15 months)
1000 s mm22) 47% for LC
oropharynx
No signal for
pre-treatment N/A
1.5 or 3.0 T; ADC
Pre-IC 18 patients OS (median
TR/TE:
Berrak Retrospective, and (Stage IV)/ follow-up Aliveincrease in
4 s/89 ms
et al38 (2011) IC 1 CRT 3 weeks LNs only 72% approximately ADC 22%,
(b 5 0, 500,
post-IC oropharynx 19 months)
1000 s mm22) deceased None
decrease in
ADC 33%

(Continued)

5 of 13 birpublications.org/bjr Br J Radiol;90:20160768
BJR Wong et al

Table 1. (Continued)

Number of
patients/
Design and Scan DW End points Suggested
ROIs
Study treatment time scanning (response Results ADC
analyzed/
modalities points protocol criterion) thresholds
primary
sites
Responders vs
1.5 T or 3.0 T; 24 patients
partial/ No signal from
Prospective, TR/TE: (Stage IIIIV)/
Chawla Pre-RT non-responders pre-treatment
CRT and 4 s/89 ms both PTs and N/A
et al35 (2013) only (median ADC alone (LN
IC 1 CRT (b 5 0, 500, LNs 94%
follow-up and PTs)
1000 s mm22) oropharynx
23.7 months)
No signal from
35 patients pre-treatment N/A
1.5 T; TR/TE: (Stage IIIIV)/ Locoregional ADC
Pre-RT
Matoba Prospective, 4 s/68 ms both PTs and control (median
and Week A larger increase
et al37 (2014) CRT (b 5 0, 90, LNs 57% follow-up
3 RT
800 s mm22) larynx/ 30.8 months) in PT ADC at 24% increase
hypopharynx Week 3 RT in PT ADC
predicts for LC
69 patients
Higher
(Stage IIIIV)/
3.0 T; TR/TE: 3-year neck pre-treatment LN
LNs only
Ng Prospective, Pre-RT 8.2 s/84 ms control (median ADC is an 1.14 3 1023
(largest) 53%
et al42 (2014) CRT only (b 5 0, follow-up independent mm2 s21
oropharynx,
800 s mm22) 31 months) predictor of poor
47%
neck control
hypopharynx
Lower LN ADC is
86 patients an independent 1.14 3 1023
(Stage IIIIV)/ predictor of longer mm2 s21
3.0 T; TR/TE: PFS and OS PFS but not OS
both PTs and
Ng Prospective, Pre-RT 8.2 s/84 ms (median
LNs 52% PT ADC is
et al45 (2016) CRT only (b 5 0, follow-up
oropharynx,
800 s mm22) 36 months) not an
48% independent N/A
hypopharynx predictor factor
for PFS or OS
20 patients
Complete
(Stage IIIIV)/
remission
Pre-IC, 1.5 T; TR/TE: both PTs and No signal from
3 months
Wong Prospective, post-1st 13 s/61 ms LNs 90% pre-treatment
post-CRT N/A
et al39 (2016) IC 1 CRT and 2nd (b 5 50, 400, oropharynx ADC or
(median
cycle IC 800 s mm22) and 10% changes post-IC
follow-up
larynx/
14 months)
hypopharynx
ADC, apparent diffusion coefficient; CR, complete response; CRT, radical chemoradiotherapy; IC, induction chemotherapy; LC, local control; LN, lymph
node; N/A, not applicable; OS, overall survival; PFS, progression-free survival; PR, partial response; PT, primary tumour; ROI, region of interest; RT,
radical radiotherapy; TE, echo time; TR, repetition time.

accountable for the discrepancy in their ndings. Some inves- ,1.111.14 3 1023 mm2 s21 to be predictive of favourable out-
tigators used different magnetic eld strengths (1.5 T or 3.0 T) even come following radiotherapy.40,41,44,45 A contradictory result
within the same study and the number of b-values used ranged by Nakajo et al,46 where they found patients with low pre-
from 2 to 5. Whilst almost all studies excluded apparent tumour treatment primary ADC ,0.88 3 1023 mm2 s21 to have an
necrosis from ADC calculation, there is a lack of consensus on unfavourable 2-year outcome, somewhat highlighted the
the analytical methods: some studies only chose the tumour potential negative inuence of including inhomogeneous
with the largest diameter (primary or LN), whereas others analyzed tumour sites and treatment modalities in the study ndings:
them in isolation. However, the pre-treatment ADC thresholds 54% patients undergoing primary surgery instead of radio-
suggested by the positive studies appear to be fairly concordant therapy, with higher recurrence rate in patients undergoing
with primary ADC ,0.790.88 3 1023 mm2 s21 and nodal ADC surgery.

6 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

Another noteworthy observation by Wong et al39 is that HPV- shortens the longitudinal relaxation time (T1). This leads to
positive OP tumours in patients who achieved complete re- increased signal intensity in perfused tissue regions in T1
mission following chemoradiotherapy exhibited a wide range weighted images. The temporal changes in signal intensity
of pre-treatment ADC (0.91.54 3 1023 mm2 s21), thereby obtained by DCE MRI are related to the underlying permeability
undermining its predictive value. HPV-positive OP cancer and perfusion of tumour microenvironment, all of which are
(tonsil and base of tongue) is known to exhibit unique his- known to inuence treatment response. The gadolinium
topathological features such as indistinct cell borders and concentrationtime curve can also be tted to a two-
comedo necrosis, unlike other subsites or HPV-negative compartment pharmacokinetic model to yield various kinetic
disease.47,48 These features may have contributed to the and volumetric parameters such as the transfer coefcient from
high ADC in some HPV-positive tumours but, importantly, plasma to the interstitial space (Ktrans), the extracellular extra-
they do not have the same negative biological impact on vascular volume fraction (Ve) and plasma volume fraction (Vp).
treatment outcome as on a patient who is HPV negative.48
The potential impact of HPV status on ADC measurements in Enhancement patterns of DCE MRI have shown correlations
OP cancer has not been explored in other published studies; with malignancy,52 angiogenesis,53 proliferation54 and
hence, caution should be exercised when interpreting pre- hypoxia.55,56 One of the rst studies to evaluate the relationship
treatment ADC alone. between tumour perfusion and local control (LC) using DCE
MRI in patients with HNC was published by Hoskin et al.57
Whilst the value of pre-treatment ADC remains unclear, 13 patients underwent DCE MRI before and on completion of
treatment-induced changes of ADC during radiotherapy have accelerated radiotherapy. LC was found to be related to maxi-
been more consistently demonstrated in several clinical mum tumour enhancement following radiotherapy and the
studies.36,37,40,49 The cumulative results suggest that tumours difference in time taken to reach maximum tumour enhance-
that show a lower increase or even a decrease in ADC 14 weeks ment pre- and post-radiotherapy. These results suggested that
into radiotherapy (ADC ,1424%) are more likely to fail tumours with lower perfusion at the end of radiotherapy were
treatment. It is speculated that tumours with a good treatment most sensitive to treatment and those with greater tumour en-
response show a higher frequency of apoptosis or necrosis earlier hancement were likely to fail locally.
in the course of treatment than those with a poor response.
Thus, the ADC tends to increase in responding tumours because Since then, several other groups have evaluated the ability of pre-
of the presence of fewer barrier structures to the movement of treatment DCE parameters to provide prognostic information
tissue water such as cell membranes. The optimal timing for for patients with HNC undergoing radical chemoradiotherapy
early intratreatment assessment using DW MRI, however, still (CRT). These studies have been summarized in two systematic
needs to be established (between Week 1 and Week 4 of RT), as reviews published in recent years.58,59 The most commonly
development of mature scar tissues may falsely decrease ADC reported pre-treatment DCE parameters with predictive or
in responders.50 prognostic value are Ktrans, followed by Ve and Vp. Two earlier
clinical studies have shown low pre-treatment nodal tumour
The majority of published studies have calculated mean or Ktrans to be correlated with poor locoregional control and
median ADC from dened regions of interest to quantitate disease-free survival.60,61 No threshold was suggested, but non-
the observed differences in DW MRI between patients at responders in both studies have an average Ktrans value of
baseline and during treatment. More sophisticated analyses of 0.150.21 min21. However, one of the larger study to date by
DW MRI data are possible and may enable DW MRI to be Shukla-Dave et al62 reported that it was the skewness rather than
used as a more sensitive and specic biomarker to provide an the mean value of nodal tumour Ktrans which was the strongest
early prediction of response to chemoradiotherapy. Galban predictor of progression-free survival (PFS). The authors
et al51 described a voxelwise approach to the evaluation of therefore recommended calculation of skewness to be a better
ADC changes during treatment that involves the calculation measure of tumour heterogeneity, but did not indicate whether
of parametric response maps (PRM) in 12 patients. This ap- a positive or negative skew predicted for a better outcome.
proach uses registered baseline and intratreatment ADC maps
to calculate regional tumour response and they concluded Another large multimodality, parametric functional imaging
that PRM may be more sensitive to cellular changes than study of 69 patients by Ng et al42 reported low pre-treatment
measurements of the change in the mean ADC over whole nodal Ve (,0.23) to be an independent poor prognostic factor
regions of interest. One lingering uncertainty, however, is how for 3-year neck control for patients undergoing CRT. This is one
well deformable image registration accounts for volumetric or of the rst study reported in HNC to incorporate multimodality
positional changes in tumour between the scanning time functional imaging in uorine-18 udeoxyglucose-PET/CT,
points to allow condent per-voxel analysis. Moreover, de- DCE and DW MRI, given emerging evidence from correlation
formable image registration itself remains a matter of research studies that these modalities provide different, but comple-
with lack of clinical validation. Further studies on larger mentary biological tumour information,63 thereby improving
numbers of patients are, therefore, required. predictive power. It is noteworthy that none of these studies
reported DCE parameters for PTs and this may be related to
Dynamic contrast-enhanced MRI technical difculties, e.g. motion caused by swallowing or sus-
DCE MRI assesses changes in signal intensity following the in- ceptibility artefacts. A more recent study by King et al64 in
jection of a paramagnetic contrast agent, e.g. gadolinium that 49 patients assessed the predictive value of pre-treatment DCE

7 of 13 birpublications.org/bjr Br J Radiol;90:20160768
BJR Wong et al

parameters in both primary and nodal tumours but failed to likely to benet from such a strategy. PET-based techniques
show any correlation with response to chemoradiotherapy. Ng using tracers such as uorine-18 uoromisonidazole are cur-
et al45 also provided an update on their previous study after rently the most widely studied functional imaging to charac-
recruiting additional 17 patients and including PT in analysis. terize hypoxia in HNC. However, these PET techniques are
This showed a similar result, i.e. low pre-treatment nodal Ve expensive, time consuming and suffer from a poor spatio-
independently predicted for shorter PFS and overall sur- temporal resolution and signal-to-noise ratio.73
vival (OS).
An alternative hypoxia-specic imaging technique is ISW MRI,
It is possible to conclude from the cumulative results shown also known as blood oxygen level-dependent MRI. It exploits the
above that pre-treatment DCE-derived Ktrans or Ve in LNs could paramagnetic properties of deoxyhaemoglobin in erythrocytes to
predict for outcome following radiotherapy in HNC. However, it create contrast. Essentially, deoxyhaemoglobin creates magnetic
is impossible to deduce which are the optimal pre-treatment susceptibility perturbations around blood vessels and the
parameters and the inconsistencies in the reported results are transverse MR relaxation rate R2* (R2* 5 1/T2*) of water in
invariably attributable to the differences in DCE scanning pro- blood and the surrounding tissues increases in proportion to
tocols, pharmacokinetic models and arterial input function tissue deoxyhaemoglobin concentration. Because oxygenation of
used. For example, there are up to four methodologies available haemoglobin is proportional to arterial blood polarographic
to estimate arterial input function, e.g. population averaged,65 oxygen levels, tumour R2* is a sensitive index of tissue oxy-
reference tissue based,66 contrast concentration in adjacent genation and a surrogate marker of hypoxia.
arteries67 and independent component analysis.68 Consequently,
large collective efforts are required to optimize and standardize ISW MRI has been typically performed by measuring changes in
DCE protocols for future studies. R2* with hyperoxic gas challenge such as carbogen. Clinical
studies conducted in patients with HNC have consistently dem-
In contrast, there is a paucity of data on the role of intratreat- onstrated that the tumour R2* decreases following inhalation of
ment DCE MRI to assess and predict response to radiotherapy. hyperoxic gas, indicative of improved blood oxygenation which
The number of patients in these studies are very small (n , 15), may further increase the radiosensitivity.74,75 The use of hyperoxic
precluding any denitive conclusion or clinical translation. One gas breathing in clinical practice remains limited owing to added
of the rst study by Cao et al69 reported an increase of blood complexity and side effects such as breathlessness. However, there
volume (BV) in PT 2 weeks into chemoradiotherapy to be as- are now emerging data that suggest that useful information may
sociated with LC. Wang et al70 subsequently reported on still be obtained from ISW MRI, even in the absence of oxygen
a cluster analysis method to identify biologically relevant tu- challenge. A study in cervical cancer by Li et al76 was one of the
mour subvolumes using DCE MRI. The sizes of the cluster rst studies to demonstrate the ability of using baseline tumour
analysis-dened tumour subvolumes with low BV, before and R2* alone to predict response to chemoradiotherapy: responders
during Week-2 radiotherapy, were signicantly greater in the had a lower baseline R2* than non-responders. This study also
patients with local treatment failure (LF) than that in those with found baseline R2* to be an independent prognostic factor for
LC. Whilst the total PT volumes were reduced from baseline to PFS and OS. However, these results are yet to be replicated in
Week 2 to a similar extent for both patients with LF and LC, the other tumour sites.
percentage decreases in the subvolumes of the PTs with low BV
in the same time interval were signicantly smaller for the A study by Panek et al77 has demonstrated tumour R2* mea-
patients with LF than that for those with LC (p , 0.05).70 This surement to be a sensitive and reproducible quantitative imaging
illustrates the potential utility of DCE parameters to identify technique in detecting clinically relevant changes in tumour ox-
a biological target volume for radiotherapy dose escalation and ygenation for HNC. However, reliable interpretation of R2* as
using it to monitor response. a stand-alone parameter is not possible without additional in-
formation such as tumour BV.77 This is supported by other
Baer et al71 investigated the feasibility of using PRM of DCE MRI observations that hypoxic tumours with high blood ow had high
to predict survival following CRT in 10 patients. They found that R2*,78 whereas hypoxic tumours with low BV were found to have
the reduction in Ktrans per voxel measured through PRM after low R2* instead.79 Serial weekly changes in R2* alone throughout
2 weeks of radiotherapy was a better predictor of OS in com- chemoradiotherapy in patients with HNC also did not appear to
parison with the whole tumour mean or median signal changes. show any clear pattern.80 Therefore, a better understanding of
Similar to the DW PRM study, this needs to be validated with how to interpret R2* measurements with BV needs to be ascer-
more patients. The authors themselves acknowledged that the tained to improve its performance as an imaging biomarker.
process to analyze PRM is complex and requires special attention
during delineation owing to limitations in image resolution. Challenges with integration of functional MRI into
clinical practice
Intrinsic susceptibility-weighted MRI Whilst there is little doubt that F-MRI can provide predictive
Hypoxia is a well-recognized factor of resistance to chemo- and prognostic information for HNC, its integration into clin-
therapy and radiotherapy in HNC. A meta-analysis has dem- ical practice remains limited. One of the main reasons is the lack
onstrated hypoxia modication to be a valid therapeutic of consensus of optimal modalities, scanning protocols and
strategy;72 thus, a non-invasive mean to detect tumour hypoxia analytical methodologies, leading to discrepancy in the reported
is highly desirable to guide identication of patients who are results (as described above). This is partly due to the constant

8 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

Figure 4. A schematic summarizing the potential applications of MRI in the management, radiotherapy planning and delivery of
head and neck cancer. BTV, biological target volume; DCE, dynamic contrast-enhanced; DW, diffusion-weighted; F-MRI, functional
MRI; HPV, human papilloma virus; ISW, intrinsic susceptibility-weighted; OAR, organ at risk; RT, radical radiotherapy.

evolution of machinery and acquisition techniques over the which precluded the analysis of PT owing to gross motion ar-
years. Consequently, a meta-analysis of F-MRI studies is not tefact. Consequently, tolerability may be an issue for some
possible for the same reason. Efforts to standardize these pro- patients owing to the additional discomfort with immobilization
tocols for clinical trials are under way and may be further fa- and scanning time required in comparison with standard ana-
cilitated through collaboration between MR-Linac consortiums. tomical MRI. Considerations should also be given to the po-
tential pressure on the hospital resources, e.g. time on scanners
Another important issue to consider is that physiological and additional expertise required to process F-MRI images.
parameters, such as perfusion and oxygenation, are dynamic and
potentially unstable parameters that may uctuate signicantly CONCLUSION
in the absence of therapy. Only limited data exist on the stability Anatomical and functional imaging capabilities offered by MRI
of F-MRI parameters in patients with cancer prior to treatment provide potential opportunities to optimize radiotherapy strat-
and there are no data on this aspect that are specic to patients egies for HNC through improved target delineation, high-risk
with HNC receiving primary CRT. However, two studies in disease subvolume identication, early intratreatment monitor-
patients with a variety of cancers enrolled in Phase 1 clinical drug ing and adaptation based on response (Figure 4). Based on
trials do provide some reassuring data on the reproducibility of current evidence, baseline tumour vascular permeability/
F-MRI parameters. Koh et al81 found ADC measurements from hypoxia and early cell apoptosis during radiotherapy, as measured
DW-MRI to be highly reproducible, with a coefcient of re- by DCE and DW MRI, respectively, appear to be the most
peatability of 13.3%. Messiou et al82 also reported the intrapatient promising biomarkers to predict radiotherapy outcome and
coefcients of variation (CVs) for all DCE-derived parameters to tailor treatment strategies for HNC. The studies conducted to
be within the range of 830%, except for Vp.82 The most re- date have laid a solid foundation for the exciting prospect of
producible DCE-derived parameters were the enhancing fraction integrating functional imaging into MR-Linac in the future.
(CV 5 8.6%), followed by Ktrans (CV 5 13.9%) and initial This added dimension will further enhance the appeal of using
area under the time-concentration curve over 60 seconds MRI as a single platform for radiotherapy planning and de-
(CV 5 15.5%). The stability of these parameters in HNC need to livery in HNC. However, an important next step to bridge the
be established further prior to clinical implementation. translational gaps for F-MRI is a collective effort to determine
the optimal methodology to allow standardization and perform
Furthermore, integration of F-MRI into radiotherapy treatment clinical validation through a large multicentre study.
planning requires special attention in terms of image quality and
geometrical accuracy, which means that image acquisition with FUNDING
immobilization in the radiotherapy treatment position is es- The authors of this article are supported by the Cancer Research
sential. This approach was lacking in most earlier F-MRI studies, UK head and neck programme grant C7224/A13407.

9 of 13 birpublications.org/bjr Br J Radiol;90:20160768
BJR Wong et al

REFERENCES

1. Nutting CM, Morden JP, Harrington KJ, patterns of failure. Int J Radiat Oncol Biol a resolution of 20 ms. NMR Biomed 2010; 23:
Urbano TG, Bhide SA, Clark C, et al. Parotid- Phys 2010; 76: 133946. doi: https://doi.org/ 98694. doi: https://doi.org/10.1002/
sparing intensity modulated versus conven- 10.1016/j.ijrobp.2009.04.006 nbm.1585
tional radiotherapy in head and neck cancer 10. Ahmed M, Schmidt M, Sohaib A, Kong C, 19. Paulson ES, Bradley JA, Wang D, Ahunbay
(PARSPORT): a phase 3 multicentre rando- Burke K, Richardson C, et al. The value of EE, Schultz C, Li XA. Internal margin
mised controlled trial. Lancet Oncol 2011; 12: magnetic resonance imaging in target volume assessment using cine MRI analysis of de-
12736. doi: https://doi.org/10.1016/S1470- delineation of base of tongue tumours glutition in head and neck cancer radiother-
2045(10)70290-4 a study using exible surface coils. Radiother apy. Med Phys 2011; 38: 17409. doi: https://
2. Wippold FJ. Head and neck imaging: the role Oncol 2010; 94: 1617. doi: https://doi.org/ doi.org/10.1118/1.3560418
of CT and MRI. J Magn Reson Imaging 2007; 10.1016/j.radonc.2009.12.021 20. Ding Y, Mohamed ASR, Yang J, Colen RR,
25: 45365. doi: https://doi.org/10.1002/ 11. Emami B, Sethi A, Petruzzelli GJ. Inuence of Frank SJ, Wang J, et al. Prospective observer
jmri.20838 MRI on target volume delineation and IMRT and software-based assessment of magnetic
3. Hsu WC, Loevner LA, Karpati R, Ahmed T, planning in nasopharyngeal carcinoma. Int J resonance imaging quality in head and neck
Mong A, Battineni ML, et al. Accuracy of Radiat Oncol Biol Phys 2003; 57: 4818. doi: cancer: should standard positioning and
magnetic resonance imaging in predicting https://doi.org/10.1016/S0360-3016(03) immobilization be required for radiation
absence of xation of head and neck cancer 00570-4 therapy applications? Pract Radiat Oncol
to the prevertebral space. Head Neck 2005; 12. Rasch C, Keus R, Pameijer FA, Koops W, de 2015; 5: e299308. doi: https://doi.org/
27: 95100. doi: https://doi.org/10.1002/ Ru V, Muller S, et al. The potential impact of 10.1016/j.prro.2014.11.003
hed.20128 CT-MRI matching on tumor volume de- 21. Barker JL, Garden AS, Ang KK, ODaniel JC,
4. Baulch J, Gandhi M, Sommerville J, Panizza lineation in advanced head and neck cancer. Wang H, Court LE, et al. Quantication of
B. 3T MRI evaluation of large nerve peri- Int J Radiat Oncol Biol Phys 1997; 39: 8418. volumetric and geometric changes occurring
neural spread of head and neck cancers. J doi: https://doi.org/10.1016/S0360-3016(97) during fractionated radiotherapy for head-
Med Imaging Radiat Oncol 2015; 59: 57885. 00465-3 and-neck cancer using an integrated CT/
doi: https://doi.org/10.1111/1754-9485.12338 13. Fortunati V, Verhaart RF, Verduijn GM, van linear accelerator system. Int J Radiat Oncol
5. Kato H, Kanematsu M, Watanabe H, Mizuta der Lugt A, Angeloni F, Niessen WJ, et al. Biol Phys 2004; 59: 96070. doi: https://doi.
K, Aoki M. Metastatic retropharyngeal lymph MRI integration into treatment planning of org/10.1016/j.ijrobp.2003.12.024
nodes: comparison of CT and MR imaging head and neck tumors: can patient immobi- 22. Bhide SA, Davies M, Burke K, McNair HA,
for diagnostic accuracy. Eur J Radiol 2014; 83: lization be avoided? Radiother Oncol 2015; Hansen V, Barbachano Y, et al. Weekly volume
115762. doi: https://doi.org/10.1016/j. 115: 1914. doi: https://doi.org/10.1016/j. and dosimetric changes during chemoradio-
ejrad.2014.02.027 radonc.2015.03.021 therapy with intensity-modulated radiation
6. Samuels SE, Vainshtein J, Spector ME, 14. Hanvey S, McJury M, Tho LM, Glegg M, therapy for head and neck cancer: a pro-
Ibrahim M, McHugh JB, Tao Y, et al. Impact Thomson M, Grose D, et al. The inuence of spective observational study. Int J Radiat Oncol
of retropharyngeal adenopathy on distant MRI scan position on patients with oropha- Biol Phys 2010; 76: 13608. doi: https://doi.
control and survival in HPV-related oropha- ryngeal cancer undergoing radical radiother- org/10.1016/j.ijrobp.2009.04.005
ryngeal cancer treated with chemoradiother- apy. Radiat Oncol 2013; 8: 129. doi: https:// 23. Schwartz DL, Garden AS, Thomas J, Chen Y,
apy. Radiother Oncol 2015; 116: 7581. doi: doi.org/10.1186/1748-717X-8-129 Zhang Y, Lewin J, et al. Adaptive radiotherapy
https://doi.org/10.1016/j.radonc.2015.06.006 15. Welsh L, Panek R, McQuaid D, Dunlop A, for head-and-neck cancer: initial clinical
7. Loo SW, Geropantas K, Wilson P, Martin Schmidt M, Riddell A, et al. Prospective, outcomes from a prospective trial. Int J
WMC, Roques TW. Target volume denition longitudinal, multi-modal functional imaging Radiat Oncol Biol Phys 2012; 83: 98693. doi:
for intensity-modulated radiotherapy after for radical chemo-IMRT treatment of locally https://doi.org/10.1016/j.ijrobp.2011.08.017
induction chemotherapy and patterns of advanced head and neck cancer: the INSIGHT 24. Kataria T, Gupta D, Goyal S, Bisht SS, Basu
treatment failure after sequential chemo- study. Radiat Oncol 2015; 10: 112. doi: https:// T, Abhishek A, et al. Clinical outcomes of
radiotherapy in locoregionally advanced doi.org/10.1186/s13014-015-0415-7 adaptive radiotherapy in head and
oropharyngeal squamous cell carcinoma. 16. Clayman DA, Murakami ME, Vines FS. neck cancers. Br J Radiol 2016; 89:
Clin Oncol (R Coll Radiol) 2013; 25: 16270. Compatibility of cervical spine braces with 201600856. doi: https://doi.org/10.1259/
doi: https://doi.org/10.1016/j. MR imaging: a study of nine nonferrous bjr.20160085
clon.2012.07.015 devices. AJNR Am J Neuroradiol 1990; 25. Olteanu LA, Berwouts D, Madani I, De
8. Sanguineti G, Gunn GB, Endres EJ, Chaljub 11: 38590. Gersem W, Vercauteren T, Duprez F, et al.
G, Cheruvu P, Parker B. Patterns of locore- 17. Schmidt MA, Panek R, Colgan R, Hughes J, Comparative dosimetry of three-phase
gional failure after exclusive IMRT for Sohaib A, Saran F, et al. Slice encoding for adaptive and non-adaptive dose-painting
oropharyngeal carcinoma. Int J Radiat Oncol metal artefact correction in magnetic reso- IMRT for head-and-neck cancer. Radiother
Biol Phys 2008; 72: 73746. doi: https://doi. nance imaging examinations for radiotherapy Oncol 2014; 111: 34853. doi: https://doi.org/
org/10.1016/j.ijrobp.2008.01.027 planning. Radiother Oncol 2016; 120: 35662. 10.1016/j.radonc.2014.02.017
9. Daly ME, Le QT, Maxim PG, Loo BW, doi: https://doi.org/10.1016/j. 26. Chen L, Liu M, Bao J, Xia Y, Zhang J, Zhang
Kaplan MJ, Fischbein NJ, et al. Intensity- radonc.2016.05.004 L, et al. The correlation between apparent
modulated radiotherapy in the treatment of 18. Uecker M, Zhang S, Voit D, Karaus A, diffusion coefcient and tumor cellularity in
oropharyngeal cancer: clinical outcomes and Merboldt KD, Frahm J. Real-time MRI at patients: a meta-analysis. PLoS One 2013; 8:

10 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

e790089. doi: https://doi.org/10.1371/jour- treatment response. Eur Radiol 2010; 20: 44. Ohnishi K, Shioyama Y, Hatakenaka M,
nal.pone.0079008 221320. doi: https://doi.org/10.1007/ Nakamura K, ABE K, Yoshiura T, et al.
27. Chawla S, Kim S, Wang S, Poptani H. s00330-010-1769-8 Prediction of local failures with a combina-
Diffusion-weighted imaging in head and 37. Matoba M, Tuji H, Shimode Y, Toyoda I, tion of pretreatment tumor volume and
neck cancers. Future Oncol 2009; 5: 95975. Kuginuki Y, Miwa K, et al. Fractional change apparent diffusion coefcient in patients
doi: https://doi.org/10.2217/fon.09.77 in apparent diffusion coefcient as an treated with denitive radiotherapy for
28. Vandecaveye V, De Keyzer F. Evaluation of imaging biomarker for predicting treatment hypopharyngeal or oropharyngeal squamous
the larynx for tumour recurrence by diffu- response in head and neck cancer treated cell carcinoma. J Radiat Res 2011; 52: 52230.
sion-weighted MRI after radiotherapy: initial with chemoradiotherapy. AJNR Am J Neuro- doi: https://doi.org/10.1269/jrr.10178
experience in four cases. Br J Radiol 2014; radiol 2014; 35: 37985. doi: https://doi.org/ 45. Ng SH, Lin CY, Chan SC, Lin YC, Yen TC,
79: 6817. 10.3174/ajnr.A3706 Liao CT, et al. Dynamic contrast-enhanced
29. Moeller BJ, Rana V, Cannon BA, Williams 38. Berrak S, Chawla S, Kim S, Quon H, MRI, diffusion-weighted MRI and 18F-FDG
MD, Sturgis EM, Ginsberg LE, et al. Pro- Sherman E, Loevner LA, et al. Diffusion PET/CT for the prediction of survival in
spective risk-adjusted [18F]uorodeoxyglu- weighted imaging in predicting progression oropharyngeal or hypopharyngeal squamous
cose positron emission tomography and free survival in patients with squamous cell cell carcinoma treated with chemoradiation.
computed tomography assessment of radia- carcinomas of the head and neck treated with Eur Radiol 2016; 26: 416272. doi: https://
tion response in head and neck cancer. J Clin induction chemotherapy. Acad Radiol 2011; doi.org/10.1007/s00330-016-4276-8
Oncol 2009; 27: 250915. doi: https://doi.org/ 18: 122532. doi: https://doi.org/10.1016/j. 46. Nakajo M, Nakajo M, Kajiya Y, Tani A,
10.1200/JCO.2008.19.3300 acra.2011.06.009 Kamiyama T, Yonekura R, et al. FDG PET/CT
30. Schakel T, Hoogduin JM, Terhaard CH, 39. Wong KH, Panek R, Welsh L, McQuaid D, and diffusion-weighted imaging of head and
Philippens ME. Diffusion weighted MRI in Dunlop A, Riddell A, et al. The predictive neck squamous cell carcinoma: comparison
head-and-neck cancer: geometrical accuracy. value of early assessment after 1 cycle of of prognostic signicance between primary
Radiother Oncol 2013; 109: 3947. doi: induction chemotherapy with 18F-FDG PET/ tumor standardized uptake value and appar-
https://doi.org/10.1016/j.radonc.2013.10.004 CT and diffusion-weighted MRI for response ent diffusion coefcient. Clin Nucl Med 2012;
31. Jezzard P, Balaban RS. Correction for geo- to radical chemoradiotherapy in head and 37: 47580. doi: https://doi.org/10.1097/
metric distortion in echo planar images from neck squamous cell carcinoma. J Nucl Med RLU.0b013e318248524a
B0 eld variations. Magn Reson Med 1995; 2016; 57: 184350. doi: https://doi.org/ 47. El-Mofty SK, Lu DW. Prevalence of human
34: 6573. doi: https://doi.org/10.1002/ 10.2967/jnumed.116.174433 papillomavirus type 16 DNA in squamous
mrm.1910340111 40. Kim S, Loevner L, Quon H, Sherman E, cell carcinoma of the palatine tonsil, and not
32. Wu M, Chang LC, Walker L, Lemaitre H, Weinstein G, Kilger A, et al. Diffusion- the oral cavity, in young patients: a distinct
Barnett AS, Marenco S, et al. Comparison of weighted magnetic resonance imaging for clinicopathologic and molecular disease en-
EPI distortion correction methods in diffu- predicting and detecting early response to tity. Am J Surg Pathol 2003; 27: 146370.
sion tensor MRI using a novel framework. chemoradiation therapy of squamous cell 48. Chernock RD, El-Mofty SK, Thorstad WL,
Med Image Comput Comput Assist Interv carcinomas of the head and neck. Clin Cancer Parvin CA, Lewis JS. HPV-related nonker-
2008; 11: 3219. Res 2009; 15: 98694. doi: https://doi.org/ atinizing squamous cell carcinoma of the
33. Koyasu S, Iima M, Umeoka S, Morisawa N, 10.1158/1078-0432.CCR-08-1287 oropharynx: utility of microscopic features in
Porter DA, Ito J, et al. The clinical utility of 41. Hatakenaka M, Nakamura K, Yabuuchi H, predicting patient outcome. Head Neck Pathol
reduced-distortion readout-segmented echo- Shioyama Y, Matsuo Y, Ohnishi K, et al. 2009; 3: 18694. doi: https://doi.org/10.1007/
planar imaging in the head and neck region: Pretreatment apparent diffusion coefcient of s12105-009-0126-1
initial experience. Eur Radiol 2014; 24: the primary lesion correlates with local 49. Vandecaveye V, Dirix P, De Keyzer F, de Beeck
308896. doi: https://doi.org/10.1007/ failure in head-and-neck cancer treated with KO, Vander Poorten V, Roebben I, et al.
s00330-014-3369-5 chemoradiotherapy or radiotherapy. Int J Predictive value of diffusion-weighted mag-
34. Schouten CS, de Bree R, van der Putten L, Radiat Oncol Biol Phys 2011; 81: 33945. doi: netic resonance imaging during chemoradio-
Noij DP, Hoekstra OS, Comans EFI, et al. https://doi.org/10.1016/j.ijrobp.2010.05.051 therapy for head and neck squamous cell
Diffusion-weighted EPI- and HASTE-MRI 42. Ng SH, Lin CY, Chan SC, Lin YC, Yen TC, carcinoma. Eur Radiol 2010; 20: 170314. doi:
and 18F-FDG-PET-CT early during chemo- Liao CT, et al. Clinical utility of multi- https://doi.org/10.1007/s00330-010-1734-6
radiotherapy in advanced head and neck modality imaging with dynamic contrast- 50. Padhani AR, Koh DM, Collins DJ. Whole-
cancer. Quant Imaging Med Surg 2014; 4: enhanced MRI, diffusion-weighted MRI, and body diffusion-weighted MR imaging in
23950. doi: https://doi.org/10.3978/j. 18F-FDG PET/CT for the prediction of neck cancer: current status and research direc-
issn.2223-4292.2014.07.15 control in oropharyngeal or hypopharyngeal tions. Radiology 2011; 261: 70018. doi:
35. Chawla S, Kim S, Dougherty L, Wang S, squamous cell carcinoma treated with che- https://doi.org/10.1148/radiol.11110474
Loevner LA, Quon H, et al. Pretreatment moradiation. PLoS One 2014; 9: e115933. 51. Galban CJ, Mukherji SK, Chenevert TL,
diffusion-weighted and dynamic contrast- doi: https://doi.org/10.1371/journal. Meyer CR. A feasibility study of parametric
enhanced MRI for prediction of local treat- pone.0115933 response map analysis of diffusion-weighted
ment response in squamous cell carcinomas 43. Kato H, Kanematsu M, Tanaka O, Mizuta K, magnetic resonance imaging scans of head
of the head and neck. AJNR Am J Neuroradiol Aoki M, Shibata T, et al. Head and neck and neck cancer patients for providing early
2013; 200: 3543. squamous cell carcinoma: usefulness of detection of therapeutic efcacy. Transl Oncol
36. King AD, Mo FK, Yu KH, Yeung DK, Zhou diffusion-weighted MR imaging in the pre- 2009; 2: 18490.
H, Bhatia KS, et al. Squamous cell carcinoma diction of a neoadjuvant therapeutic effect. 52. Hulka CA, Smith BL, Sgroi DC, Tan L,
of the head and neck: diffusion-weighted MR Eur Radiol 2009; 19: 1039. doi: https://doi. Edmister WB, Semple JP, et al. Benign and
imaging for prediction and monitoring of org/10.1007/s00330-008-1108-5 malignant breast lesions: differentiation with

11 of 13 birpublications.org/bjr Br J Radiol;90:20160768
BJR Wong et al

echo-planar MR imaging. Radiology 1995; 61. Kim S, Loevner LA, Quon H, Kilger A, of outcome in advanced head-and-neck
197: 338. doi: https://doi.org/10.1148/ Sherman E, Weinstein G, et al. Prediction of cancer based on tumor blood volume alter-
radiology.197.1.7568850 response to chemoradiation therapy in ations during therapy: a prospective study.
53. Buckley DL, Drew PJ, Mussurakis S, Monson squamous cell carcinomas of the head and Int J Radiat Oncol Biol Phys 2008; 72:
JR, Horsman A. Microvessel density of neck using dynamic contrast-enhanced MR 128790. doi: https://doi.org/10.1016/j.
invasive breast cancer assessed by dynamic imaging. AJNR Am J Neuroradiol 2010; 31: ijrobp.2008.08.024
Gd-DTPA enhanced MRI. J Magn Reson 2628. doi: https://doi.org/10.3174/ 70. Wang P, Popovtzer A, Eisbruch A, Cao Y. An
Imaging 1997; 7: 4614. doi: https://doi.org/ ajnr.A1817 approach to identify, from DCE MRI,
10.1002/jmri.1880070302 62. Shukla-Dave A, Lee NY, Jansen JF, Thaler HT, signicant subvolumes of tumors related to
54. Konouchi H, Asaumi JI, Yanagi Y, Shigehara Stambuk HE, Fury MG, et al. Dynamic outcomes in advanced head-and-neck cancer.
H, Hisatomi M, Matsuzaki H, et al. Evalua- contrast-enhanced magnetic resonance im- Med Phys 2012; 39: 52779. doi: https://doi.
tion of tumor proliferation using dynamic aging as a predictor of outcome in head-and- org/10.1118/1.4737022
contrast enhanced-MRI of oral cavity and neck squamous cell carcinoma patients with 71. Baer AH, Hoff BA, Srinivasan A, Galban CJ,
oropharyngeal squamous cell carcinoma. nodal metastases. Int J Radiat Oncol Biol Phys Mukherji SK. Feasibility analysis of the
Oral Oncol 2003; 39: 2905. doi: https://doi. 2012; 82: 183744. parametric response map as an early pre-
org/10.1016/S1368-8375(02)00119-7 63. Han M, Kim SY, Lee SJ, Choi JW. The dictor of treatment efcacy in head and neck
55. Newbold K, Castellano I, Charles-Edwards E, correlations between MRI perfusion, diffu- cancer. AJNR Am J Neuroradiol 2015; 36:
Mears D, Sohaib A, Leach M, et al. An sion parameters, and 18F-FDG PET meta- 75762. doi: https://doi.org/10.3174/
exploratory study into the role of dynamic bolic parameters in primary head-and-neck ajnr.A4296
contrast-enhanced magnetic resonance im- cancer: a cross-sectional analysis in single 72. Overgaard J. Hypoxic modication of radio-
aging or perfusion computed tomography for institute. Medicine (Baltimore) 2015; 94: therapy in squamous cell carcinoma of the
detection of intratumoral hypoxia in head- e2141. doi: https://doi.org/10.1097/ head and necka systematic review and
and-neck cancer. Int J Radiat Oncol Biol Phys md.0000000000002141 meta-analysis. Radiother Oncol 2011; 100:
2009; 74: 2937. doi: https://doi.org/10.1016/ 64. King AD, Chow SK, Yu KH, Mo FK, Yeung 2232. doi: https://doi.org/10.1016/j.
j.ijrobp.2008.07.039 DK, Yuan J, et al. DCE-MRI for pre- radonc.2011.03.004
56. Egeland TA, Gaustad JV, Vestvik IK, Benja- treatment prediction and post-treatment 73. Wahl RL, Herman JM, Ford E. The promise
minsen IC, Mathiesen B, Rofstad EK. As- assessment of treatment response in sites of and pitfalls of positron emission tomography
sessment of fraction of radiobiologically squamous cell carcinoma in the head and and single-photon emission computed to-
hypoxic cells in human melanoma xenografts neck. PLoS One 2015; 10: e0144770. doi: mography molecular imaging-guided radia-
by dynamic contrast-enhanced MRI. Magn https://doi.org/10.1371/journal. tion therapy. Semin Radiat Oncol 2011; 21:
Reson Med 2006; 55: 87482. doi: https://doi. pone.0144770 88100. doi: https://doi.org/10.1016/j.
org/10.1002/mrm.20852 65. Parker GJ, Roberts C, Macdonald A, Buo- semradonc.2010.11.004
57. Hoskin PJ, Saunders MI, Goodchild K, naccorsi GA, Cheung S, Buckley DL, et al. 74. Rijpkema M, Kaanders J, Joosten F, van der
Powell ME, Taylor NJ, Baddeley H. Dynamic Experimentally-derived functional form for Kogel AJ, Heerschap A. Effects of breathing
contrast enhanced magnetic resonance scan- a population-averaged high-temporal-reso- a hyperoxic hypercapnic gas mixture on
ning as a predictor of response to accelerated lution arterial input function for dynamic blood oxygenation and vascularity of head-
radiotherapy for advanced head and neck contrast-enhanced MRI. Magn Reson Med and-neck tumors as measured by magnetic
cancer. Br J Radiol 1999; 72: 10938. doi: 2006; 56: 9931000. doi: https://doi.org/ resonance imaging. Int J Radiat Oncol Biol
https://doi.org/10.1259/bjr.72.863.10700827 10.1002/mrm.21066 Phys 2002; 53: 118591. doi: https://doi.org/
58. Bernstein JM, Homer JJ, West CM. Dynamic 66. Yu Y, Jiang Q, Miao Y, Li J, Bao S, Wang H, et 10.1016/S0360-3016(02)02825-0
contrast-enhanced magnetic resonance im- al. Quantitative analysis of clinical dynamic 75. Kotas M, Schmitt P, Jakob PM, Flentje M.
aging biomarkers in head and neck cancer: contrast-enhanced MR imaging for evaluat- Monitoring of tumor oxygenation changes in
potential to guide treatment? A systematic ing treatment response in human breast head-and-neck carcinoma patients breathing
review. Oral Oncol 2014; 50: 96370. doi: cancer. Radiology 2010; 257: 4755. doi: a hyperoxic hypercapnic gas mixture with
https://doi.org/10.1016/j. https://doi.org/10.1148/radiol.10092169 a noninvasive MRI technique. Strahlenther
oraloncology.2014.07.011 67. McGrath DM, Bradley DP, Tessier JL, Lacey Onkol 2009; 185: 1926. doi: https://doi.org/
59. Noij DP, de Jong MC, Mulders LG, Marcus T, Taylor CJ, Parker GJ. Comparison of 10.1007/s00066-009-1870-6
JT, de Bree R, Lavini C, et al. Contrast- model-based arterial input functions for 76. Li XS, Fan HX, Fang H, Song YL, Zhou CW.
enhanced perfusion magnetic resonance im- dynamic contrast-enhanced MRI in tumor Value of R2* obtained from T2*-weighted
aging for head and neck squamous cell bearing rats. Magn Reson Med 2009; 61: imaging in predicting the prognosis of
carcinoma: a systematic review. Oral Oncol 117384. doi: https://doi.org/10.1002/ advanced cervical squamous carcinoma trea-
2015; 51: 12438. doi: https://doi.org/ mrm.21959 ted with concurrent chemoradiotherapy. J
10.1016/j.oraloncology.2014.10.016 68. Mehrabian H, Chandrana C, Pang I, Chopra Magn Reson Imaging 2015; 42: 6818. doi:
60. Chawla S, Kim S, Loevner LA, Hwang WT, R, Martel AL. Arterial input function calcu- https://doi.org/10.1002/jmri.24837
Weinstein G, Chalian A, et al. Prediction of lation in dynamic contrast-enhanced MRI: an 77. Panek R, Welsh L, Dunlop A, Wong KH,
disease-free survival in patients with squa- in vivo validation study using co-registered Riddell AM, Koh DM, et al. Repeatability
mous cell carcinomas of the head and neck contrast-enhanced ultrasound imaging. Eur and sensitivity of T2* measurements in
using dynamic contrast-enhanced MR imag- Radiol 2012; 22: 173547. doi: https://doi. patients with head and neck squamous cell
ing. AJNR Am J Neuroradiol 2011; 32: org/10.1007/s00330-012-2418-1 carcinoma at 3T. J Magn Reson Imaging
77884. doi: https://doi.org/10.3174/ 69. Cao Y, Popovtzer A, Li D, Chepeha DB, 2016; 44: 7280. doi: https://doi.org/
ajnr.A2376 Moyer JS, Prince ME, et al. Early prediction 10.1002/jmri.25134

12 of 13 birpublications.org/bjr Br J Radiol;90:20160768
Review article: MRI in head and neck radiotherapy BJR

78. Taylor NJ, Baddeley H, Goodchild KA, Powell 80. Min M, Lee MT, Lin P, Holloway L, coefcients of solid tumours treated with
ME, Thoumine M, Culver LA, et al. BOLD Wijesekera D, Gooneratne D, et al. As- combretastatin A4 phosphate and bevacizu-
MRI of human tumor oxygenation during sessment of serial multi-parametric func- mab in a two-centre Phase I clinical trial. Eur
carbogen breathing. J Magn Reson Imaging tional MRI (diffusion-weighted imaging Radiol 2009; 19: 272838. doi: https://doi.
2001; 14: 15663. doi: https://doi.org/ and R2*) with 18F-FDG-PET in patients org/10.1007/s00330-009-1469-4
10.1002/jmri.1166 with head and neck cancer treated with 82. Messiou C, Orton M, Ang JE, Collins DJ,
79. Rodrigues LM, Howe FA, Grifths JR, radiation therapy. Br J Radiol 2016; 89: Morgan VA, Mears D, et al. Advanced solid
Robinson SP. Tumor R2* is a prognostic 20150530. doi: https://doi.org/10.1259/ tumors treated with cediranib: comparison of
indicator of acute radiotherapeutic response bjr.20150530 dynamic contrast-enhanced MR imaging and
in rodent tumors. J Magn Reson Imaging 81. Koh DM, Blackledge M, Collins DJ, Padhani CT as markers of vascular activity. Radiology
2004; 19: 4828. doi: https://doi.org/10.1002/ AR, Wallace T, Wilton B, et al. Reproduc- 2012; 265: 42636. doi: https://doi.org/
jmri.20024 ibility and changes in the apparent diffusion 10.1148/radiol.12112565

13 of 13 birpublications.org/bjr Br J Radiol;90:20160768

You might also like