ArticlePDF Available

Opportunities and Challenges of 7 Tesla Magnetic Resonance Imaging: A Review

Authors:

Abstract

The desire to achieve clinical ultra-high magnetic resonance imaging (MRI) systems stems from the fact that higherfield strength leads to higher signal-to-noise ratio (SNR), contrast-to-noise ratio (CNR), and spatial resolution. During last few years 7T MRI systems have become a quasi standard for ultra-high field MRI (UhFMRI) systems. This review presents a detailed account of opportunities and challenges associated with a clinical 7T MRI system for cranial and extracranial imaging. As with all of the previous transitions to higher field strengths, the switch from high to UhFMRI is not easy. The engineering and scientific community have to overcome challenges like magnetic field inhomogeneity, patient safety and comfort issues, and cost and related problems in order to achieve a clinically viable UhFMRI system. In addition, a large number of clinical studies are still required to show the improvements in quality of diagnostics that would come with 7T MRI, in order to bring such a research tool to the clinic.
Critical ReviewsTM in Biomedical Engineering, 44 (1-2): 73–89 (2016)
Opportunities and Challenges of 7 Tesla Magnetic
Resonance Imaging: A Review
Muhammad Irfan Karamat,1,Sahar Darvish-Molla,1& Alejandro Santos-Diaz2,3
1Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada;
2McMaster School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; 3Imaging Research Centre,
St. Joseph’s Healthcare, Hamilton, ON, Canada
All authors contributed equally to this paper.
Address all correspondence to: Muhammad Irfan Karamat, Department of Medical Physics and Applied Radiation Sciences, McMaster University,
1280 Main Street W, Hamilton, ON, Canada L8S 4K1; Tel.: +1 905 521 2100 ext. 73389; karamami@mcmaster.ca
ABSTRACT: The desire to achieve clinical ultra-high magnetic resonance imaging (MRI) systems stems from the fact that higher
field strength leads to higher signal-to-noise ratio (SNR), contrast-to-noise ratio (CNR), and spatial resolution. During last few years
7T MRI systems have become a quasi standard for ultra-high field MRI (UhFMRI) systems. This review presents a detailed account
of opportunities and challenges associated with a clinical 7T MRI system for cranial and extracranial imaging. As with all of the
previous transitions to higher field strengths, the switch from high to UhFMRI is not easy. The engineering and scientific community
have to overcome challenges like magnetic field inhomogeneity, patient safety and comfort issues, and cost and related problems
in order to achieve a clinically viable UhFMRI system. In addition, a large number of clinical studies are still required to show the
improvements in quality of diagnostics that would come with 7T MRI, in order to bring such a research tool to the clinic.
KEY WORDS: ultra high field magnetic resonance imaging, 7T MRI system, B0inhomogeneity, B1inhomogeneity, specific ab-
sorption rate, brain MRI, extracranial MRI, magnetic resonance spectroscopy
ABBREVIATIONS: AD, Alzheimer’s disease; CE-MRI, contrast-enhanced MRI; CNR, contrast-to-noise; CSLE, chemical shift
localization error; DIR, double inversion recovery; DWI, diffusion-weighted imaging; FDA, Federal Drug Agency; FLAIR, fluid at-
tenuation inversion recovery; FLASH, fast low-angle shot; FSE, fast spin-echo; FSPGR, fast spoiled gradient echo; GRAPPA, gen-
eralized autocalibrating partially parallel acquisition; MPRAGE, magnetization prepared rapid gradient echo; MRS, magnetic reso-
nance spectroscopy; MS, multiple Sclerosis; PD, Parkinson’s disease; RF, radiofrequency; SAR, specific absorption rate; SENSE,
sensitivity encoding; SMASH, simultaneous acquisition of spatial harmonics; SNR, signal-to-noise; SSFP, steady state free pre-
cession; TOF-MRA, time of flight MR angiography; TSE, turbo spin-echo; UhFMRI, ultra-high field MRI; VIBE, volumetric
interpolated breath-hold examination; VOI, volume of interest
I. INTRODUCTION
Since the beginning of the magnetic resonance imag-
ing (MRI) era in the 20th latter part of the century, an
urge to achieve the highest possible magnetic field in
clinical settings has been a major motivation for sci-
entists and engineers.1,2The efforts towards achiev-
ing higher magnetic field stem from the fact that
higher field strength implies higher signal-to-noise
(SNR), contrast-to-noise (CNR), and spatial resolu-
tion. However, transition from the first generation
MRI scanners (0.5T) to the conventional (1.0–1.5T)
and then to high field 3T scanners was gradual with
each transition presenting a different set of challenges
for the scientific and engineering community.3The
transition from conventional to high magnetic field
(3.0T) clinical MRI scanners was not straightforward
either. It took almost 15 years before being accepted
widely by the clinical community. Over the years,
many ultra-high field MRI (UhFMRI) systems rang-
ing from 7 to 11.7T have been designed, but a 7T sys-
tem has evolved as a quasi standard currently.2There-
fore, in this review UhFMRI will refer to a 7T scanner
unless stated otherwise. In order to reach the goal of
clinical 7T MRI scanner and to gain the above men-
tioned advantages, a considerable amount of work has
been done and efforts are still continued by the re-
search and scientific community to solve problems
like safety, susceptibility related artifacts, B0and B1
inhomogeneities and other technical issues. The cur-
0278–940X/16/$35.00 c
°2016 by Begell House, Inc. www.begellhouse.com 73
74 Karamat, Darvish-Molla, & Santos-Diaz
rent review focuses on the efforts and developments
towards transformation of a 7T MR imaging system
from a topic of research to a routinely used clinical
MR imaging system. In this regard, a detailed account
of opportunities, challenges and the ways proposed
to overcome these challenges, and clinical studies to
identify possible advantages offered by UhFMRI in
cranial and extracranial imaging applications with an
intent to achieve a clinical 7T system, is given in the
review.
II. 7T MRI: OPPORTUNITIES
A. Enhancement of Signal-to-Noise Ratio
(SNR)
There is a lack of consensus among MRI scientists
about the definition of SNR. However, it is generally
accepted that SNR is directly proportional to the field
strength B0.1,3There are still some who think SNR
increases more steeply with B0:4
SNR B7/4
0.(1)
To be on the safe side, one assumes a linear relation-
ship between SNR and B0. Under such an assump-
tion, the difference between parallel and antiparal-
lel spins, calculated using the Boltzmann probabil-
ity distribution, is as small as 10 spins in one mil-
lion at B0=3T that implies around 2.3 times (i.e.,
23 spins in one million) more spins per million at
7T.1,3Such an increase in SNR with B0implies more
refined spin manipulation and image reconstruction
with fewer spins. In addition, greater SNR provides a
signal enhancement to nuclei other than protons, such
as sodium-23 (23Na) and phosphorus-31 (31 P).5The
signal boost given to these nuclei by UhFMRI pro-
vides a mean to study novel relaxation mechanisms,
different metabolic pathways, and cellular phenom-
ena.
B. Spatial Resolution
The UhFMRI provides SNR that can be spent on res-
olution. It has been theorized that the spatial resolu-
tion is related to a factor Nby which field B0is in-
creased as:5
Spatial Resolution N1/3.(2)
The above relationship implies about 30% improve-
ment in resolution when B0is changed from 3T to
7T. It must be emphasized here that resolution is
not dependent only on B0and can further be en-
hanced in practice by improvements in receiver and
gradient design and by the use of imaging sequence
that involves improvement in contrast-to-noise ratio.5
This increase in spatial resolution promises to reveal
anatomical and pathological details that were never
resolved before by other lower-field MR imaging sys-
tems. The examples of such revolutionary detail de-
piction by UhMRI are given in almost all the follow-
ing sections of this review.
C. Enhancement of T1 Relaxation
The prolongation of T1relaxation time can be gener-
alized as a heuristic relation based on human brain
studies or extrapolated from animal brain studies
as:5,6
T1 = 1
a1+a2Ba3
0
.(3)
In order to give the reader an idea about the T1pro-
longation, constants a1,a2, and a3for cortical gray
matter are found approximately to be 0.35, 0.64, and
–0.7, respectively, in SI units.79This suggests an
overall prolongation of about 26% in the case of 7T
MRI compared to 3T MRI. This prolongation of T1
relaxation time with field strength may be regarded
as a disadvantage, as longer T1leads to lesser SNR
recovery per unit time. However, the increase in T1
in applications that involve labeling of magnetiza-
tion before imaging can be an advantage as it implies
less decay of the label. Arterial spin labeling that in-
volves labeling of blood to obtain perfusion weighted
images10 and cardiac tagging, in which saturation of
magnetization lines enable visualization of cardiac
muscle contraction over the cardiac cycle,11 are the
examples of imaging applications in which prolonga-
tion of T1is an advantage. Another such application
where increase in T1value could be advantageous is
the time-of-flight angiography where the background
signal is suppressed with the help of a relatively high
flip angle and repetition times much shorter than T1
are used to prevent signal recovery.1214 Such back-
ground suppression enables improved signal from the
fresh blood entering the tissue that has not previously
experienced RF pulse.
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 75
D. Reduction of T2 Relaxation
A heuristic expression for T2similar to that de-
scribed for T1[Eq. (3)] in Sec. II.C. can be written
as:5,6
T2=1
a4+a5B0
.(4)
In the case of cortical gray matter a4and a5are
7 and 3.5, respectively, in SI units.79This rela-
tion implies a reduction in T2by a factor of about
0.56 at 7T compared to 3T for cortical gray matter.
The shortening of the T2time constant at UhFMRI
makes it more sensitive to the presence of calcium
and iron in microbleeds and hemorrhages. This leads
to an improved depiction rate of microbleeds in
patients,1516 detection of small venules17,18 and
envisioning of deep brain structures.19,20 Also, in-
creased blood oxygen level dependent (BOLD) con-
trast (due to enhanced T2dephasing caused by de-
oxygenated blood)21 and better visualization of cap-
illary beds (due to higher SNR)22,23 enables high-
resolution functional MRI (fMRI) at the highest field
strength possible.
E. Enhancement of the Chemical Shift
The difference in molecular distribution of different
compounds results in different resonance frequen-
cies for the compounds. This molecular distribution
based variation of resonance frequencies is known
as chemical shift. The extent of resonance frequency
spread, 4ν, is directly proportional to field strength
B0i.e.,4,5
4ν∝ 4B0.(5)
According to Eq. (5), molecular distribution based
spectral changes become more pronounced at higher
field and provide higher spectral resolution in ap-
plications involving MR spectroscopy in UhFMRI.
Higher spectral resolution and increased SNR would
result in a greater number of metabolites detected at
7T compared to lower field strengths.5Such an in-
crease in chemical shift at UhFMRI is also beneficial
in achieving better suppression due to considerable
enhancement in frequency differences between water
and fat at higher field strength.24
F. Magnetic Susceptibility Effects
The magnetic susceptibility (χ) is the inherent prop-
erty of the tissue or material that describes the re-
sponse of that tissue to external magnetic field.3The
sensitivity of χto magnetic field strength, B0, is de-
picted in the following expression (6).5
χB0×T E (6)
The expression (6) shows that at higher B0, shorter
TEs can be used to achieve the same phase effect,
which in conjunction with higher SNR can be used
to achieve susceptibility weighted images (SWI) at
higher resolution and contrast.3,5This increase in χ-
effects with increase in B0allows more effective de-
piction of microvasculatures, microbleeds, iron and
calcium deposits25 and better differentiation between
blood containing iron and calcification.26 Also, it is
worth mentioning here that SNR in phase images in-
creases quadratically with field strength that can help
to achieve an order of magnitude better CNR in phase
images compared to magnitude images in brain.27
III. CHALLENGES OF 7T MRI
In order to take full advantage of the opportunities de-
scribed in Sec. II and to develop MRI at 7T in clini-
cal practice, a number of technical, patient safety, and
practical issues need to be addressed. The technical
issues include B0inhomogeneity, B1inhomogene-
ity, susceptibility related artifacts and chemical shift
error. The quadratic increase in radiofrequency (RF)
specific absorption rate (SAR) with B0is the main
patient safety concern at UhFMRI.3The location of
7T MRI and its cost are practical considerations in-
volved in clinical 7T MRI system. Some additional
patient comfort and safety requirements of UhFMRI
include noise level, peripheral nerve stimulation, and
other ultra-high field related physiological effects that
will also be addressed below.
A. Technical Challenges
B0Inhomogeneity. The magnetic susceptibility, χ,
scales linearly with B0, as does the inhomogeneity in
B0.3,5The inhomogeneities in B0in UhFMRI lead
to geometrical and intensity related distortions in the
Volume 44, Issues 1-2, 2016
76 Karamat, Darvish-Molla, & Santos-Diaz
MR images. Especially, rapid single- or few-shot ac-
quisition schemes are more prone to susceptibility re-
lated artifacts that mainly lead to geometric distortion
caused by the B0inhomogeneity. The B0variations
in UhFMRI also introduce not only shifts, but also
cause broadening of metabolite peaks in MR spec-
troscopic applications. It also makes it extremely dif-
ficult to use selective frequency pulses designed for
spectral band specific data sampling. The intensity
distortions caused by B0inhomogeneity also make
fat and water suppression relatively more problem-
atic in UhFMRI. More powerful referencing schemes,
dynamic B0shimming and smaller voxel size are re-
quired to overcome the B0inhomogeneity issue.5
B1Inhomogeneity. Is the most difficult prob-
lem to solve at ultra-high B0.3,5The wavelength
of RF transmit field in UhFMRI becomes so small
(11.2 cm at 7T) that the phase effect of the hu-
man body no longer remains negligible at these
wavelengths.3,28 These material dependent phase
changes caused by the reduction in wavelength (due
to higher RF frequency required at UhFMRI) result
in nonuniform flip angle distribution and unexpected
changes in SNR and CNR over the volume of in-
terest (VOI). The central brightening within brain
commonly manifests due to B1inhomogeneity in
UhFMRI.
One possible solution proposed to overcome B1
inhomogeneity is to use specially designed adiabatic
pulses to obtain uniform flip angle distribution, in-
dependent of B1inhomogeneity, above a certain am-
plitude known as “adiabatic threshold”.29 There are
pulse sequence design methods available to tailor adi-
abatic pulses that are behaviorally similar to conven-
tional RF pulses.30
Another solution offered to overcome B1inhomo-
geneity is to use specifically designed RF pulses tai-
lored using rapidly acquired accurate B1maps. Even
though these 2D pulses are used to compensate for
B1inhomogeneity at reduced RF power consumption
compared to adiabatic pulses, they may get affected
by B0inhomogeneity.31,32
Use of multichannel parallel transmit arrays also
offers not only a method to mitigate B1inhomogene-
ity, they also help to overcome SAR related issues.
These arrays divide VOI into partially overlapping
spatial regions and overcome B1inhomogeneity ei-
ther by (1). changing RF amplitude or phase (B1
shimming) at each transmit channel to compensate
for B1inhomogeneity, or (2) using customized RF
pulse for each of the transmit channels to overcome
B1variations.28,33
Chemical Shift Localization Error (CSLE). Is
the measure of spatial offset in precise location
with RF frequency and resonance frequency of the
metabolite in MR spectroscopy within VOI, specified
at the scanner. The CSLE varies as:5
CSLE B0×Slice thickness
(Transmit RF bandwidth).(7)
As depicted in Eq. (7), the CSLE is proportional to
field strength and slice thickness and inversely pro-
portional to transmit RF bandwidth. CSLE scales lin-
early with frequency shift which leads to a reduction
of usable volume for MR spectroscopy, due to spatial
offset introduced by multiple metabolites involved in
spectroscopic data acquisition at 7T.5
B. Patient Safety Challenge
The SAR increases with the square of B0which puts
a more stringent limit on the number, duration, and
amplitude of applied RF pulse at 7T compared to 3T
for a given time period. The use of MRI pulse se-
quences like turbo spin-echo (TSE) or fast spin-echo
(FSE) that involve dense high flip-angle RF pulses
also limits the number of image slices that can be ob-
tained at 7T.3,5
Parallel imaging, described earlier in this section,
with segmented readout to accelerate image data ac-
quisition used to mitigate field inhomogeneity, can
also be used to overcome some limitations posed
by SAR.34 In order to reduce RF energy deposition
lower-flip angle sequences have also been designed
with an intent to achieve contrast similar to more SAR
intensive pulse sequences like TSE or FSE.35
C. Practical Considerations
Siting of whole body 7T clinical MR system. Larger
7T magnet compared to 3T and a requirement of 200-
400 metric tons of passive iron shielding to reduce
stray magnetic field2are no longer the limiting factors
in siting options for a 7T MRI system. The use of
active shielding has greatly improved the feasibility
to use these UhFMRI systems in clinical settings.5
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 77
Cost. The two factors that govern the cost of clin-
ical 7T MRI systems are (1) helium boil-off and (2)
Economy of scale. In newer 7T systems, the helium
boil-off problem has been addressed and these sys-
tems are called ”Zero helium boil-off” systems with
very little helium leak. Since the 7T systems are not
being used in common research and clinical practice,
these systems are not produced in large numbers. The
cost of these systems will definitely go down with
widespread adoption of these systems by institutions
and clinics. The cost may vary, currently (in mid-
2015) it costs about $10 million to install a 7T system
in the USA.5
FDA Approval. Although, federal drug agency
(FDA) has declared MRI scanners operating at 8T
and below as non-significant risk to adults and chil-
dren, but clinical use of 7T has not yet been approved
by the FDA.5
D. Patient Comfort and Safety
Risks associated with a 7T scanner are similar to
those for lower-field MRI systems. However, there
are some additional patient comfort and safety con-
siderations related to a 7T MRI system which are dis-
cussed in this section. One of the major patient safety
issues related to higher SAR related to 7T systems has
already been discussed in Sec. III. Continuous SAR
monitoring systems are installed on these systems to
ensure that the SAR limits set by the FDA they do not
exceed.5
Noise level and peripheral nerve stimulation.
Acoustic noise is produced by bulk vibrations in gra-
dient coils. These vibration are caused by Lorentz
force that scales directly with the magnetic field for
a given gradient coil orientation.1A 7T system gen-
erates acoustic noise that is about 7.4 dB higher than a
3T system for a given field and gradient orientation.2
In order to achieve a safe noise level below 99 dB,
modern systems are tested for this specified comfort
level.2,5
The peripheral nerve stimulation, which is related
to the switching speed of the gradients, is also moni-
tored and limited on all 7T systems.2,5
Ultra-high field related physiological effects.
Among the most commonly reported transitory phys-
iological effects caused by ultra-high magnetic field
brain imaging are dizziness and vertigo. These ef-
fects are caused by magnetohydrodynamic forces ex-
erted on the ionic fluids when a person moves through
the fringe field. In order to minimize these effects
the subjects are instructed to avoid quick head mo-
tion during the scan and also the scan table is moved
slowly to ensure patient speed reduction through the
fringe field.5In extracranial UhFMRI, specific ab-
sorption rate, especially when imaging deeply lying
structures, is the most important limiting factor that
needs careful monitoring.3
Metallic implants. To date, metallic implants of
almost any sort in the body are considered as a con-
traindication for scanning at 7T systems. So far only
two contrast injectors and one radiofrequency identi-
fication chip are approved for 7T.5However, studies
to test a wide range of implantable devices, to allow
a scanning of wider range of patients at 7T, are being
performed.3638
A summary of major challenges of a clinical 7T
MRI system is provided in Table 1.
TABLE 1: Major challenges of clinical 7T MRI system
Challenges Proposed solution
B0inhomogeneity More powerful referencing schemes, dynamic B0shimming, and smaller
voxel size5
B1inhomogeneity Custom designed adiabatic pulses,29 multichannel parallel transmit array28,33
Chemical shift
localization error
SAR Multichannel parallel transmit array28,33 ; lower flip angle sequences35
Metallic implants More studies to overcome contraindication are being performed3638
Volume 44, Issues 1-2, 2016
78 Karamat, Darvish-Molla, & Santos-Diaz
IV. 7T MRI IN NEUROIMAGING
Most of the research geared towards developing the
clinical applicability for 7T MRI systems has focused
on examining anatomical aspects of the brain due
to the very high resolution achievable at this field
strength in T1-weighted imaging, high CNR in fluid
attenuation inversion recovery (FLAIR) imaging, and
time-of-flight MR angiography (TOF-MRA), as well
as the ability of the system to detect microbleeds with
T2-weighted images due to the higher susceptibility
effect. On the functional side of MR applications, ad-
vantages offered by 7T rely on two main points, first
the increased SNR to measure the BOLD effect for
functional MRI (fMRI) making more accurate the lo-
calization of brain activity due to a specific task and,
second, the increased spectral separation in metabo-
lite peaks that improve the resolution and quantifica-
tion for magnetic resonance spectroscopy MRS. In
this section we describe major contributions of 7T
MRI in brain imaging with a view to its validation
as a clinical tool.
A. Anatomical Brain Imaging
Ultra-high field MR creates a complete new challenge
in terms of understanding and rediscovering the hu-
man brain. Both normal and disease conditions need
to be fully understood to address the real benefits on
7T MR in a clinical environment involving studies on
the brain. Because of this caveat, most brain imag-
ing at this field strength has found its way in study-
ing multiple sclerosis (MS), cerebrovascular diseases
(CVS), degenerative brain diseases, brain tumors, and
epilepsy.5,39
1. Multiple Sclerosis (MS)
MS is a demyelinating disease with prevalence
among relatively young patients (20s–30s) and its
pathogenesis is unknown. There are very few measur-
able biomarkers that can predict treatment response
and disease progression in patients with MS. The aim
of MRI to assess MS focuses on imaging white mat-
ter lesions, as they might be a key point in the eval-
uation of disease. In particular, cortical lesions are a
challenge because of the relatively low resolution at
clinical field strengths (1.5T and 3T) and some stud-
ies have shown its relevance in getting a better insight
into the pathogenesis.40 Moving to ultra-high field
MR could provide the resolution and CNR needed to
visualize these lesions with better accuracy. Graff et
al. showed improved identification of cortical lesions
caused by MS using 7T FLAIR imaging comparing it
with 3T.4Other high-resolution sequences like T2-,
double inversion recovery (DIR), and MPRAGE
imaging have also been used to examine MS cortical
lesions.40,4244 Among these, some lesions showed
characteristics not seen before at lower fields; for ex-
ample, the appearance of peripheral distortion rings
caused by susceptibility effect.40,42
Another characteristic that has been related with
the development of MS is the relationship between
white matter cortical lesions and veins; this has been
explored throughout T2-weighted images. Tallan-
tyre et al. found that 80% of lesions in patients with
MS were perivenous compared with 20% in normal
aging white matter lesions.45,46 This relationship was
clearer at 7T when compared to 3T, with a higher
percentage of lesions showing veins. Thus, such en-
hanced recognition may be helpful to definitely diag-
nose MS in uncertain cases; however, no studies have
yet showed superiority of 7T to diagnose MS, when
compared to 3T MRI.
2. Cerebrovascular Diseases
The assessment of cerebrovascular diseases using
MRI is mainly through high resolution MR angiog-
raphy (MRA) and T2-weighted sequences able to
show microbleeds. The optimization of MRA for 7T
has allowed the visualization of lenticulostriate arte-
rial branches,47 the small vessel that feed deep brain
structures like the basal ganglia. Thus, small vessels
disease like infarcts in such structures can be studied
now, increasing the interest for the field. Kang et al.48
found a reduction in the number and configuration of
lenticulostriate arteries in patients with chronic stroke
compared with controls. They also found reduced
visible arteries in hypertensive patients compared to
the control group.49 It should be noted that despite
the capabilities of high-resolution MRA at 7T, even
smaller vessels such as resistance arterioles are still
out of reach with inflow-based methods. Therefore,
improvement in these techniques is still needed to as-
sess small artery disease that results in white matter
lesions.
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 79
The accurate detection of aneurysms is another ca-
pability of high-resolution 7T MRA, where the clear
depiction of characteristics in the aneurysm, like the
presence of small perforating branches, can be made.
These branches could result in an infarct if not no-
ticed in treatment planning. Only a few studies have
compared the capabilities of 7T time-of-flight-MRA
versus clinical field strength in its capability to detect
aneurysms,50,51 showing a better-rated image qual-
ity at 7T. However, it is yet to be determined if 7T
MRA is better than 1.5T and 3T in detecting clin-
ically relevant aneurysms. In terms of follow-up of
treated aneurysms 7T is contraindicated for clips and
coils.
The study of microbleeds using MRI has gained
attention especially at high field strengths. Due to
the paramagnetic nature of microbleeds, it is pos-
sible to visualize them using T2-weighted images.
The increased sensitivity of higher fields to detect
microbleeds is because of the increased susceptibil-
ity effect and the higher resolution available. Fur-
thermore, due to the increased susceptibility at 7T,
microbleeds can be detected even using very short
echo times (TE = 2.5 ms).52 Using lower resolu-
tion (and lower field) the intensity difference be-
tween microbleeds can be lost in the noise. Conijn
et al. showed that 7T is better than 1.5T for detecting
microbleeds.16
The study of intracranial atherosclerosis is another
area that may benefit from the high SNR at 7T. In or-
der to get information regarding the nature of steno-
sis, it is necessary to visualize the intracranial vessel
wall. At clinical field strengths this wall was depicted
only in the presence of large atherosclerotic lesions or
vasculitis.5355 The increased resolution at 7T may
be exploited due to its capacity to depict the intracra-
nial vessel wall even in healthy subjects.56
3. Degenerative Brain Diseases
Dementia [vascular dementia and Alzheimer’s dis-
ease (AD)] and Parkinson’s disease are three major
pathologies where 7T MRI has found applications.
Theysohn et al. assessed the presence of white mat-
ter lesions using T2-weighted sequences at 7T and
found an increased sensitivity for finding these le-
sions when comparing with 1.5T in patients with vas-
cular dementia.57
One of the biggest objectives to be achieved study-
ing 7T in degenerative brain disease is the direct
detection of amyloid plaques. Sequences to acquire
T2-weighted images used at very high resolution
may show changes in tissue due to the presence of
amyloid, these changes have been elucidated during
post-mortem.58 However, it is not clear if such a goal
can be achieved at shorter scan times.
The evaluation of detailed substructures of the hip-
pocampus also plays a main role in the assessment of
dementia. In this regard, 7T MRI has shown an in-
creased depiction of substructures of the hippocam-
pus when compared with clinical field strengths,59,60
due to a high contrast between tissues. The differenti-
ation of these substructures may be important due to
its possible relation with memory loss. Currently, it
is not known which sequences are better to depict the
hippocampus substructures. In this regard, high res-
olution T2-, FLAIR, T1- and T2-weighted images
have shown promising results.5961
In regard Parkinson’s disease (PD), the use of 7T
MRI has focused on imaging the substantia nigra,
thought to play a meaningful role in PD’s pathogene-
sis. Cho et al. showed the superiority of 7T MRI de-
lineating substructures in the substantia nigra com-
pared with 1.5 and 3T.19 Changes in these substruc-
tures, like increased iron content62 and differences in
anatomical patterns,63 have also been found in pa-
tients with PD versus normal subjects.
4. Brain Tumors
The most important clinical marker to assess brain tu-
mors using MRI is the enhancement due to a MR-
gadolinium-based test. It allows differentiation and
a rough grading of tumors. Such an enhancement
is due to the disruption of the blood-brain barrier
causing contrast agent leakage. In this regard, 7T
MRI has not shown differences in the enhancement
area compared with lower field strengths.64 Other
methods at 7T like T2-weighted imaging may show
added value when examining the arterial microvas-
culature of the tumor. Tumor metabolism could also
probably be assessed using this technique, as the in-
creased metabolism of the tumor may lead to an
increase in deoxyhemoglobin signal in veins. Cur-
rently, only small samples of brain tumor patients
have been studied at 7T64 and there is no evidence
Volume 44, Issues 1-2, 2016
80 Karamat, Darvish-Molla, & Santos-Diaz
that shows an added value of 7T MRI compared with
3T MRI.
5. Epilepsy
The increased SNR and high resolution at 7T MRI
plus its novel contrast mechanisms can provide delin-
eation of epileptic foci not often visible at lower field
strengths, thus improving treatment planning and pa-
tient outcome. So far, 7T MRI has shown value when
characterizing features related with epilepsy like hip-
pocampal sclerosis,65 cortical dysplasia66 and vascu-
lar malformations.67
B. Functional Brain imaging
The increased SNR/CNR available at 7T MRI needs
to be exploited on every front in order to become a
clinical tool that can be used on a daily basis. Func-
tional magnetic resonance imaging (fMRI) is not the
exception of it as this technique has gained power for
diagnostic and treatment planning purposes in lower
field strengths. The improved sensitivity in BOLD
signal at 7T seems promising and its benefits have
already been demonstrated when comparing versus
1.5T and 3T for healthy subjects.68 Furthermore, a
study from Beisteiner et al. has shown evidence of
the benefit at 7T over 3T in a patient population.69
However, issues related with high field systems like
increased susceptibility artifacts, B1inhomogeneity,
SAR limitations, and patient motion (especially in pa-
tients with brain pathology) still remain as a challenge
and clear advantages are yet to be determined for the
jump of field strength in this regard. In addition to
acquisition methodology, post processing techniques
need to be studied in greater detail. Yang et al. showed
statistical distribution differences in the data when
comparing 7T and 3T.70
Magnetic resonance spectroscopy (MRS) is an-
other technique useful in the assessment of brain
function due to its capability to measure the concen-
tration of different metabolites in the tissue. The in-
creased magnetic field at 7T results in a separation
of metabolite peaks leading to a higher resolution
and better quantification. In applications related to the
brain, MRS imaging at 7T has been shown to pro-
vide a better measuring of metabolic markers in tu-
mors when compared with 3T.71 There is an increase
of 35% in SNR of 7T over 3T leading to a clear su-
periority of the higher field in measuring glutamate,
glutamine, and GABA.72 Thus an understanding of
metabolic processes in healthy and disease conditions
can be improved at higher field strength.
V. 7T MRI BEYOND THE BRAIN
A. Musculoskeletal Imaging
Despite the issues discussed earlier in Sec. III, such
as homogeneous radiofrequency (RF) coil design, in-
creased chemical shift artifacts, susceptibility arti-
facts, RF energy deposition, and changes in relax-
ation times, MRI at 7T likely will provide some ex-
cellent opportunities for high-resolution morphologic
imaging and functional imaging of musculoskele-
tal systems.7381 Regatte and Schweitzer in 200776
studied the feasibility of acquiring high-resolution
in vivo images of the musculoskeletal system in 15
healthy human volunteers on the knee, ankle, and sev-
eral muscle groups at 7T using high-resolution ax-
ial/sagittal morphological imaging [3D fast low-angle
shot (FLASH)]. They reported 70%–80% higher car-
tilage SNR at 7T compared to corresponding regions
at 3T. Generally, there is not any signal intensity from
menisci on clinical scanners (1.5T and 3T) at the
usual imaging TE values, due to low SNR and res-
olution. However at 7T, the contrast and high SNR
can enable better detection of subtle morphological
changes by improving image resolution. It is con-
cluded that MRI at 7T provides a powerful, rapid,
high-resolution functional imaging tool of muscu-
loskeletal systems (1H, 23Na, and 31 P).
In 2009 the same group77 demonstrated, for the
first time, the feasibility of acquiring high-resolution,
isotropic 3D-sodium knee images of healthy and os-
teoarthritis patients at 7T in less than 15 minutes.
Their preliminary results suggest that for osteoarthri-
tis imaging, lesion localization, and clinical diagno-
sis, sodium imaging at 7T may be a better alterna-
tive. Sodium MRI has been shown to correlate with
glycosaminoglycan (GAG) concentration in many
studies.8285 Compared to proton MRI, the sodium
MRI has a low SNR, poor spatial resolution, and long
acquisition times due to the low natural abundance of
sodium, and rapid biexponential T2signal decay.77
Hence, in vivo sodium and phosphorous imaging at
clinical magnetic field strengths poses major chal-
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 81
lenges and is rather limited. However ultra-high field
systems with improved gradient hardware and pulse
sequences have potential for improving the spatial-
temporal resolution of sodium MRI of the knee in
vivo, in clinically acceptable scan times.77
Jordan et al.80 carried out a study to compare the
relaxation times of musculoskeletal tissues at 3T and
7T in order to use these measurements to select ap-
propriate parameters for musculoskeletal protocols at
7T. T1and T2relaxation times of cartilage, mus-
cle, synovial fluid, bone marrow, and subcutaneous
fat were measured at both 3T and 7T, in the knees
of five healthy volunteers. It was shown that at 7T,
the T1relaxation times increased by an average of
51%, whereas the T2relaxation times decreased by
an average of 21%. Further parameter optimization
for clinical musculoskeletal imaging protocols at 7T
will be viable with the knowledge of these relaxation
parameters.
Recently, it was shown that there is a substantial
benefit from UhFMRI of ankles with routine clinical
sequences at 7T compared to 3T.78 Three clinical se-
quences: (1) 3D gradient-echo, T1-weighted; (2) 2D
fast spin-echo, PD-weighted; and (3) 2D spin-echo,
T1-weighted were used on both systems to examine
10 healthy volunteers. SNR was calculated for car-
tilage, bone, muscle, synovial fluid, Achilles tendon,
and Kager’s fat-pad. It was shown that the mean SNR
significantly increased by 60.9% and 86.7% at 7T
compared to 3T for 3D GRE, and 2D TSE, respec-
tively. However, an average SNR decrease by 25%
in the 2D SE sequence was also observed. The CNR
was obtained for cartilage/bone, cartilage/fluid, carti-
lage/muscle, and muscle/fat-pad, and showed an in-
crease for 2D TSE images, and in most 3D GRE im-
ages.
The first clinically comprehensive hip MRI pro-
tocol at 7T was developed by Chang et al. (2014),
to demonstrate the feasibility of performing bone
microarchitecture, high-resolution cartilage.81 AT1-
weighted 3D fast low-angle shot (3D FLASH) se-
quence (0.23 ×0.23 ×1–1.5 mm3) was used for
bone microarchitecture imaging, and T1-weighted
3D FLASH and volumetric interpolated breath-hold
examination (VIBE) sequences (0.23 ×0.23 ×
1.5 mm3) with saturation or inversion recovery-
based fat suppression for cartilage imaging and
2D intermediate-weighted fast spin-echo (FSE) se-
quences without and with fat saturation (0.27 ×0.27
×2 mm3) for clinical imaging. It was found that us-
ing 7T MRI (1) depicting individual trabeculae within
the proximal femur in vivo; (2) performing high-
resolution cartilage MRI of the hip joint with T1-
weighted 3D gradient echo sequences, and (3) per-
forming fast spin-echo imaging both without and with
fat suppression would be possible. As a future per-
spective, unique insight into the pathogenesis of dif-
ferent hip disorders or the ability to detect disease
changes in the hip joint in its earliest stages will be
achievable using an amalgamation of high-resolution
morphologic proton imaging methods and physio-
logic imaging methods at 7T.81
B. Breast
Currently imaging of breast using MRI is mainly per-
formed by 1.5T and 3T MRI systems8688 in which
the diagnostic performance yields a sensitivity of 0.90
and a specificity of 0.72.89 Recently, ultra-high field
7T MRI has become available in a clinical research
setting. At 7T, substantial improvements in image
quality can be provided and there is a potential to
improve diagnosis and staging of breast cancer pa-
tients if technical challenges can be overcome with
7T breast MRI.9093
Stehouwer et al.91 carried out the first clinical ex-
perience with contrast-enhanced MRI (CE-MRI) of
breast cancer at 7T, compared to 3T and histopathol-
ogy. T1weighted CE-MRI with unilateral breast
coil results obtained at 7T and 3T showed en-
hanced SNR as well as the enhanced contrast be-
tween the tumor and glandular tissue at 7T. A sin-
gle dose of gadolinium-based contrast agent was used
in both cases. Contrast enhancement-to-noise ratio
was found to be 4.6 at 7T and 2.8 at 3T when
comparing contrast to noise of the mass before and
after contrast administration. It was concluded that
clinical contrast-enhanced MRI of the breast at 7T
is technically feasible. Ultra-high spatial and tem-
poral resolution contrast-enhanced breast MR imag-
ing was reported using parallel imaging at 7T.94,95
High potential of breast MRI at 7T with sensitiv-
ity of 100% and a specificity of 92% were reported
by Pinker et al. in 2014.96 Images without signif-
icant artifacts and satisfactory fat suppression were
obtained using an isotropic spatial resolution of 0.7
Volume 44, Issues 1-2, 2016
82 Karamat, Darvish-Molla, & Santos-Diaz
×0.7 ×0.7 mm3with a temporal resolution of
14 s.
Diffusion-weighted imaging (DWI) has a high po-
tential for characterizing breast tumors and monitor-
ing or predicting treatment response.97 Compared to
dynamic contrast-enhanced MRI (DCE-MRI), DWI
has lower spatial resolution. If the spatial resolu-
tion of 1 ×1×2.5 mm3, which is the recom-
mended resolution for CE-MRI,98 could be achiev-
able for DWI sequences as well, both the detection
of smaller lesions and the morphological evaluation
of the breast may be possible. Hence, there is a need
of measure at higher magnetic field strength, imag-
ing technique and hardware improvement.92 Just re-
cently, Pinker et al.93 carried out a feasibility study of
multiparametric MR imaging of the breast in com-
bination with DCE imaging and DWI at 7T and
to investigate any improvement in diagnostic accu-
racy. Twenty-five patients with BI-RADS category
4 and 15 patients with BI-RADS category 5, to-
tal of 40 patients, age 18 years or older, not preg-
nant and not breastfeeding underwent bilateral mul-
tiparametric MR imaging of the breast at 7T and a
dedicated four-channel double-tuned 31P/1H breast
coil. They concluded the feasibility of multiparamet-
ric MR imaging of the breast with DCE MR imag-
ing and DWI at 7T in clinical practice and diag-
nosis of breast cancer with a high diagnostic accu-
racy of 94.1% (44 of 46 lesions) and excellent inter-
rater agreement (k = 0.833 to 1) would be achiev-
able.
C. Prostate
MRI has been widely used for a variety of clinical
purposes in the prostate, including tumor detection
and localization, treatment planning, and assessment
of aggressiveness.99 There is a need of biopsy in the
case of suspected aggressive prostate cancer, in which
small malignancies could be easily missed.3To iden-
tify tumor tissue magnetic resonance spectroscopy is
a better option to image metabolites such as spermine,
choline, creatine, and citrate.100 This is where ultra-
high field strength has advantages of increased sep-
aration of spectral peaks compared to lower strength
fields.101 However, as it was mentioned imaging the
human body at 7T is very challenging. There have
been studies dedicated to the investigating and over-
coming these challenges in prostate imaging by de-
signing an external transmit-receive coil array,102 im-
proving SNR and B1transmit field for an endorec-
tal coil103,104 and assessing image quality and cancer
visibility of prostate MRI at 7T.105
Rosenkrantz et al. in 2013102 developed a more
simply designed system for 7T prostate MRI, with
two transmit-receive elements and six receive-only
elements, avoiding parallel transmission and RF
shimming, for the goal of optimization of coil ar-
rangement for high-resolution T2-weighted prostate
MRI at 7T and substantial improvements in SNR
compared with 3T MRI. It was shown that prostate
tumors diagnosed or confirmed with MR guided
biopsy or prostatectomy were well visualized with 7T
MRI.106
D. Heart
Despite the major challenges in ultra-high field
MRI, a number of research groups already have
demonstrated the feasibility of cardiac imaging at
7T and have compared it to 3T.107112 Due to a
higher SNR in a higher strength field, 7T MRI is
practically advantageous in the areas that are crit-
ical to diagnosis of cardiac disease, such as coro-
nary artery angiography.113,114 Also cardiac MR
at 7T offers the potential for imaging of cardiac
physiology such as perfusion, coronary lumen and
wall, tissue oxygenation, and cardiac metabolism
which are substandard at 1.5T or even at 3T at
present.112
Temporally-resolved and high spatial resolution
myocardial T2mapping is another application of
ultra-high field cardiac MRI which was recently re-
ported by Hezel et al. to assess myocardial iron depo-
sition or myocardial perfusion deficits to character-
ize myocardial tissue.115 In another study,116 a direct
quantitative and qualitative comparison between 7T
and 3T for right coronary MR angiography in young
healthy volunteers was undertaken. It was shown that
the SNR of the blood pool measured on the 7T im-
ages was 60% higher compared to 3T. Also compared
to 3T, improved CNR between the blood pool and
the epicardial fat, and increased vessel sharpness at
7T were observed. Suttie et al.112 carried out a com-
parison of cardiovascular magnetic resonance imag-
ing (CMR) at 1.5T, 3T, and 7T field strengths using
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 83
steady state free precession (SSFP) and fast low-angle
shot (FLASH) cine sequences to assess cardiac func-
tion. No significant differences in cardiac volume or
mass were found using SSFP at 3T or 7T when com-
paring with the clinical gold standard of 1.5T SSFP
imaging. There were also no significant differences
between cardiac volume and mass determined using
FLASH imaging at 1.5T, 3T, and 7T field strengths.
A detailed review on human cardiac magnetic reso-
nance at ultrahigh fields was published by Niendorf et
al. in 2013.117 The authors addressed many channel
radiofrequency (RF) technology concepts, and basic
principles of mapping and shimming of transmission
fields including RF power deposition considerations
for the ultra-high field. Clinical application of car-
diac MR at 7T, including assessment of cardiac func-
tion, myocardial tissue characterization, and MR an-
giography of large and small vessels as well as het-
eronuclear MR of the heart and the skin were dis-
cussed.
E. Angiography
MR angiography at ultra-high field has been often
used for cerebrovascular imaging and has yielded
measurable benefits in assessing cerebrovascular
diseases as described before.118120 Whole body
MR angiography images have also been recently
recorded. As the SNR decreases due to accelera-
tion techniques such as parallel acceleration121 and
compressed sensing,122,123 the increase in SNR us-
ing ultra-high strength MR is favorable for MR an-
giography in which high spatiotemporal resolution
is essential.124 The feasibility of aortic 4D flow at
7.0T for both contrast enhanced and non-contrast en-
hanced was investigated by Hess et al. in 2015.125
They compared and quantified SNR as a function of
field strength and contrast enhancement for 1.5T, 3T,
and 7T. The non-enhanced acquisition at 7T SNR was
found to be 2.2 times that of 3T. It is interesting to
note that the SNR of 3T MRI is 1.7 times that of
1.5T.
Metzger et al.126 investigated the feasibility of
non-contrast enhanced renal MR angiography at 7T
using a respiratory-gated turbo-FLASH sequence
consisting of a slab-selective inversion and chemi-
cal shift selective fat suppression followed by signal
readout. Vessel conspicuity, which would be bene-
ficial for several clinical applications including pre-
operative evaluation of renal vascular anatomy, un-
controlled hypertension, and the evaluation of pa-
tients with renal insufficiency or failure, is the motiva-
tion behind performing these studies at 7T. They ac-
quired high quality images of the renal arteries with-
out venous and background signal artifacts by using
more efficient B1shimming solutions for the inver-
sion preparation and more homogeneous solutions for
the excitation. Likewise, in another study by Umutlu
et al.,127 the feasibility of 7T non-enhanced high-
field MR imaging of the renal vasculature was stud-
ied using non-enhanced T1-weighted FLASH imag-
ing. It was concluded that both methods are promis-
ing techniques for good quality non-enhanced renal
artery assessment at 7T. Contrast-enhanced MR an-
giography is currently a valuable tool in the diagno-
sis of patients with peripheral arterial disease. How-
ever, there are patients who are at risk of nephro-
genic systemic fibrosis due to the gadolinium contrast
agent.128 Hence, feasibility of non-enhanced MR an-
giography technique at 7T might be an important
development.129131
VI. CONCLUSIONS
SNR increases linearly with field strength, B0. There-
fore, achieving a higher B0is a motivation to im-
prove capabilities of the MRI. Such a scanner im-
proves MRI capabilities as (1) the SNR starved tech-
niques, like fMRI and DTI, benefited greatly with
higher field strength; (2) faster imaging sequences,
such as SENSE, SMASH, and GRAPPA can also
get a sensitivity boost due to the increase in SNR at
higher field strength; and (3) higher spectral disper-
sion and sensitivity achievable at higher field strength
greatly improve MRS outcomes.
In order to take advantage of the research poten-
tial offered by 7T MRI systems, a significant amount
of progress has been made in the development of
software and hardware solutions to address major is-
sues such as B0and B1nonuniformities, reduction of
SAR, and susceptibility related artifacts.
UhFMR imaging has shown significant potential
in revealing the anatomical abnormalities that would
remain unresolved at lower field strengths. A large
number of studies in brain imaging have revealed
the ability of higher field strength to visualize fine
Volume 44, Issues 1-2, 2016
84 Karamat, Darvish-Molla, & Santos-Diaz
structural abnormalities associated with different dis-
eases; depiction of different metabolites and their
distribution present in smaller structures; detection
of microbleeds, microvasculature, and blood prod-
ucts; better spectral dispersion and sensitivity for
the signals related to underlying neuronal activity
and tapping into the signal for nuclei other than
1H.
Although in extracranial imaging applications,
UhFMRI has shown promise in terms of improved
SNR and resolution, it is still a challenge to de-
velop more robust and safe protocols to acquire uni-
form images, especially for deep seated structures.
Another important question that needs attention is,
“Does higher SNR and spatial resolution offered by
UhFMRI improves diagnosis of disease compared to
MRI machines with lower fields, in extracranial ap-
plications?” Introduction of dedicated coils for organs
like breast and knee has greatly improved the situa-
tion in terms of safety and robustness for these or-
gans.
Based on the studies performed in neuroimaging,
it can be concluded that the combination of high-
resolution anatomic, spectroscopic, and functional
imaging at 7T has the ability to become a strong, non-
invasive tool for diagnosis and treatment of different
neurological diseases and disorders. However, more
technological developments and several clinical stud-
ies are required to show diagnostic improvements, if
any, provided by a 7T MRI system in extracranial
imaging applications.
In short, continued technical developments to
achieve better B0and B1homogeneity and lower
SAR, reduction in number of MRI contraindications
related to implantable devices at 7T, more clinical
studies to prove added diagnostic value with 7T, and
making the cost of a 7T system comparable to current
clinical MR imaging systems are the key factors that
would lead the way for ushering the 7T scanner from
research laboratories to the clinic.
ACKNOWLEDGMENT
The authors would like to thank Dr. Mike Nosewor-
thy of Department of Biomedical Engineering, Mc-
Master University, for all his help, guidance and for
the precious time he spent in reviewing this arti-
cle.
REFERENCES
1. Brown RW, Cheng YN, Haacke EM, Thompson MR,
Venkatesan R. Magnetic resonance im aging: physical prin-
ciples and sequence design. J Wiley & Sons Inc. 2014, New
Jersey, 2 ed.
2. Schmitt F, Potthast A, Stoeckel B, Triantafyllou C, Wig-
gins C. J, Wiggins G, Wald LL. Aspects of clinical imag-
ing at 7T. In: Robitaille PM, Berliner L (Eds.). Ultra High
Field Magnetic Resonance Imaging, vol. 26, pp. 59–104.
Springer Science, USA, 2006.
3. Zwanenburg JJM, van der Kolk AG, Luijten PR. Ultra-high-
field MR imaging: research tool or clinical need? PET Clin.
2013, 8:311–328.
4. Nakada T. Clinical application of high and ultra-high field
MRI. Brain Dev. 2007, 29:325–335.
5. Balchandani P, Naidich TP. Ultra-high-field MR neu-
roimaging. Am J Neuroradiol. 2015, Jul; 36(7):1204–1215.
6. Duyn JH. NeuroImage The future of ultra-high field MRI
and fMRI for study of the human brain. NeuroImage. 2012,
62(2):1241–1248.
7. Gr¨
ohn HI, Michaeli S, Garwood M, Kauppinen RA, Gr¨
ohn
OHJ. Quantitative T(1ρ) and adiabatic CarrPurcell T2 mag-
netic resonance imaging of human occipital lobe at 4T.
Magn Reson Med. 2005, 54(1):14–19.
8. Peters AM, Brookes MJ, Hoogenraad FG, Gowland PA,
Francis ST, Morris PG, Bowtell R. T24measurements in
human brain at 1.5, 3 and 7T. Magn Reson Imaging. 2007,
25(6):748–753.
9. Uludag K, Muller-Bierl B, Ugurbil K. An integrative model
for neuronal activity-induced signal changes for gradi-
ent and spin echo functional imaging. NeuroImage. 2009,
48:150–162.
10. Golay X, Petersen ET. Arterial spin labeling: benefits and
pitfalls of high magnetic field. Neuroimaging Clin N Am.
2006, 16(2):259–268.
11. Ibrahim el-SH. Myocardial tagging by cardiovascular mag-
netic resonance: evolution of techniques, pulse sequences,
analysis algorithms, and applications. J Cardiovasc Magn
Reson. 2011, 13:36.
12. Hendrikse J, Zwanenburg JJ, Visser F, TakaharaT, Luijten P.
Noninvasive depiction of the lenticulostriate arteries with
time-of-flight MR angiography at 7.0T. Cerebrovasc Dis.
2008, 26(6):624–629.
13. Cho Z, Kang C, Han J, Kim S, Kim K. Observation of the
lenticulostriate arteries in the human brain in vivo using
7.0T MR angiography. Stroke. 2008, 39(5):1604–1606.
14. Johst S, Wrede KH, Ladd ME, Maderwald S. Timeof-flight
magnetic resonance angiography at 7T using venous satu-
ration pulses with reduced flip angles. Invest Radiol. 2012,
47(8):445–450.
15. Brundel M, Heringa SM, de Bresser J, Koek HL, Zwanen-
burg JJM, Jaap Kappelle L, Luijten PR, Biessels GJ. High
prevalence of cerebral microbleeds at 7T MRI in patients
with early Alzheimer’s disease. J Alzheimers, 2012 Jan;
31(2):259–263.
16. Conijn MMA, Geerlings MI, Biessels GJ, Takahara T,
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 85
Witkamp TD, Zwanenburg JJM, Luijten PR, Hendrikse
J. Cerebral microbleeds on MR imaging: Comparison be-
tween 1.5 and 7T. Am J Neuroradiol. 2011, 32(6):1043–
1049.
17. Tallantyre EC, Brookes MJ, Dixon JE, Morgan PS, Evan-
gelou N, Morris PG. Demonstrating the perivascular dis-
tribution of MS lesions in vivo with 7T MRI. Neurology.
2008, 70(22):2076–2078.
18. Ge Y, Zohrabian VM, Grossman RI. Seven-Tesla magnetic
resonance imaging: new vision of microvascular abnormal-
ities in multiple sclerosis. Arch Neuro. 2008, 65(6):812–
816.
19. Cho Z-H, Min H, Oh S, Han J-Y, Park C-W, Chi J-G, Kim
Y-B, Paek SH, Lozano AM, Lee KH. Direct visualization of
deep brain stimulation targets in Parkinson disease with the
use of 7T magnetic resonance imaging. J Neurosurg. 2010,
113(3):639–647.
20. Lenglet C, Abosch A, Yacoub E, de Martino F, Sapiro G,
Harel N. Comprehensive in vivo mapping of the human
basal ganglia and thalamic connectome in individuals using
7T MRI. PLoS ONE. 2012, 7(1):e29153.
21. Sanchez-panchuelo RM, Besle J, Beckett A, Bowtell R,
Schluppeck D, Francis S. Within-digit functional parcella-
tion of brodmann areas of the human primary somatosen-
sory cortex using functional magnetic resonance imaging at
7T. J Neurosci. 2012, 32(45):15815–15822.
22. Duong TQ, Yacoub E, Adriany G, Hu XU, Gurbil K, Kim
SG. Microvascular BOLD contribution at 4 and 7T in
the human brain: Gradient-echo and spin-echo fMRI with
suppression of blood effects. Magn Reson Med. 2003,
49(6):1019–1027.
23. Donahue MJ, Hoogduin H, Zijl PCMV, Jezzard P, Lui-
jten PR, Hendrikse J. Blood oxygenation leveldependent
(BOLD) total and extravascular signal changes and R2
in human visual cortex at 1.5, 3.0 and 7.0T. NMR Biomed.
2011 Jan; 24:25–34.
24. Ivanov D, Sch¨
afer A, Streicher MN, Heidemann RM, Tram-
pel R, Turner R. A simple low-SAR technique for chemical-
shift selection with high-field spin-echo imaging. Magn Re-
son Med. 2010, 64(2):319–326.
25. Haacke EM, Mittal S, Wu Z, Neelavalli J, Cheng Y-CN. Sus-
ceptibility weighted imaging: technical aspects and clinical
applications, part 1. Am J Neuroradiol. 2009, 30(1):19–30.
26. Schweser F, Deistung A, Lehr BW, Reichenbach JR. Dif-
ferentiation between diamagnetic and paramagnetic cere-
bral lesions based on magnetic susceptibility mapping. Med
Phys. 2010, 37(10):5165–5178.
27. Duyn JH, van Gelderen P, Li T-Q, de Zwart JA, Koretsky
AP, Fukunaga M. High-field MRI of brain cortical substruc-
ture based on signal phase. Proc Nat Acad Sci USA. 2007,
104(28):11796–11801.
28. Ladd ME. High-field-strength magnetic resonance: potential
and limits. Top Magn Reson Imag; 2007, 18(2):139–152.
29. Balchandani P, Glover G, Pauly J, Spielman D. Improved
slice-selective adiabatic excitation. Magn Reson Med.
2014, 71(1):75–82.
30. Balchandani P, Pauly J, Spielman D. Designing adiabatic ra-
dio frequency pulses using the Shinnar-Le Roux algorithm.
Magn Reson Med. 2010, 64(3):843–851.
31. Saekho S, Yip CY, Noll DC, Boada FE, Stenger VA. Fast-kz
three-dimensional tailored radiofrequency pulse for reduced
B1 inhomogeneity. Magn Reson Med. 2006, 55(4):719–
724.
32. Cloos MA, Boulant N, Luong M, Ferrand G, Giacomini E,
Le Bihan D, Amadon A. K T-points: Short threedimensional
tailored RF pulses for flip-angle homogenization over an
extended volume. Magn Reson Med. 2012, 67(1):72–80.
33. Zhu Y. Parallel excitation with an array of transmit coils.
Magn Reson Med. 2004, 51:775–784.
34. Heidemann RM, Porter DA, Anwander A, Feiweier T,
Heberlein K, Kn¨
osche TR, Turner R. Diffusion imaging in
humans at 7T using readout-segmented EPI and GRAPPA.
Magn Reson Med. 2010, 64(1):9–14.
35. Saranathan M, Tourdias T, Kerr AB, Bernstein J, Kerchner
G, Han M, Rutt B. Optimization of magnetizationprepared
3-dimensional fluid attenuated inversion recovery imaging
for lesion detection at 7T. Invest Radiol. 2014, 49(5):290–
298.
36. Vogt J, Renz W, Celal O, Santoro D, Winter L, Mu A, Nien-
dorf T, Gra A. Detailing radio frequency heating induced
by coronary stents: a 7.0T magnetic resonance study. PLoS
One. 2012, 7(11):e49963.
37. Wezel J, Brink WJ, Kooij B, Webb A. Simulation study on
safety of dental implants at 7T. In Proceedings of ISMRM
20th Scientific Meeting and Exhibition; Melbourne Aus-
tralia. 2012, p. 2766.
38. Ansems J, van der Kolk AG, Kroeze H, Van Den Berg CA,
de Borst GJ, Luijten PR. MR imaging of patients with stents
is safe at 7.0T. In Proceedings of ISMRM 20th Scientific
Meeting and Exhibition; Melbourne, Australia. 2012, p.
2766.
39. van der Kolk AG, Hendrikse J, Zwanenburg JJM, Visser F,
Luijten PR. Clinical applications of 7T MRI in the brain.
Eur J Radiol. 2013, 82:708–718.
40. Rudick RA, Trapp BD. Gray-matter injury in multiple scle-
rosis. N Engl J Med. 2009, 361(15):1505–1506.
41. De Graaf WL, Castelijns JA, Wattjes MP, Geurts J, Pouwels
P, Polman CH, Barkhof F, Luijten PR. 7T 3D-FLAIR and
3D-DIR: high sensitivity in cortical regions in multiple scle-
rosis Proc. Intl. Soc. Mag. Reson. Med. 2010, 18:20–67.
42. Kollia K, Maderwald S, Putzki N, Schlamann M, Theysohn
JM, Kraff O. First clinical study on ultra-highfield MR
imaging in patients with multiple sclerosis: comparison of
1.5T and 7T. Am J Neuroradiol. 2009, 30(4):699–702.
43. Mainero C, Benner T, Radding A, van der Kouwe A, Jensen
R, Rosen BR, Kinkel RP. In vivo imaging of cortical pathol-
ogy in multiple sclerosis using ultra-high field MRI. Neu-
rology. 2009, 73(12):941–948.
44. Tallantyre EC, Morgan PS, Dixon JE, Al-Radaideh A,
Brookes MJ, Morris PG, Evangelou N. 3T and 7T MRI of
multiple sclerosis cortical lesions. J Magn Reson Imaging :
JMRI. 2010, 32(4):971–977.
45. Tallantyre EC, Morgan PS, Dixon JE, Al-Radaideh A,
Brookes MJ, Evangelou N, Morris PG. A comparison of 3T
Volume 44, Issues 1-2, 2016
86 Karamat, Darvish-Molla, & Santos-Diaz
and 7T in the detection of small parenchymal veins within
MS lesions. Invest Radiol. 2009, 44(9):491–494.
46. Tallantyre EC, Morgan PS, Dixon JE, Al-Radaideh A,
Brookes MJ, Morris PG, Evangelou N. 3T and 7T MRI of
multiple sclerosis cortical lesions. J Magn Reson Imaging.
2010, 32(4):971–977.
47. Kang CK, Park CW, Han JY, Kim SH, Park CA, Kim KN,
Hong SM, Kim YB, Lee KH, Cho ZH. Imaging and analysis
of lenticulostriate arteries using 7.0T magnetic resonance
angiography. Magn Reson Med. 2009, 61(1):136–144.
48. Kang CK, Park CK, Park CW, Lee YB, Cho ZH, Kim YB.
Lenticulostriate arteries in chronic stroke patients visualised
by 7T magnetic resonance angiography. Int J Stroke. 2010,
5(5):374–380.
49. Kang CK, Park CA, Lee H, Kim SH, Park CW, Kim YB,
Cho ZH. Hypertension correlates with lenticulostriate arter-
ies visualized by 7T magnetic resonance angiography. Hy-
pertension. 2009, 54(5):1050–1056.
50. M¨
onninghoff C, Maderwald S, Theysohn JM, Kraff O, Ladd
SC, Ladd ME, Forsting M, Quick HH, Wanke I. Evalua-
tion of intracranial aneurysms with 7T versus 1.5T time-
of-flight MR angiography – Initial experience. R¨
oFo. 2009,
181(1):16–23.
51. Monninghoff C, Maderwald S, Theysohn J, Kraff O, Ladd
M, Regel J, M¨
oller-Hartmann C, Forsting M, Wanke I. Eval-
uation of intracranial aneurysms with 7T versus 1.5T MR
angiography and digital subtraction angiography. R¨
oFo.
2008, 180(02):16–23.
52. Conijn MMA, Geerlings MI, Luijten PR, Zwanenburg JJM,
Visser F, Biessels GJ, Hendrikse J. Visualization of cerebral
microbleeds with dual-echo T2-weighted magnetic reso-
nance imaging at 7.0T. Methods. 2010, 59(1):52–59.
53. Swartz RH, Bhuta SS, Farb RI, Agid R, Willinsky
RA, Butany J, Wasserman BA, Johnstone DM, Silver
FL, Mikulis DJ. Intracranial arterial wall imaging using
high-resolution 3-tesla contrast-enhanced MRI. Neurology.
2009, 72(7):627–634.
54. Ryu CW, Jahng GH, Kim EJ, Choi WS, Yang DM. High res-
olution wall and lumen MRI of the middle cerebral arteries
at 3T. Cerebrovasc Dis. 2009, 27(5):433–442.
55. Xu W-H, Li M-L, Gao S, Ni J, Zhou L-X, Yao M, Peng
B, Feng F, Jin Z-Y, Cui L-Y. In vivo high-resolution MR
imaging of symptomatic and asymptomatic middle cere-
bral artery atherosclerotic stenosis. Atherosclerosis. Octo-
ber 2010, 212(2):507–511.
56. van der Kolk AG, Zwanenburg JJM, Brundel M, Biessels G-
J, Visser F, Luijten PR, Hendrikse J. Intracranial vessel wall
imaging at 7.0T MRI. Stroke. 2011 Sept; 42(9):2478–2484.
57. Theysohn JM, Kraff O, Maderwald S, Barth M, Ladd SC,
Forsting M, Ladd ME, Gizewski ER. 7T MRI of microb-
leeds and white matter lesions as seen in vascular dementia.
J Magn Reson Imaging. 2011, 33(4):782–791.
58. van Rooden S, Maat-Schieman MLC, Nabuurs RJA, van
der Weerd L, van Duijn S, van Duinen SG, Natt´
e R, van
Buchem MA, van der Grond J. Cerebral amyloidosis: post-
mortem detection with human 7.0T MR imaging system.
Radiology. 2009, 253(3):788–796.
59. Theysohn JM, Kraff O, Maderwald S, Schlamann MU, de
Greiff A, Forsting M, Ladd SC, Ladd ME, Gizewski ER.
The human hippocampus at 7T in vivo MRI. Hippocampus.
2009, 19(1):1–7.
60. Kerchner GA, Hess CP, Hammond-Rosenbluth KE, Xu D,
Rabinovici GD, Kelley DA. C, Vigneron DB, Nelson SJ,
Miller BL. Hippocampal CA1 apical neuropil atrophy in
mild Alzheimer disease visualized with 7T MRI. Neurol-
ogy. 2010, 75(15):1381–1387.
61. Thomas BP, Welch EB, Niederhauser BD, Whetsell WO,
Anderson AW, Gore JC, Avison MJ, Creasy JL. High-
resolution 7T MRI of the human hippocampus in vivo. J
Magn Reson Imaging. 2008, 28(5):1266–1272.
62. Bajaj N, Schafer A, Wharton S, Gontu V, Bowtell R, Auer
DP, Gowland P, Schwartz S, Lotfipour A. PATH53 magnetic
susceptibility of substantia nigra in Parkinson’s disease: a
7T in vivo MRI study. J Neurol Neurosurg Psychi. 2010,
81(11):e22.
63. Cho Z-H, Oh S-H, Kim J-M, Park S-Y, Kwon D-H, Jeong
H-J, Kim Y-B, Chi J-G, Park C-W, Huston J, Lee KH, Jeon
BS. Direct visualization of Parkinsons disease by in vivo
human brain imaging using 7.0T magnetic resonance imag-
ing. Mov Disorders. 2011, 26(4):713–718.
64. Moenninghoff C, Maderwald S, Theysohn JM, Kraff O,
Ladd ME, El Hindy N, Van De Nes J, Forsting M, Wanke
I. Imaging of adult astrocytic brain tumours with 7T MRI:
Preliminary results. Eur Radiol. 2010, 20(3):704–713.
65. Breyer T, Wanke I, Maderwald S, Woermann FG, Kraff O,
Theysohn JM, Ebner A, Forsting M, Ladd ME, Schlamann
M. Imaging of patients with hippocampal sclerosis at 7T.
Acad Radiol. 2010, 17(4):421–426.
66. Madan N, Grant PE. New directions in clinical imaging of
cortical dysplasias. Epilepsia. 2009, 50:9–18.
67. Schlamann M, Maderwald S, Becker W, Kraff O, Theysohn
JM, Mueller O, Sure U, Wanke I, Ladd ME, Forsting
M, Schaefer L, Gizewski ER. Cerebral cavernous heman-
giomas at 7T. Acad Radiol. 2010, 17(1):3–6.
68. Zwaag WVD, Francis S, Head K, Peters A, Gowland P,
Morris P, Bowtell R. fMRI at 1.5 , 3 and 7T: characteris-
ing BOLD signal changes. NeuroImage. 2009, 47(4):1425–
1434.
69. Beisteiner R, Robinson S, Wurnig M, Hilbert M, Merksa K,
Rath J, H¨
ollinger I, Klinger N, Marosi C, Trattnig S, Geißler
A. Clinical fMRI: Evidence for a 7T benefit over 3T. Neu-
roImage. 2011, 57(3):1015–1021.
70. Yang X, Holmes MJ, Newton AT, Morgan VL, Landman BA.
A comparison of distributional considerations with statisti-
cal analysis of resting state fMRI at 3T and 7T. Pro Soc
Photo Opt Instrum Eng. 2012, 23(8314):1–10.
71. Lupo JM, Li Y, Hess CP, Nelson SJ. Advances in ultrahigh
field MRI for the clinical management of patients with brain
tumors. Curr Opin Neurol. 2011, 24(6):605–615.
72. Stephenson MC, Gunner F, Napolitano A, Greenhaff PL,
Macdonald IA, Saeed N, Vennart W, Francis ST, Mor-
ris PG. Applications of multi-nuclear magnetic resonance
spectroscopy at 7T.W J Radiol. 2011, 3(4):105–113.
73. Pakin SK, Cavalcanti C, La Rocca R, Schweitzer ME, Re-
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 87
gatte RR. Ultra-high-field MRI of knee joint at 7.0T: pre-
liminary experience. Acad Radiol. 2006, 13(9):1135–1142.
74. Krug R, Carballido-Gamio J, Banerjee S, Burghardt AJ,
Link TM, Majumdar S. In vivo ultra-high-field magnetic
resonance imaging of trabecular bone microarchitecture at
7T. J Magn Reson Imaging. 2008 Apr; 27(4):854–859.
75. Stahl R, Krug R, Kelley DA. C, Zuo J, Ma CB, Majumdar S,
Link TM. Assessment of cartilage-dedicated sequences at
ultra-high-field MRI: comparison of imaging performance
and diagnostic confidence between 3.0 and 7.0T with re-
spect to osteoarthritis-induced changes at the knee joint.
Skeletal Radiol. 2009, 38(8):771–783.
76. Regatte RR, Schweitzer ME. Ultra-high-field MRI of the
musculoskeletal system at 7.0T. J Magn Reson Imaging.
2007, 25(2):262–269.
77. Wang L, Wu Y, Chang G, Oesingmann N, Schweitzer ME,
Jerschow A, Regatte RR. Rapid isotropic 3Dsodium MRI of
the knee joint in vivo at 7T. J Magn Reson Imaging. 2009,
30(3):606–614.
78. Juras V, Welsch G, B¨
ar P, Kronnerwetter C, Fujita H, Trat-
tnig S. Comparison of 3T and 7T MRI clinical sequences
for ankle imaging. Eur J Radiol. 2012, 81(8):1846–1850.
79. Trattnig S, Zb´
yˇ
n V, Juras V, Szomolanyi P, Do-
mayer S, Welsch G, Schmitt B. Ultra high field 7T
MRI for musculoskeletal applications. Available from:
http://www.healthcare.siemens.com. 2012.
80. Jordan CD, Saranathan M, Bangerter NK, Hargreaves BA,
Gold GE. Musculoskeletal MRI at 3.0T and 7.0T: a com-
parison of relaxation times and image contrast. Eur J Ra-
diol. 2013 May, 82(5):734–739.
81. Chang G, Deniz CM, Honig S, Egol K, Regatte RR, Zhu
Y, Sodickson DK, Brown R. MRI of the hip at 7T: Fea-
sibility of bone microarchitecture, high-resolution carti-
lage, and clinical imaging. J Magn Reson Imaging. 2014,
39(6):1384–1393.
82. Reddy R, Insko EK, Noyszewski EA, Dandora R, Kneeland
JB, Leigh JS. Sodium MRI of human articular cartilage in
vivo. Magn Reson Med. 1998, 39(5):697–701.
83. Reddy R, Li S, Noyszewski E. A, Kneeland J. B, Leigh J.
S. In vivo sodium multiple quantum spectroscopy of human
articular cartilage. Magn Reson Med. 1997, 38(2):207–214.
84. Shapiro EM, Borthakur A, Dandora R, Kriss A, Leigh JS,
Reddy R. Sodium visibility and quantitation in intact bovine
articular cartilage using high field 23Na MRI and MRS. J
Magn Reson. 2000, 142(1):24–31.
85. Bashir A, Gray ML, Hartke J, Burstein D. Nondestructive
imaging of human cartilage glycosaminoglycan concentra-
tion by MRI. Magn Reson Med. 1999, 41(5):857–865.
86. Peters N, van Esser S, Bos van dench M, Storm R, Plaisier P,
van Dalen T, Diepstraten S, Weits T, Westenend P, Stapper
G, Fernandez-Gallardo M, Borel Rinkes I, van Hillegers-
berg R, Mali W, Peeters P. Preoperative MRI and surgi-
cal management in patients with nonpalpable breast cancer:
The MONET – Randomised controlled trial. Eur J Cancer.
2011, 47(6):879–886.
87. Bassett LW, Dhaliwal SG, Eradat J, Khan O, Farria DF,
Brenner RJ, Sayre JW. National trends and practices in
breast MRI. Am J Roentol. 2008, 191(2):332–339.
88. Kuhl CK, Jost P, Morakkabati N, Zivanovic O, Schild HH,
Gieseke J. Contrast-enhanced MR imaging of the breast at
3.0 and 1.5T in the same patients: initial experience. Radi-
ology. 2006, 239(3):666–676.
89. Peters NHGM, Borel Rinkes IHM, Zuithoff NPA, Mali
WPTM, Moons KGM, Peeters PHM. Metaanalysis of MR
imaging in the diagnosis of breast lesions. Radiology. 2008,
246(1):116–124.
90. Brown RD, Mcgorty K, Moy L, DeGregorio S, Sodickson
DK, Wiggins GC. Sub-millimeter breast imaging and re-
laxivity characterization at 7T. Proc. Intl. Soc. Mag. Reson.
Med. 2011, 19:30–92.
91. Stehouwer BL, Klomp DWJ, Korteweg MA, Verkooijen
HM, Luijten PR, Mali WPTM, Van Den Bosch MAAJ,
Veldhuis WB. 7T versus 3T contrast-enhanced breast mag-
netic resonance imaging of invasive ductulolobular carci-
noma: first clinical experience. Magn Reson Imaging. 2013,
31(4):613–617.
92. Bogner W, Minarikova L, Pinker K, Baltzer P, Zaric O,
Chmelik M, Zb´
yˇ
n S, Helbich T, Trattnig S, Gruber S. Clin-
ical multiparametric MR imaging of breast tumors at 7T.
Available from: http://www.healthcare.siemens.com. 2014.
93. Pinker K, Baltzer P, Bogner W, Leithner D, Trattnig S,
Zaric O, Dubsky P, Bago-Horvath Z, Rudas M, Gruber
S, Weber M, Helbich TH. Multiparametric MR imag-
ing with high-resolution dynamic contrast-enhanced and
diffusion-weighted imaging at 7T improves the assessment
of breast tumors: a feasibility study. Radiology. 2015 Aug;
276(2):360–370.
94. van de Bank BL, Voogt IJ, Italiaander M, Stehouwer BL,
Boer VO, Luijten PR, Klomp DWJ. Ultra high spatial and
temporal resolution breast imaging at 7T. NMR Biomed.
2012, Apr; 26(4):367–375.
95. Kuroki Y, Nasu K, Kuroki S, Murakami K, Hayashi T,
Sekiguchi R, Nawano S. Diffusion-weighted imaging of
breast cancer with the sensitivity encoding technique: anal-
ysis of the apparent diffusion coefficient value. Magn Reson
Med Sci. 2004, 3(2):79–85.
96. Pinker K, Bogner W, Baltzer P, Trattnig S, Gruber S,
Abeyakoon O, Bernathova M, Zaric O, Dubsky P, Bago-
Horvath Z, Weber M, Leithner D, Helbich TH. Clinical ap-
plication of bilateral high temporal and spatial resolution
dynamic contrast-enhanced magnetic resonance imaging of
the breast at 7T. Eur Radiol. 2014, 24(4):913–920.
97. Pickles MD, Gibbs P, Lowry M, Turnbull LW. Diffusion
changes precede size reduction in neoadjuvant treatment of
breast cancer. Magn. Reson. Imaging. 2006, 24(7):843–847.
98. Korteweg MA, Veldhuis WB, Visser F, Luijten PR, Mali
WPTM, van Diest PJ, van den Bosch MAAJ, Klomp DJ.
Feasibility of 7T breast magnetic resonance imaging deter-
mination of intrinsic sensitivity and high-resolution mag-
netic resonance imaging, diffusion-weighted imaging, and
1H-magnetic resonance spectroscopy of breast cancer pa-
tients receiving neoadj. Invest Radiol. 2011, 46(6):370–376.
99. Hoeks CMA, Barentsz JO, Hambrock T, Yakar D, Som-
ford DM, Heijmink SWTPJ, Scheenen TWJ, Vos PC, Huis-
Volume 44, Issues 1-2, 2016
88 Karamat, Darvish-Molla, & Santos-Diaz
man H, van Oort IM, Witjes JA, Heerschap A, Futterer JJ.
Prostate cancer: multiparametric MR imaging for detection,
localization, and staging. Radiology. 2011, 261(1):46–66.
100. Wu C-L, Jordan KW, Ratai EM, Sheng J, Adkins CB, De-
feo EM, Jenkins BG, Ying L, McDougal WS, Cheng LL.
Metabolomic imaging for human prostate cancer detection.
Sci Transl Med. 2010, 2(16):16ra8.
101. Kobus T, Bitz AK, van Uden MJ, Lagemaat MW, Rothgang
E, Orzada S, Heerschap A, Scheenen TWJ. In vivo 31P MR
spectroscopic imaging of the human prostate at 7T: safety
and feasibility. Magn Reson Med. 2012, 68(6):1683–1695.
102. Rosenkrantz AB, Zhang B, Ben-Eliezer N, Le-Nobin J,
Melamed J, Deng FM, Taneja SS, Wiggins GC. T2-
weighted prostate MRI at 7T using a simplified external
transmit-receive coil array: correlation with radical prosta-
tectomy findings in two prostate cancer patients. J Magn
Reson Imaging. 2013, 232:226–232.
103. Arteaga De Castro CS, Van Den Bergen B, Luijten PR,
Van Der Heide UA, Van Vulpen M, Klomp DWJ. Improv-
ing SNR and B1 transmit field for an endorectal coil in 7T
MRI and MRS of prostate cancer. Magn Reson Med. 2012,
68(1):311–318.
104. Metzger GJ, Snyder C, Akgun C, Vaughan T, Ugurbil K,
Van De Moortele PF. Local B1 + shimming for prostate
imaging with transceiver arrays at 7T based on subjectde-
pendent transmit phase measurements. Magn Reson Med.
2008, 59(2):396–409.
105. Vos EK, Lagemaat MW, Barentsz JO, F¨
utterer JJ, Z´
amecnik
P, Roozen H, Orzada S, Bitz AK, Maas MC, Scheenen TWJ.
Image quality and cancer visibility of T2-weighted mag-
netic resonance imaging of the prostate at 7T. Eur J Radiol.
2014, 24(8):1950–1958.
106. Maas MC, Vos EK, Lagemaat MW, Bitz AK, Orzada S,
Kobus T, Kraff O, Maderwald S, Ladd ME, Scheenen
TWJ. Feasibility of T2-weighted turbo spin echo imaging
of the human prostate at 7 tesla. Magn Reson Med. 2014,
71(5):1711–1719.
107. Snyder CJ, DelaBarre L, Metzger GJ, van de Moortele P-F,
Akgun C, Ugurbil K, Vaughan JT. Initial results of cardiac
imaging at 7T. Magn Reson Med. 2009, 61(3):517–524.
108. Vaughan JT, Snyder CJ, Delabarre LJ, Bolan PJ, Tian J,
Bolinger L, Adriany G, Andersen P, Strupp J, Ugurbil K.
Whole-body imaging at 7T: Preliminary results. Magn Re-
son Med. 2009, 61(1):244–248.
109. Maderwald S. 7T Human in vivo cardiac imaging with an
8-channel transmit/receive array introduction; Magn Reson
Med. 2009, 17:2716.
110. Versluis MJ, Tsekos N, Smith NB, Webb AG. Simple RF de-
sign for human functional and morphological cardiac imag-
ing at 7T. J Magn Reson. 2009, 200(1):161–166.
111. Frauenrath T, Hezel F, Heinrichs U, Kozerke S, Utting JF,
Kob M, Butenweg C, Boesiger P, Niendorf T. Feasibility
of cardiac gating free of interference with electromagnetic
fields at 1.5T, 3.0T and 7.0T using an MR-stethoscope. In-
vest Radiol. 2009, 44(9):539–547.
112. Suttie JJ, Delabarre L, Pitcher A, van de Moortele PF, Dass
S, Snyder CJ, Francis JM, Metzger GJ, Weale P, Ugurbil
K, Neubauer S, Robson M, Vaughan T. 7T (T) human car-
diovascular magnetic resonance imaging using FLASH and
SSFP to assess cardiac function: validation against 1.5T and
3T. NMR in biomedicine. 2012, 25(1):27–34.
113. Stuber M, Botnar RM, Fischer SE, Lamerichs R, Smink J,
Harvey P, Manning WJ. Preliminary report on in vivo coro-
nary MRA at 3T in humans. Magn Reson Med. September
2002, 48(3):42–59.
114. Hinton DP, Wald LL, Pitts J, Schmitt F. Comparison of car-
diac MRI on 1.5 and 3.0T clinical whole body systems. In-
vest Radiol. 2003, 38(7):436–442.
115. Hezel F, Thalhammer C, Waiczies S, Schulz-menger J,
Niendorf T. High spatial resolution and temporally resolved
T2mapping of normal human myocardium at 7.0T: an
ultrahigh field magnetic resonance feasibility study. PLoS
One. 2012, 7(12):1–14.
116. van Elderen SGC, Versluis MJ, Westenberg JJM, Agarwal
H, Smith NB, Stuber M, de Roos A, Webb AG. Right coro-
nary MR angiography at 7T: a direct quantitative and quali-
tative comparison with 3T in young healthy volunteers. Ra-
diology. 2010, 257(1):254–259.
117. Niendorf T, Graessl A, Thalhammer C, Dieringer MA,
Kraus O, Santoro D, Fuchs K, Hezel F, Waiczies S, Itter-
mann B, Winter L. Progress and promises of human cardiac
magnetic resonance at ultrahigh fields: A physics perspec-
tive. J Magn Reson. 2013, 229:208–222.
118. Morze CV, Xu D, Purcell DD, Hess CP, Mukherjee P, Sa-
loner D, Kelley DAC, Vigneron DB. Intracranial time-of-
flight MR angiography at 7T with comparison to 3T. J Magn
Reson Imaging. 2007, 904:900–904.
119. Nowinski WL, Puspitasaari F, Volkau I, Marchenko Y,
Knopp MV. Comparison of magnetic resonance angiogra-
phy scans on 1.5, 3, and 7T units: a quantitative study
of 3-dimensional cerebrovasculature. J Neuroimag. 2013,
23(1):86–95.
120. Stamm AC, Wright CL, Knopp MV, Schmalbrock P, Hever-
hagen JT. Phase contrast and time-of-flight magnetic reso-
nance angiography of the intracerebral arteries at 1.5, 3 and
7T. Magn Reson Imaging. 2012, 31(4):545–549.
121. Stalder AF, Dong Z, Yang Q, Bock J, Hennig J, Markl
M, Li K. Four-dimensional flow-sensitive MRI of the tho-
racic aorta: 12-Versus 32-channel coil arrays. J Magn Reson
Imaging. 2012, 35(1):190–195.
122. Kwak Y, Nam S, Akc¸akaya M, Basha TA, Goddu B, Man-
ning WJ, Tarokh V, Nezafat R. Accelerated aortic flow as-
sessment with compressed sensing with and without use of
the sparsity of the complex difference image. Magn Reson
Med. 2013, 70(3):851–858.
123. Baltes C, Kozerke S, Hansen MS, Pruessmann KP, Tsao J,
Boesiger P. Accelerating cine phase-contrast flow measure-
ments using k-t BLAST and k-t SENSE. Magn Reson Med.
2005, 54(6):1430–1438.
124. Niendorf T, Paul K, Oezerdem C, Graessl A, Klix S,
Huelnhagen T, Hezel F, Rieger J, Waiczies H, Frahm J,
Nagel AM, Oberacker E, Winter L. Whither human cardiac
and body magnetic resonance at ultrahigh fields? Technical
Critical ReviewsTM in Biomedical Engineering
MRI at 7T from Research to Clinic 89
advances, practical considerations, applications, and clini-
cal opportunities. NMR in Biomedicine.
125. Hess AT, Bissell MM, Ntusi NA. B, Lewis AJM, Tunni-
cliffe EM, Greiser A, Stalder AF, Francis JM, Myerson SG,
Neubauer S, Robson MD. Aortic 4D flow: Quantification of
signal-to-noise ratio as a function of field strength and con-
trast enhancement for 1.5T, 3T, and 7T. Magn Reson Med.
2014, 871:1864–1871.
126. Metzger GJ, Auerbach EJ, Akgun C, Simonson J, Bi X,
Uˇ
gurbil K, van de Moortele P-F. Dynamically applied
B1 + shimming solutions for non-contrast enhanced re-
nal angiography at 7.0T. Magn Reson Med. 2013, 69(1):
114–126.
127. Umutlu L, Maderwald S, Kraff O, Kinner S, Schaefer LC,
Wrede K, Antoch G, Forsting M, Ladd ME, Lauenstein TC,
Quick HH. New look at renal vasculature: 7T nonenhanced
T1-weighted FLASH imaging. J Magn Reson Imag-
ing. 2012, 36(3):714–721.
128. Schlaudecker JD, Bernheisel CR. Gadoliniumassociated
nephrogenic systemic fibrosis. Am Fam Phys. 2009,
80(7):711–714.
129. O’Hare A, Johansen K. Lower-extremity peripheral arterial
diseaseamong patients with end-stage renal disease. J. Am.
Soc. Nephrol. 2001, 12(12):2838–2847.
130. Fischer A, Maderwald S, Orzada S, Johst S, Sch¨
afer LC,
Ladd ME, Nassenstein K, Umutlu L, Lauenstein TC.
Nonenhanced magnetic resonance angiography of the lower
extremity vessels at 7T: initial experience. Invest Radiol.
2013, 48(7):525–534.
131. Fischer A, Maderwald S, Johst S, Orzada S, Ladd ME,
Umutlu L, Lauenstein TC, Kniemeyer H-W, Nassenstein K.
Initial evaluation of non-contrast-enhanced magnetic reso-
nance angiography in patients with peripheral arterial oc-
clusive disease at 7T. Invest Radiol. 2014, 49(5):331–338.
Volume 44, Issues 1-2, 2016
... It arises from the variation in the dielectric properties of tissues and the interference of RF waves. 4 The latter becomes especially pronounced for ultrahigh-field MRI, as the Larmor frequency increases with the field strength, and the electromagnetic (EM) wavelength shortens and reaches the dimension of the imaged organ. The B 1 + inhomogeneity leads to changes in the signal intensity and contrast, complicating image evaluation and diagnosis. ...
... 14 Therefore, safety rules consider the heating measure directly related to the EM nature of the energy dissipation-the specific absorption rate (SAR). 4,15,16 It corresponds to the average RF power absorbed by a unit mass of an object and directly depends on the electrical field. ...
Article
Full-text available
Purpose Testing an RF coil prototype on subjects involves laborious verifications to ensure its safety. In particular, it requires preliminary electromagnetic simulations and their validations on phantoms to accurately predict the specific absorption rate (SAR). For coil design validation with a simpler safety procedure, the restricted SAR (rS) mode is proposed, enabling representative first experiments in vivo. The goal of the developed approach is to accelerate the transition of a custom coil system from prototype to clinical use. Methods The restricted specific absorption rate (SAR) (rS) mode imposes a radical limitation on the transmitted RF power based on a worst‐case scenario of local RF power absorption. The limitations used are independent of the SAR spatial distribution, making this approach unconditionally safe. The developed rS protocol contains the sequences required for coil evaluation and satisfies the imposed rS conditions. It provides a quantitative characterization of the coil transmission and reception profiles and a qualitative evaluation of the anatomical images. Protocol validation was performed on commercial and pre‐industrial prototype coils on a small cohort of healthy volunteers. Results The proposed rS protocol enables coil evaluation within an acquisition time compatible with common clinical protocol duration. The total time of all evaluation steps does not exceed 17 min. At the same time, the global SAR remains 100 times less than the International Electrotechnical Commission safety limit for played sequences. Conclusion The rS protocol allows characterizing and comparing coil prototypes on volunteers without extensive electromagnetic calculations and phantom validations in an unconditionally safe way.
... [10][11][12] Nevertheless, the increase of the static field strength (i.e., operating frequency) leads to challenges, such as the shortening of the radiofrequency (RF) wavelength in tissue and the appearance of standing waves in the RF field distribution. [13][14][15] When imaging the head at 7T, this leads to a pronounced inhomogeneity of the transmit RF field-with a maximum B + 1 field in the brain's center and pronounced intensity drops near the temporal lobes. ...
Article
Full-text available
Purpose Ultra‐high field MRI offers unprecedented detail for noninvasive visualization of the human brain. However, brain imaging is challenging at 7T due to the B 1+$$ {}_1^{+} $$ field inhomogeneity, which results in signal intensity drops in temporal lobes and a bright region in the brain center. This study aims to evaluate using a metasurface to improve brain imaging at 7T and simplify the investigative workflow. Methods Two flexible metasurfaces comprising a periodic structure of copper strips and parallel‐plate capacitive elements printed on an ultra‐thin substrate were optimized for brain imaging and implemented via PCB. We considered two setups: (1) two metasurfaces located near the temporal lobes and (2) one metasurface placed near the occipital lobe. The effect of metasurface placement on the transmit efficiency and specific absorption rate was evaluated via electromagnetic simulation studies with voxelized models. In addition, their impact on signal‐to‐noise ratio (SNR) and diagnostic image quality was assessed in vivo for two male and one female volunteers. Results Placement of metasurfaces near the regions of interest led to an increase in homogeneity of the transmit field by 5% and 10.5% in the right temporal lobe and occipital lobe for a male subject, respectively. SAR efficiency values changed insignificantly, dropping by less than 8% for all investigated setups. In vivo studies also confirmed the numerically predicted improvement in field distribution and receive sensitivity in the desired ROI. Conclusion Optimized metasurfaces enable homogenizing transmit field distribution in the brain at 7T. The proposed lightweight and flexible structure can potentially provide MR examination with higher diagnostic value images.
... It also provides better mapping of eloquent cortical regions and white matter tracts critical for surgical planning. However, limitations include image distortions, limited accessibility, the potential for increased radiofrequency power deposition in the brain, and incidental findings that could lead to unnecessary testing and anxiety [105][106][107]. Using multiple techniques in combination is necessary for a more complete understanding of brain function. ...
Article
Full-text available
Electrophysiology is the study of the electrical properties of biological tissues, which involves the movement of ions across cell membranes. The analysis of the movement of electrical charges through the body has a wide range of biomedical applications, such as diagnosing and planning treatment in cardiovascular, nervous systems, muscular, and gastrointestinal disorders. The dielectric properties of biological tissues change according to the water content in the tissue and are measured as permittivity and conductivity relative to the frequency of the electrical field. This principle has been applied in diagnostics and therapeutics using microwave energysuch as imaging and ablation, etc. This review article summarizes the potential use of measuring dielectric properties using microwave imaging and how it can augment electrophysiological studies in medicine.
... High field MRI brings significantly improved SNR and spatial resolution when compared to MRI at 1.5 T and 3 T. [1][2][3][4] To maximize the potential of high field MRI, however, key challenges remain. Two important challenges are B 1 inhomogeneity 2,4-6 and higher specific absorption rate levels (SAR) 4,7,8 associated with the shorter electromagnetic wavelengths. ...
Article
Full-text available
Purpose Mitigating coupling effects between coil elements represents a continuing challenge. Here, we present a 16‐bowtie slot volume coil arranged in eight independent dual‐slot modules without the use of any decoupling circuits. Methods Two electrically short “bowtie” slot antennas were used to form a “module.” A bowtie configuration was chosen because electromagnetic modeling results show that bowtie slots exhibit improved B1+Pin$$ \frac{B_1^{+}}{\sqrt{P_{in}}} $$ efficiency when compared to thin rectangular slots. An eight‐module volume coil was evaluated through electromagnetic modeling, bench tests, and MRI experiments at 4.7 T. Results Bench tests indicate that worst‐case coupling between modules did not exceed −14.5 dB. MR images demonstrate well‐localized patterns about single excited modules confirming the low coupling between modules. Homogeneous MR images were acquired from a synthesized quadrature birdcage transmit mode. MRI experiments show that the RF power requirements for the proposed coil are 9.2 times more than a birdcage coil. Whereas from simulations performed to assess the proposed coil losses, the total power dissipated in the phantom was 1.1 times more for the birdcage. Simulation results at 7 T reveal an equivalent B1⁺ homogeneity when compared with an eight‐dipole coil. Conclusion Although exhibiting higher RF power requirements, as a transmit coil when the power availability is not a restriction, the inherently low coupling between electrically short slots should enable the use of many slot elements around the imaging volume. The slot module described in this paper should be useful in the design of multi‐channel transmit coils.
... Meanwhile, MRI at ultra-high field encounters numerous challenges, especially in abdominal imaging. The two main challenges of 7 T MRI are transmit B1 + inhomogeneities in large body cross sections and radiofrequency (RF) power deposition in tissue [10]. The abdominal imaging applications at 7 T are known to be impaired due to the short wavelength of the 298 MHz RF field and enhanced tissue absorption, leading to strong flip angle variations and limited penetration depth [11]. ...
Article
Full-text available
Background Recently, a whole-body 5 T MRI scanner was developed to open the door of abdominal imaging at high-field strength. This prospective study aimed to evaluate the feasibility of renal imaging at 5 T and compare the image quality, potential artifacts, and contrast ratios with 3 T. Methods Forty healthy volunteers underwent MRI examination both at 3 T and 5 T. MRI sequences included T1-weighted gradient-echo (GRE), T2-weighted fast spin echo, diffusion-weighted imaging, and multi-echo GRE T2* mapping. Image quality and presence of artifacts were assessed for all sequences using four-point scales. For anatomical imaging, the signal-to-noise ratio (SNR) and contrast ratio (CR) of abdomen organ tissues were calculated. Besides, for functional imaging, the contrast-to-noise ratio of cortex/medulla was calculated. Wilcoxon signed rank-sum test was used to compare the visual evaluation scores and quantitative measurements between 3 and 5 T images. Results Compared to 3 T examination, T1-weighted sequence at 5 T showed significantly better image quality with higher conspicuity of the renal veins and arteries, and comparable artifacts. Image quality was comparable between both field strengths on T2-weighted images, whereas a significantly higher level of artifacts was observed at 5 T. Besides, 5 T MRI contributed to higher SNR and CR for abdomen organ tissues. For functional imaging, 5 T MRI showed improved corticomedullar discrimination. There was no significant difference between apparent diffusion coefficient of renal at 3 T and 5 T, while 5 T MRI resulted in significantly shorter T2* values in both cortex and medulla. Conclusions 5 T MRI provides anatomical and functional images of the kidney with sufficient image quality.
Article
Both Parkinson disease (PD) and Essential tremor (ET) are movement disorders causing tremors in elderly individuals. Although PD and ET are different disease, they often present with similar initial symptoms, making their differentiation challenging with magnetic resonance imaging (MRI) techniques. This study aimed to identify structural brain differences among PD, ET, and health controls (HCs) using 7-Tesla (T) MRI. We assessed the whole-brain parcellation in gray matter volume, thickness, subcortical volume, and small regions of basal ganglia in PD (n = 18), ET (n = 15), and HCs (n = 18), who were matched for age and sex. Brain structure analysis was performed automatic segmentation through Freesurfer software. Small regions of basal ganglia were manually segmented by ITK-SNAP. Additionally, we examined the associations between clinical indicators (symptom duration, unified Parkinson diseases rating scale (UPDRS), and clinical rating scale for tremor (CRST)) and brain structure. PD showed a significant reduction in gray matter volume in the postcentral region compared to ET. ET showed a significant reduction in cerebellum volume compared to HCs. There was a negative correlation between CRST scores (B and C) and gray matter thickness in right superior frontal in ET. This study demonstrated potential of 7T MRI in differentiating brain structure differences among PD, ET, and HCs. Specific findings, such as parietal lobe atrophy in PD compared to ET and cerebellum atrophy in ET compared to HCs, the importance of advanced imaging techniques in accurately diagnosing and distinguishing between movement disorders that present with similar initial symptoms.
Article
Full-text available
Objectives Locally low-rank (LLR) denoising of functional magnetic resonance imaging (fMRI) time series image data is extended to multi-echo (ME) data. The proposed method extends the capabilities of non-physiologic noise suppression beyond single-echo applications with a dedicated ME algorithm. Materials and Methods Following an institutional review board (IRB) approved protocol, resting-state fMRI data were acquired in 7 healthy subjects. A compact 3T scanner enabled whole-brain acquisition of multiband ME fMRI data at high spatial resolution (1.4 × 1.4 × 2.8 mm ³ ) with a 1810 ms repetition time (TR). Image data were denoised with ME-LLR preceding functional processing. The results of connectivity maps generated from denoised data were compared with maps generated with equivalent processing of non-denoised images. To assess ME-LLR as a method to reduce scan time, comparisons were made between maps computed from image data with full and retrospectively truncated durations. Assessments were completed with seed-based connectivity analyses using echo-combined image data. In a feasibility assessment, nondenoised and denoised full-duration echo-combined data were equivalently processed with independent component analysis (ICA) and compared. Results ME-LLR denoising yielded strengthened resting-state network connectivity maps after nuisance regression and seed-based connectivity analysis. In assessing ME-LLR as a scan reduction mechanism, maps generated from denoised data at half scan time showed comparable quality with maps generated from full-duration, non-denoised data, at both single subject and group levels. ME-LLR substantially increased temporal signal-to-noise ratio (tSNR) for image data respective to each individual echo and for image data after nuisance regression. Among echo-specific image volumes, increases in tSNR yielded by ME-LLR were most pronounced for image data with the longest echo time and thereby lowest SNR. ICA showed resting-state networks consistently identified between non-denoised and denoised data, with clearer demarcation of networks for ME-LLR. Conclusions ME-LLR is demonstrated to suppress non-physiologic noise, enhance functional connectivity map quality, and could potentially facilitate scan time reduction in ME-fMRI.
Article
Purpose: MRI has become an essential diagnostic imaging modality for peripheral nerve pathology. Early MR imaging for peripheral nerve depended on inferred nerve involvement by visualizing downstream effects such as denervation muscular atrophy; improvements in MRI technology have made possible direct visualization of the nerves. In this paper, we share our early clinical experience with 7T for benign neurogenic tumors. Materials: Patients with benign neurogenic tumors and 7T MRI examinations available were reviewed. Cases of individual benign peripheral nerve tumors were included to demonstrate 7T MRI imaging characteristics. All exams were performed on a 7T MRI MAGNETOM Terra using a 28-channel receive, single-channel transmit knee coil. Results: Five cases of four pathologies were selected from 38 patients to depict characteristic imaging features in different benign nerve tumors and lesions using 7T MRI. Conclusion: The primary advantage of 7T over 3T is an increase in signal-to-noise ratio which allows higher in plane resolution so that the smallest neural structures can be seen and characterized. This improvement in MR imaging provides the opportunity for more accurate diagnosis and surgical planning in selected cases. As this technology continues to evolve for clinical purposes, we anticipate increasing applications and improved patient care using 7T MRI for the diagnosis of peripheral nerve masses.
Article
Full-text available
Purpose Introduce Shimming Toolbox ( https://shimming‐toolbox.org), an open‐source software package for prototyping new methods and performing static, dynamic, and real‐time B0 shimming as well as B1 shimming experiments. Methods Shimming Toolbox features various field mapping techniques, manual and automatic masking for the brain and spinal cord, B0 and B1 shimming capabilities accessible through a user‐friendly graphical user interface. Validation of Shimming Toolbox was demonstrated in three scenarios: (i) B0 dynamic shimming in the brain at 7T using custom AC/DC coils, (ii) B0 real‐time shimming in the spinal cord at 3T, and (iii) B1 static shimming in the spinal cord at 7T. Results The B0 dynamic shimming of the brain at 7T took about 10 min to perform. It showed a 47% reduction in the standard deviation of the B0 field, associated with noticeable improvements in geometric distortions in EPI images. Real‐time dynamic xyz‐shimming in the spinal cord took about 5 min and showed a 30% reduction in the standard deviation of the signal distribution. B1 static shimming experiments in the spinal cord took about 10 min to perform and showed a 40% reduction in the coefficient of variation of the B1 field. Conclusion Shimming Toolbox provides an open‐source platform where researchers can collaborate, prototype and conveniently test B0 and B1 shimming experiments. Future versions will include additional field map preprocessing techniques, optimization algorithms, and compatibility across multiple MRI manufacturers.
Article
Objective This study investigates a locally low-rank (LLR) denoising algorithm applied to source images from a clinical task-based functional MRI (fMRI) exam before post-processing for improving statistical confidence of task-based activation maps. Methods Task-based motor and language fMRI was obtained in eleven healthy volunteers under an IRB approved protocol. LLR denoising was then applied to raw complex-valued image data before fMRI processing. Activation maps generated from conventional non-denoised (control) data were compared with maps derived from LLR-denoised image data. Four board-certified neuroradiologists completed consensus assessment of activation maps; region-specific and aggregate motor and language consensus thresholds were then compared with nonparametric statistical tests. Additional evaluation included retrospective truncation of exam data without and with LLR denoising; a ROI-based analysis tracked t-statistics and temporal SNR (tSNR) as scan durations decreased. A test-retest assessment was performed; retest data were matched with initial test data and compared for one subject. Results fMRI activation maps generated from LLR-denoised data predominantly exhibited statistically significant ( p = 4.88×10 –4 to p = 0.042; one p = 0.062) increases in consensus t-statistic thresholds for motor and language activation maps. Following data truncation, LLR data showed task-specific increases in t-statistics and tSNR respectively exceeding 20 and 50% compared to control. LLR denoising enabled truncation of exam durations while preserving cluster volumes at fixed thresholds. Test-retest showed variable activation with LLR data thresholded higher in matching initial test data. Conclusion LLR denoising affords robust increases in t-statistics on fMRI activation maps compared to routine processing, and offers potential for reduced scan duration while preserving map quality.
Article
Full-text available
The objective of this study was to document and review advances and groundbreaking progress in cardiac and body MR at ultrahigh fields (UHF, B0 ≥ 7.0 T) with the goal to attract talent, clinical adopters, collaborations and resources to the biomedical and diagnostic imaging communities. This review surveys traits, advantages and challenges of cardiac and body MR at 7.0 T. The considerations run the gamut from technical advances to clinical opportunities. Key concepts, emerging technologies, practical considerations, frontier applications and future directions of UHF body and cardiac MR are provided. Examples of UHF cardiac and body imaging strategies are demonstrated. Their added value over the kindred counterparts at lower fields is explored along with an outline of research promises. The achievements of cardiac and body UHF-MR are powerful motivators and enablers, since extra speed, signal and imaging capabilities may be invested to overcome the fundamental constraints that continue to hamper traditional cardiac and body MR applications. If practical obstacles, concomitant physics effects and technical impediments can be overcome in equal measure, sophisticated cardiac and body UHF-MR will help to open the door to new MRI and MRS approaches for basic research and clinical science, with the lessons learned at 7.0 T being transferred into broad clinical use including diagnostics and therapy guiding at lower fields. Copyright © 2015 John Wiley & Sons, Ltd. Copyright © 2015 John Wiley & Sons, Ltd.
Article
Full-text available
Objectives: To assess the image quality of T2-weighted (T2w) magnetic resonance imaging of the prostate and the visibility of prostate cancer at 7 Tesla (T). Materials & methods: Seventeen prostate cancer patients underwent T2w imaging at 7T with only an external transmit/receive array coil. Three radiologists independently scored images for image quality, visibility of anatomical structures, and presence of artefacts. Krippendorff's alpha and weighted kappa statistics were used to assess inter-observer agreement. Visibility of prostate cancer lesions was assessed by directly linking the T2w images to the confirmed location of prostate cancer on histopathology. Results: T2w imaging at 7T was achievable with 'satisfactory' (3/5) to 'good' (4/5) quality. Visibility of anatomical structures was predominantly scored as 'satisfactory' (3/5) and 'good' (4/5). If artefacts were present, they were mostly motion artefacts and, to a lesser extent, aliasing artefacts and noise. Krippendorff's analysis revealed an α = 0.44 between three readers for the overall image quality scores. Clinically significant cancer lesions in both peripheral zone and transition zone were visible at 7T. Conclusion: T2w imaging with satisfactory to good quality can be routinely acquired, and cancer lesions were visible in patients with prostate cancer at 7T using only an external transmit/receive body array coil. Key points: • Satisfactory to good T2-weighted image quality of the prostate is achievable at 7T. • Periprostatic lipids appear hypo-intense compared to healthy peripheral zone tissue at 7T. • Prostate cancer is visible on T2-weighted MRI at 7T.
Article
Full-text available
To report design of a simplified external transmit-receive coil array for 7 Tesla (T) prostate MRI, including demonstration of the array for tumor localization using T2-weighted imaging (T2WI) at 7T before prostatectomy. Following simulations of transmitter designs not requiring parallel transmission or radiofrequency-shimming, a coil array was constructed using loop elements, with anterior and posterior rows comprising one transmit-receive element and three receive-only elements. This coil structure was optimized using a whole-body phantom. In vivo sequence optimization was performed to optimize achieved flip angle (FA) and signal to noise ratio (SNR) in prostate. The system was evaluated in a healthy volunteer at 3T and 7T. The 7T T2WI was performed in two prostate cancer patients before prostatectomy, and localization of dominant tumors was subjectively compared with histopathological findings. Image quality was compared between 3T and 7T in these patients. Simulations of the B1 (+) field in prostate using two-loop design showed good magnitude (B1 (+) of 0.245 A/m/w(1/2) ) and uniformity (nonuniformity [SD/mean] of 10.4%). In the volunteer, 90° FA was achieved in prostate using 225 v 1 ms hard-pulse (indicating good efficiency), FA maps confirmed good uniformity (14.1% nonuniformity), and SNR maps showed SNR gain of 2.1 at 7T versus 3T. In patients, 7T T2WI showed excellent visual correspondence with prostatectomy findings. 7T images demonstrated higher estimated SNR (eSNR) in benign peripheral zone (PZ) and tumor compared with 3T, but lower eSNR in fat and slight decreases in tumor-to-PZ contrast and PZ-homogeneity. We have demonstrated feasibility of a simplified external coil array for high-resolution T2-weighted prostate MRI at 7T.J. Magn. Reson. Imaging 2013. © 2013 Wiley Periodicals, Inc.
Article
Purpose To ascertain whether multiparametric magnetic resonance (MR) imaging of the breast in combination with dynamic contrast material-enhanced (DCE) imaging and diffusion-weighted imaging (DWI) at 7 T is feasible and improves diagnostic accuracy. Materials and Methods From December 2011 to December 2013, 40 patients with suspicious breast lesions were included in this institutional review board-approved prospective study. Before bilateral multiparametric MR imaging of the breast at 7 T, all patients gave written informed consent. Lesions were classified according to Breast Imaging Reporting and Data System (BI-RADS) and assessed for apparent diffusion coefficient (ADC) values by two readers independently. For combined analysis of DCE MR imaging and DWI, the BI-RADS-adapted reading algorithm, which adapted ADC thresholds to the BI-RADS assessment category, was used. Diagnostic values of multiparametric, DCE MR imaging, and DWI were calculated. Receiver operating characteristic curve analysis was performed. Image quality and interreader agreement were assessed. Histopathologic results were used as the highest standard. Results There were 29 malignant and 17 benign lesions (range, 6-95 mm; mean, 23.3 mm). Multiparametric MR imaging yielded a sensitivity of 100% (29 of 29 lesions), a specificity of 88.2% (16 of 18 lesions), and an area under the curve of 0.941, which was greater than for DCE MR imaging (P = .003), which had a sensitivity of 100% (29 of 29 lesions), a specificity of 53.2% (nine of 17 lesions), and an area under the curve of 0.765. DWI had a sensitivity of 93.1% (27 of 29 lesions), a specificity of 88.2% (15 of 17 lesions), and an area under the curve of 0.907. Multiparametric MR imaging at 7 T of the breast eliminated all false-negative findings and reduced false-positive findings, from eight false-positive findings with DCE MR imaging to two false-positive findings. Thus, if used clinically, 7-T multiparametric MR imaging may have potentially obviated unnecessary breast biopsies in six of eight lesions (P = .031). Multiparametric MR imaging demonstrated either excellent or good image quality and interreader agreement (κ = 0.89-1.00). Conclusion The clinical use of 7-T multiparametric MR imaging is feasible, provides good or excellent image quality, and has the potential to improve diagnostic accuracy. (©) RSNA, 2015 Online supplemental material is available for this article.
Article
At ultra-high magnetic fields, such as 7T, MR imaging can noninvasively visualize the brain in unprecedented detail and through enhanced contrast mechanisms. The increased SNR and enhanced contrast available at 7T enable higher resolution anatomic and vascular imaging. Greater spectral separation improves detection and characterization of metabolites in spectroscopic imaging. Enhanced blood oxygen level-dependent contrast affords higher resolution functional MR imaging. Ultra-high-field MR imaging also facilitates imaging of nonproton nuclei such as sodium and phosphorus. These improved imaging methods may be applied to detect subtle anatomic, functional, and metabolic abnormalities associated with a wide range of neurologic disorders, including epilepsy, brain tumors, multiple sclerosis, Alzheimer disease, and psychiatric conditions. At 7T, however, physical and hardware limitations cause conventional MR imaging pulse sequences to generate artifacts, requiring specialized pulse sequences and new hardware solutions to maximize the high-field gain in signal and contrast. Practical considerations for ultra-high-field MR imaging include cost, siting, and patient experience. © 2015 American Society of Neuroradiology.
Article
PurposeTo investigate for the first time the feasibility of aortic four-dimensional (4D) flow at 7T, both contrast enhanced (CE) and non-CE. To quantify the signal-to-noise ratio (SNR) in aortic 4D flow as a function of field strength and CE with gadobenate dimeglumine (MultiHance).Methods Six healthy male volunteers were scanned at 1.5T, 3T, and 7T with both non-CE and CE acquisitions. Temporal SNR was calculated. Flip angle optimization for CE 4D flow was carried out using Bloch simulations that were validated against in vivo measurements.ResultsThe 7T provided 2.2 times the SNR of 3T while 3T provided 1.7 times the SNR of 1.5T in non-CE acquisitions in the descending aorta. The SNR gains achieved by CE were 1.8-fold at 1.5T, 1.7-fold at 3T, and 1.4-fold at 7T, respectively.Conclusion The 7T provides a new tool to explore aortic 4D flow, yielding higher SNR that can be used to push the boundaries of acceleration and resolution. Field strength and contrast enhancement at all fields provide significant improvements in SNR. Magn Reson Med 73:1864-1871, 2015. © 2014 Wiley Periodicals, Inc.
Article
The aim of this study was to achieve initial experience with non-contrast-enhanced (ne) magnetic resonance angiography (MRA) of the lower leg arteries in patients with peripheral arterial occlusive disease (PAOD) at 7 T. Seven patients with PAOD were examined on a 7-T whole-body magnetic resonance system. A custom-built 16-channel transmit/receive coil and a manually positionable AngioSURF table were used for multistation imaging. For ne-MRA, an axial T1-weighted Turbo-fast low angle shot sequence (repetition time, 700 milliseconds; echo time, 3.84 milliseconds; bandwidth, 930 Hz/pixel; voxel volume, 1 × 1 × 2 mm; matrix, 384 × 288) with phonocardiogram gating was acquired at 7 T. Acquisition time of an entire angiogram covering the vasculature from pelvis to feet amounted to approximately 30 minutes, depending on the patient's heart frequency. All patients underwent a contrast-enhanced MRA (ce-MRA) at 1.5 T as standard of reference. The presence of stenosis and occlusions was evaluated segment based and compared for both MRA techniques. The degree of stenosis was defined as low grade (<50%), high grade (50%-99%), and occlusion (100%). High-grade stenosis and occlusion were considered to be hemodynamically significant stenosis. The 7-T ne-MRA enabled a homogenous, hyperintense artery signal and nearly total venous suppression with accurate delineation of arterial anatomy both proximal and distal to stenotic disease. A total of 154 artery segments were depicted with ce-MRA at 1.5 T. At 7 T, only 124 segments (80.5%) were displayed and involved for analysis, as the iliacal region was displayed incompletely in 4 patients because of the fact that the fixed coil diameter was too small to contain the lower abdomen and pelvis of these patients. In comparison with ce-MRA at 1.5 T as the reference standard, there was total agreement regarding the characterization of an artery segment as being normal or having any kind of stenosis. Of the 124 included segments, 28 segments (23%) had hemodynamically significant stenosis evaluated with 7-T ne-MRA and 26 segments (21%) assessed with 1.5-T ce-MRA. The sensitivity and specificity values of 7-T ne-MRA for detecting segments with hemodynamically significant stenosis were 93% and 98%, respectively. Non-contrast-enhanced MRA by means of T1-weighted Turbo-fast low angle shot imaging at 7 T in patients with PAOD is feasible and allowed for good visualization of stenosis and occlusions in all analyzed artery segments in this small patient group. However, this study also shows the challenges of ultrahigh-field body imaging, and more experience is required to determine the impact of 7-T ne-MRA in clinical practice.
Article
The aim of this study was to optimize the 3-dimensional (3D) fluid attenuated inversion recovery (FLAIR) pulse sequence for isotropic high-spatial-resolution imaging of white matter (WM) and cortical lesions at 7 T. We added a magnetization-prepared (MP) FLAIR module to a Cube 3D fast spin echo sequence and optimized the refocusing flip angle train using extended phase graph simulations, taking into account image contrast, specific absorption rate (SAR), and signal-to-noise ratio (SNR) as well as T1/T2 values of the different species of interest (WM, grey matter, lesions) at 7 T. We also effected improved preparation homogeneity at 7 T by redesigning the refocusing pulse used in the MP segments. Two sets of refocusing flip angle trains-(a) an SNR-optimal and (b) a contrast-optimal set-were derived and used to scan 7 patients with Alzheimer disease/cognitive impairment and 7 patients with multiple sclerosis. Conventional constant refocusing flip MP-FLAIR images were also acquired for comparison. Lesion SNR, contrast, and lesion count were compared between the 2 optimized and the standard FLAIR sequences. Whole brain coverage with 0.8 mm isotropic spatial resolution in ∼5-minute scan times was achieved using the optimized 3D FLAIR sequences at clinically acceptable SAR levels. The SNR efficiency of the SNR-optimal sequence was significantly better than that of conventional constant refocusing flip MP-FLAIR sequence, whereas the scan time was reduced more than 2-fold (∼5 vs >10 minutes). The contrast efficiency of the contrast-optimal sequence was comparable with that of the constant refocusing flip sequence. Lesion load ascertained by lesion counting was not significantly different among the sequences. Magnetization-prepared FLAIR-Cube with refocusing flip angle trains optimized for SNR and contrast can be used to characterize WM and cortical lesions at 7 T with 0.8 mm isotropic resolution in short scan times and without SAR penalty.
Article
The objective of our study was to evaluate the clinical application of bilateral high spatial and temporal resolution dynamic contrast-enhanced magnetic resonance imaging (HR DCE-MRI) of the breast at 7 T. Following institutional review board approval 23 patients with a breast lesion (BIRADS 0, 4-5) were included in our prospective study. All patients underwent bilateral HR DCE-MRI of the breast at 7 T (spatial resolution of 0.7 mm(3) voxel size, temporal resolution of 14 s). Two experienced readers (r1, r2) and one less experienced reader (r3) independently assessed lesions according to BI-RADS®. Image quality, lesion conspicuity and artefacts were graded from 1 to 5. Sensitivity, specificity and diagnostic accuracy were assessed using histopathology as the standard of reference. HR DCE-MRI at 7 T revealed 29 lesions in 23 patients (sensitivity 100 % (19/19); specificity of 90 % (9/10)) resulting in a diagnostic accuracy of 96.6 % (28/29) with an AUC of 0.95. Overall image quality was excellent in the majority of cases (27/29) and examinations were not hampered by artefacts. There was excellent inter-reader agreement for diagnosis and image quality parameters (κ = 0.89-1). Bilateral HR DCE-MRI of the breast at 7 T is feasible with excellent image quality in clinical practice and allows accurate breast cancer diagnosis. • Dynamic contrast-enhanced 7-T MRI is being developed in several centres. • Bilateral high resolution DCE-MRI of the breast at 7 T is clinically applicable. • 7-T HR DCE-MRI of the breast provides excellent image quality. • 7-T HR DCE-MRI should detect breast cancer with high diagnostic accuracy.
Article
To demonstrate the feasibility of performing bone microarchitecture, high-resolution cartilage, and clinical imaging of the hip at 7T. This study had Institutional Review Board approval. Using an 8-channel coil constructed in-house, we imaged the hips of 15 subjects on a 7T magnetic resonance imaging (MRI) scanner. We applied: 1) a T1-weighted 3D fast low angle shot (3D FLASH) sequence (0.23 × 0.23 × 1-1.5 mm(3) ) for bone microarchitecture imaging; 2) T1-weighted 3D FLASH (water excitation) and volumetric interpolated breath-hold examination (VIBE) sequences (0.23 × 0.23 × 1.5 mm(3) ) with saturation or inversion recovery-based fat suppression for cartilage imaging; 3) 2D intermediate-weighted fast spin-echo (FSE) sequences without and with fat saturation (0.27 × 0.27 × 2 mm) for clinical imaging. Bone microarchitecture images allowed visualization of individual trabeculae within the proximal femur. Cartilage was well visualized and fat was well suppressed on FLASH and VIBE sequences. FSE sequences allowed visualization of cartilage, the labrum (including cartilage and labral pathology), joint capsule, and tendons. This is the first study to demonstrate the feasibility of performing a clinically comprehensive hip MRI protocol at 7T, including high-resolution imaging of bone microarchitecture and cartilage, as well as clinical imaging. J. Magn. Reson. Imaging 2013;. © 2013 Wiley Periodicals, Inc.