You are on page 1of 15

Note: This copy is for your personal non-commercial use only.

To order presentation-ready
copies for distribution to your colleagues or clients, contact us at www.rsna.org/rsnarights.
䡲 REVIEW

1
H MR Spectroscopy of the
Brain: Absolute Quantification of
REVIEWS AND COMMENTARY

Metabolites1

Jacobus F. A. Jansen, MS
Hydrogen 1 (1H) magnetic resonance (MR) spectroscopy
Walter H. Backes, PhD
enables noninvasive in vivo quantification of metabolite
Klaas Nicolay, PhD
concentrations in the brain. Currently, metabolite concen-
M. Eline Kooi, PhD trations are most often presented as ratios (eg, relative to
creatine) rather than as absolute concentrations. Despite
the success of this approach, it has recently been sug-
gested that relative quantification may introduce substan-
tial errors and can lead to misinterpretation of spectral
data and to erroneous metabolite values. The present re-
view discusses relevant methods to obtain absolute metab-
olite concentrations with a clinical MR system by using
single-voxel spectroscopy or chemical shift imaging. Im-
portant methodological aspects in an absolute quantifica-
tion strategy are addressed, including radiofrequency coil
properties, calibration procedures, spectral fitting meth-
ods, cerebrospinal fluid content correction, macromole-
cule suppression, and spectral editing. Techniques to ob-
tain absolute concentrations are now available and can be
successfully applied in clinical practice. Although the
present review is focused on 1H MR spectroscopy of the
brain, a large part of the methodology described can be
applied to other tissues as well.

娀 RSNA, 2006

1
From the Department of Radiology, Maastricht University
Hospital, P. Debyelaan 25, 6202 AZ Maastricht, the Neth-
erlands; and Biomedical NMR, Department of Biomedical
Engineering, Eindhoven University of Technology, Eind-
hoven, the Netherlands.
Address correspondence to J.F.A.J. (e-mail: j.f.a.jansen
@tue.nl ).

姝 RSNA, 2006

318 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

ydrogen 1 (1H) magnetic reso-

H
for healthy tissue can be used for most neously recorded from multiple adjoin-
nance (MR) spectroscopy enables clinical purposes, while for focal patho- ing spatial regions (chemical shift imag-
noninvasive quantification of in logic conditions a reference spectrum ing) find increasing use in many clinical
vivo metabolite concentrations in the can be acquired from the contralateral trials (13,15,17,21). In addition, new
brain. It has proved to be a powerful addi- region of the patient’s brain. The use of developments, such as spectral editing,
tion to the clinical assessment tools for signal ratios has the great advantage macromolecule suppression, tissue seg-
numerous pathologic conditions, includ- that it is very easy to implement be- mentation, and quantitative analysis of
ing epilepsy, multiple sclerosis, stroke, cause it does not require extra imaging spectra with dedicated software, can
cancer, and metabolic diseases (1). time and time-consuming postprocess- aid in AQ of brain metabolites. This re-
In nuclear MR, the total area under ing. Furthermore, a number of prob- view will address the implementation of
a metabolite resonance in a 1H MR lems—for example, partial volume ef- these new developments in AQ strate-
spectrum is directly proportional to the fects arising from different amounts of gies.
concentration of the metabolite. Cur- cerebrospinal fluid in the selected vol-
rently, metabolite concentrations are umes— can be largely avoided. In addi-
usually expressed as ratios (relative tion, ratios are, in some cases, more Advantages and Disadvantages of
quantification) rather than as absolute sensitive in terms of detecting changes Absolute Metabolite Concentrations
concentrations. Ratios can be useful for (eg, when one metabolite increases and In relative quantification, which yields
clinical diagnosis to characterize patho- another decreases) and can be more concentrations expressed as ratios, one
logic tissue. For example, relative quan- accurate than absolute concentrations, of the metabolite peaks measured is
tification has been successfully applied owing to specific characteristics of the used as the concentration standard and
in the diagnosis of cancer (2), leukemia analysis computer program applied (7). serves as the denominator of the peak
(3), epilepsy (4), dementia (5), and If a change is observed in the ratio of ratios. As a result, the total number of
multiple sclerosis (6). A locally obtained metabolite peaks, however, it remains quantifiable metabolites is decreased by
reference data set with normal ratios uncertain which metabolite concentration one. Furthermore, alterations in the
actually changes. This information can peak ratio do not necessarily reflect a
only be obtained from absolute concen- change in the concentration of the nu-
Essentials trations. Absolute quantification (AQ, merator. The alteration may be caused
䡲 Although metabolite ratios can be also referred to as absolute quantitation) by changes in the concentration of the
useful, absolute quantification implies that concentrations are expressed numerator, the denominator, or both or
(AQ) has an added value, since in biochemical units, such as millimoles may merely be due to changes in relax-
concentrations obtained through per kilogram wet weight. Furthermore, ation behavior.
AQ procedures benefit from un- when spectroscopic results are to be com- The assumption that the concentra-
ambiguous interpretation and pared in an interdisciplinary context (eg, tion of certain reference metabolites
may be less prone to error. with biochemically derived concentra- (eg, total creatine, choline) remains
䡲 The AQ procedure is divided into tions), ratios or arbitrary units might not constant may be incorrect under normal
two steps: (a) determination of be appropriate. conditions, as well as in many patho-
accurate peak areas for the rele- Since many factors need to be con- logic states. For example, it has been
vant metabolites and (b) conver- sidered to obtain reliable absolute con- shown in patients with temporal lobe
sion of peak areas into metabolite centrations with 1H MR spectroscopy, it epilepsy (22) that the temporal lobe ip-
concentrations by using a calibra- is hard for a newcomer to comprehend silateral to the seizure focus displays a
tion procedure. all aspects and potential pitfalls con- significant increase in total creatine and
䡲 The implementation of AQ has cerned with AQ and to decide which choline; a similar finding has also been
been greatly facilitated by the de- method to use. The latest reviews on reported in patients with frontal lobe
velopment of calibration strate- AQ were written several years ago (8– epilepsy (23). Furthermore, even the
gies and the availability of spectral 11), when AQ was more or less consid- temporal lobe contralateral to the sei-
fitting routines. ered to be a basic research tool. How-
䡲 AQ is available and can improve ever, several groups have recently pub-
the diagnostic utility of 1H MR lished clinical trials that incorporate Published online
spectroscopy. AQ. These include studies of patients 10.1148/radiol.2402050314
䡲 AQ requires more time and ex- with Alzheimer disease (12), epilepsy Radiology 2006; 240:318 –332
pertise than does relative quantifi- (13–15), multiple sclerosis (16,17), leu-
Abbreviations:
cation, and one can only benefit koencephalopathy (18), amyotrophic
AQ ⫽ absolute quantification
from AQ if all additional reference lateral sclerosis (19), and cancer (20). NAA ⫽ N-acetylaspartate
steps are executed properly; oth- Furthermore, the reviews published so RF ⫽ radiofrequency
erwise, unwanted additional er- far have mainly focused on single-voxel SNR ⫽ signal-to-noise ratio
rors may be introduced. spectroscopy, whereas spectra simulta- VOI ⫽ volume of interest

Radiology: Volume 240: Number 2—August 2006 319


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

zure focus exhibits total creatine and demonstrate metabolites. Alternatively, step, accurate peak areas for the rele-
choline concentrations that are in- multiple spectra can be obtained simul- vant metabolites are determined. In the
creased, albeit not significantly (22). In taneously from multiple adjoining spa- second step, careful calibration is used
addition, if patients have global meta- tial regions (chemical shift imaging, also to convert peak areas to metabolite con-
bolic defects, comparisons with con- referred to as MR spectroscopic imag- centrations to which the metabolite sig-
tralateral brain regions (which are as- ing) (30,31). The transmit coil should nals are referenced.
sumed to be metabolically normal) may generate RF pulses with sufficient
not be feasible (24,25). power to penetrate into all tissue re- Definitions of Concentration
Clear evidence has recently been gions of interest. Different quantification strategies lead
provided showing that, in addition to The requirements for signal recep- to different concentration standards.
causing possibly ambiguous interpreta- tion are generally similar to those for Two definitions of concentration are
tion, relative quantification may intro- transmission. The penetration of the RF commonly used: (a) molarity, which is
duce larger errors than does AQ pulses of the transmit coil and the sig- the number of moles of metabolite per
(26,27). Two independent 1H MR spec- nal-to-noise ratio (SNR) from the re- liter of tissue water, and (b) molality,
troscopy studies with healthy volunteers ceive coil are strongly dependent on the which is the number of moles of metab-
in whom single-voxel spectroscopy (27) coil loading, which is determined by the olite per kilogram of tissue water. In
and chemical shift imaging (26) were electric conductivity of the coil and the principle, molal concentrations can be
used have shown that metabolite ratios volume of the object near the coil. Vari- converted into molar concentrations
exhibit higher coefficients of variation ations in the size and tissue composition (32) by using a conversion that requires
(up to 1.6-fold) than do AQ concentra- of the head positioned in the RF coil knowledge of the specific brain tissue
tions of individual metabolites. Further- affect the amount of RF energy that gets density.
more, authors of a chemical shift imag- into and the amount of signal detected
ing study in patients with temporal lobe from the volume of interest (VOI). This Comparison of in Vivo 1H MR
epilepsy (15) reported that metabolite is one of the reasons why the electric Spectroscopy to Other Quantification
ratios were less sensitive to abnormali- characteristics of the RF coil are opti- Methods
ties than were absolute values. It is, mized before measurement begins, to Most alternative quantification methods
therefore, advisable to obtain concen- achieve the most efficient performance. can only be performed in vitro, after
trations expressed in standard units brain autopsy. These methods include
(such as millimoles per kilogram wet Characteristics of MR Spectra high-pressure liquid chromatography
weight) by applying AQ. One should re- A water-suppressed brain 1H MR spec- and high-field-strength (⬎7-T) 1H MR
alize, however, that AQ requires more trum typically displays a number of sig- spectroscopy. Whereas in vivo 1H MR
time than does relative quantification, nals that correspond to several brain spectroscopy can only determine me-
and one can benefit from AQ only if all metabolites. These signals are charac- tabolites with a concentration on the
additional referencing steps are exe- terized by one or more peaks with a order of millimoles, both high-pressure
cuted properly; otherwise, unwanted certain resonance frequency, line width liquid chromatography and high-field-
additional errors may be introduced. (full width at half maximum of the strength 1H MR spectroscopy can dem-
peak’s height), line shape (eg, lorent- onstrate concentrations in the micro-
zian or Gaussian), phase, and area. The molar range. However, several metabo-
Theoretic Background peaks are separated owing to differ- lites, including N-acetylaspartate (NAA)
ences in resonance frequency, which and phosphocreatine, are generally de-
Characteristics of the Radiofrequency Coil are caused by the difference in the tected at lower concentrations by using
Radiofrequency (RF) coils are used to chemical environment of the different in vitro methods (up to 10% reduction
transmit the RF magnetic induction field nuclei. The molecular structure of a par- relative to that detected in vivo in
(B1) and to detect the resulting signal. In ticular metabolite is reflected by a typi- healthy subjects) (33,34) owing to the
the case of clinical 1H MR spectroscopy cal peak pattern. The area of a peak is rapid degradation of these metabolites
of the brain, the patient’s head is posi- directly proportional to the number of in brain tissue samples immediately af-
tioned within the head coil, which is nuclei that contribute to it and to the ter autopsy (35,36). The assessment of
used as a receive coil, while either the concentration of the metabolite to metabolites that are not subject to deg-
head or the body coil acts as the trans- which the nuclei belong. However, the radation yields similar concentrations in
mit coil. With a dedicated pulse se- peak areas are also influenced by other both in vivo and in vitro methods (37).
quence such as point-resolved spatially factors, including T1 and T2 relaxation Typical metabolite concentrations in pa-
localized spectroscopy, or PRESS (28), times. rietal lobe white matter obtained with in
or stimulated-echo acquisition mode, or vivo 1H MR spectroscopy are 9.6 mmol
STEAM (29), a spectrum can be ob- Quantification of 1H MR Spectra per kilogram (mmol/kg) wet weight ⫾
tained from a single well-defined spatial Generally, the quantification procedure 3.0 (standard deviation) for NAA, 7.0
volume (single-voxel spectroscopy) to is divided into two steps. In the first mmol/kg wet weight ⫾ 2.0 for total cre-

320 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

atine, 1.8 mmol/kg wet weight ⫾ 0.5 for Relative Quantification Method and choline. Although this method al-
choline, and 5.2 mmol/kg wet weight ⫾ One of the simplest approaches is the lows direct comparison with other bio-
3.3 for myo-inositol (38). internal endogenous marker method. chemical measurements, it is essentially
With this approach, one of the mea- a relative quantification method and
sured peaks, originating from an endog- shares all previously mentioned possible
Quantification Strategies enous metabolite, serves as a concen- merits and drawbacks.
In this section, the most widely used tration standard (Fig 1, A). Peak ratios
quantification strategies for in vivo 1H (possibly corrected for factors such as AQ Methods
MR spectroscopy are described. The relaxation) are converted into concen- External reference method.—A vial
relative merits and drawbacks of each trations by using a value from the litera- with a known reference solution and re-
approach are indicated in Table 1. The ture for the reference metabolite, laxation properties is positioned near or
schematic setup for the brain and cali- whose concentration is supposed to be inside the RF coil (Fig 1, B) (39). Imme-
bration measurement for each case is invariant. Commonly used reference diately after the acquisition of the in
shown in Figure 1. metabolites are NAA, total creatine, vivo spectrum, a reference spectrum
from the calibration sample is obtained
while the patient’s head is still inside the
Table 1 coil. The reference spectrum can also
be obtained simultaneously with the ac-
Relative Merits and Drawbacks of Quantification Strategies quisition of patient data (40). An option
Extra Imaging Extra Imaging to account for possible B1 inhomogene-
Time for Time after Preparation Ease of ity is to measure (41) or to simulate (42)
Strategy Examination Examination Time† Use Accuracy the B1 distribution of the RF coil used. It
is important to realize that the external
Internal endogenous marker ⫹ ⫹ ⫹ ⫹ ⫺
vial might introduce substantial distor-
External reference ⫺ ⫹ 0 ⫺ 0
tions of the constant magnetic induction
Replace-and-match method ⫹ ⫺ ⫺ ⫺ ⫹
field (B0) homogeneity, which will com-
Water signal reference ⫺ ⫹ ⫹ 0 0
plicate shimming (adjustment of the ho-
Principle of reciprocity 0 ⫹ ⫺ ⫺ ⫹
mogeneity of the local magnetic field)
Note.—⫹ ⫽ Merit, ⫺ ⫽ serious drawback of the strategy, 0 ⫽ neutral aspect. and water suppression.

Includes preparation of phantoms and validation of the strategy. Replace-and-match method.—The
basic principle of the replace-and-match
strategy is to replace the human subject
with a phantom that simulates (human)
Figure 1 tissue as closely as possible and to
match the coil load to the load obtained
Figure 1: Schematic illustrates
experimental setup of calibration previously in vivo (Fig 1, C) (43,44).
strategies used for quantification Then, a calibration measurement is per-
of cerebral metabolite concentra- formed, which is identical with respect
tions. Left: Setup for brain exami- to all imager settings (eg, pulse se-
nation. Right: Calibration mea- quence and size and position of the
surement. A, Internal endogenous VOI). The phantom usually contains a
marker, water signal reference solution that mimics the electric con-
method, and principle of reciproc- ductivity of human tissue. Fine adjust-
ity. B, External reference method. ment of the coil load can be made by
C, Replace-and-match method. placing the phantom further inside or
outside the coil or by inserting a small
bottle containing saline. It is important
that the load be accurately matched, be-
cause it is usually not possible to elimi-
nate the effect of a difference in load by
using a post hoc correction factor (44).
Unfortunately, the matching of the load
by maneuvering the phantom is not al-
ways straightforward and can be time
consuming. Furthermore, the B1 distri-
bution through a phantom is often con-

Radiology: Volume 240: Number 2—August 2006 321


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

Table 2
siderably different from that in the hu- strategy (also referred to as the phan-
man head. Therefore, simple matching of tom-replacement strategy) that is based Reduction of Metabolite Peak Area
the conductivity and overall load might be on this notion involves the determina- as Function of Echo Time
insufficient, because the human head has tion of the local B1 at the VOI, which is
Echo Time
structures of varying resistance that alter accomplished by measuring the voltage (msec) NAA* Macromolecules†
electric current paths. amplitude required to obtain the maxi-
Water signal reference method.— mum signal response (Vmax) (Fig 1, A). 20 4.6 33.0
Several research groups have used tis- Absolute standard units can then be ob- 30 6.9 45.1
sue water as an internal standard tained by dividing the recorded in vivo 50 11.2 63.2
(32,45–50). The in vivo measurement is MR signals by Vmax and by performing 130 26.5 92.6
performed first. Then, from the same additional calibration measurements 260 46.0 99.4
voxel, the signal from unsuppressed tis- with a solution of known concentration Note.—Data are percentage reduction in metabolite
sue water is recorded, which serves as (57–61). The principle of reciprocity ap- peak area with respect to hypothetic echo time of 0
an endogenous concentration reference plies only to transmit-receive coils. For msec.
* T2 relaxation time of 420 msec (32).
(Fig 1, A). It is best to record the water- MR systems with a receive-only coil, an †
T2 relaxation time of 50 msec (65).
suppressed spectra under conditions alternative approach based on the same
that are identical to those for the metab- principle has been designed in which
olite spectra. This can be achieved by the transmit-receive capabilities of the
detuning or switching off the water-sup- body coil are used (62). saturation, even though this increases
pression RF pulses. The use of pure ce- Institutional units.—In this quantifi- the duration of the MR acquisition.
rebrospinal fluid (recorded from a voxel cation approach, one of the previously The use of a short echo time (eg,
that contains only cerebrospinal fluid) mentioned absolute reference methods ⬍20 msec) minimizes signal losses
as a water reference to obtain molar is used, but the final calibration to stan- caused by T2 relaxation. At 1.5 T, typi-
concentrations is inadvisable, since it is dard units is not performed. Therefore, cal T2 relaxation times for several me-
usually impossible to position a voxel of this method shares the robustness of tabolites are 422 msec ⫾ 48 for NAA,
sufficiently large size in an area that the applied AQ strategy but is only use- 356 msec ⫾ 35 for choline, 214 msec ⫾
contains only cerebrospinal fluid (eg, ful for diagnostic comparison within one 23 for total creatine, and 200 msec ⫾ 20
ventricles). institution. for myo-inositol (32,51,64). The reduc-
Alternatively, the water content can tion of the NAA signal will be approxi-
be derived from proton-density– Important Considerations for AQ mately 21% if an echo time of 100 msec
weighted images (51,52). The obtained Echo time and repetition time.—When is used, while the signal loss will only be
metabolite concentrations, in molal the repetition time of a pulse sequence approximately 5% if an echo time of 20
units, can be converted into molar con- is relatively short (generally shorter msec is used (the effect of varying echo
centrations by using the density of brain than five times the longitudinal relax- times is indicated in Table 2). In con-
tissue (32). Because the determination ation time, T1), the magnetization can- trast, macromolecules (compounds with
of brain density and brain water content not totally recover before the next exci- a high effective molecular weight) have
can be quite elaborate, one usually ap- tation, which leads to a reduction of the a very short T2 relaxation time (⬍50
plies a value from the literature. How- signal (ie, signal saturation). msec at 2.1 T [65]), so the use of even
ever, one has to be cautious since sev- At 1.5 T, typical T1 relaxation times the shortest echo time leads to a large
eral pathologic conditions are known to (⫾ standard deviation) for several me- reduction in signal. For instance, an
affect the water content. For instance, it tabolites are 1430 msec ⫾ 165 for NAA, echo time of 20 msec will lead to a re-
has been determined for conditions 1330 msec ⫾ 199 for choline, 1460 duction in the macromolecule signal of
such as multiple sclerosis (53,54), brain msec ⫾ 270 for total creatine, and 1140 approximately 30%. On the other hand,
tumors (55), and hydrocephalus (55) msec ⫾ 308 for myo-inositol (32,63,64). spectra acquired with a long echo time
that the brain water content can be in- Given these values, a repetition time of (⬎150 msec) benefit from a less compli-
creased up to 12%, 50%, and 120%, 2000 msec will lead to a decrease of the cated appearance (mainly resonances
respectively, over the water content in NAA signal of approximately 25%, from uncoupled spins remain visible,
healthy subjects. whereas a repetition time of 7000 msec such as singlets from NAA, total creat-
Principle of reciprocity.—From the will cause a reduction of only 0.7%. Be- ine, and choline), improved water sup-
principle of reciprocity (55,56), it can cause the T1 values for most metabo- pression, and a flatter baseline (due to
be deduced that (if the coil is a com- lites are not the same, the acquired sig- signal reduction of components with
bined transmit-receive coil) the external nal intensity of each resonance should short T2, such as water and macromol-
voltage needed to produce a certain B1 be corrected separately for partial satu- ecules).
at a given location is inversely related to ration. Therefore, the use of a long rep- In all these cases, signal intensities
the voltage induced in the same RF coil etition time is advisable since it reduces of each resonance have to be properly
by a predefined B1. The calibration systematic errors caused by T1 signal corrected for T2 relaxation. However,

322 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

accurate determination of T1 and T2 is ute to the metabolite signal measured. It tization transfer effects can generally be
usually too time consuming to be per- is hard to determine the “true” concentra- avoided.
formed for every patient (66). Unfortu- tion level of a metabolite in a specific ho-
nately, one sometimes cannot resort to mogeneous tissue. For example, metabo-
using average values of a patient group, lite concentrations that are uncorrected Data Analysis
since relaxation times may be influ- for the contribution of cerebrospinal fluid In this section, the most important
enced by pathologic conditions and by are generally underestimated, since the methods of data analysis will be de-
the severity of a specific condition. For concentration of 1H MR spectroscopically scribed. The merits and drawbacks of
example, for diseases such as stroke detectable metabolites in cerebrospinal each analysis method are shown in Ta-
(67), amyotrophic lateral sclerosis (68), fluid is very low (70). Therefore, several ble 3. The actual quantification can be
low-grade glioma, and high-grade gli- tissue segmentation approaches have performed in either the time (76) or the
oma (69), the T2 relaxation times of been proposed, all of which rely on differ- frequency (77) domain. In the time do-
NAA can be reduced up to 42%, 22%, ences in relaxation properties as deter- main, the MR signal is represented as a
38%, and 43%, respectively. mined with a separate MR imaging mea- function of recording time, whereas in
Receiver gain instability.—In any surement. These methods include the use the frequency domain the signal is rep-
quantification strategy that requires an of calculated T1- or T2-weighted images resented as a function of resonance fre-
additional calibration measurement of a (17) or spectra in which the T2 decay of quency. The time-dependent signal can
phantom, this measurement should be water is measured as a function of echo be converted into its equivalent fre-
recorded with exactly the same ampli- time (50,71). However, since pathologic quency representation by applying a
fier gain settings as were used for the in conditions may affect relaxation times, Fourier transformation. In theory,
vivo measurement, to prevent system- one should always carefully interpret re- there are no differences between the
atic errors. Therefore, a calibration ex- sults from any segmentation procedure. two domains (78), but the data are al-
periment is preferably performed im- MR visibility.—Not all metabolite ways presented in the frequency do-
mediately after the in vivo experiment. molecules contribute equally to the MR main since this enables direct (visual)
However, if time restrictions prevent signal from a certain VOI. For example, interpretation.
this measurement, one has to monitor creatine has an invisible metabolite pool
the receiver stability on a regular basis, (2.5%) that is bound to macromolecular Integration
since it is not easily corrected for after- structures and, therefore, has low mo- The traditional procedure to determine
wards. An assessment of receiver gain bility; this results in very short T2 relax- the area of a certain peak in the fre-
stability can be performed by measuring ation times and thus broad resonances, quency domain is integration. The oper-
the background noise level in a region of which can be unobservable on conven- ator selects a frequency range, which
the spectrum “downfield” from the wa- tional MR spectra (72). A larger under- preferably contains only one peak, and
ter resonance, where no signals are ex- estimation (up to 10% for creatine [73] then performs numeric integration. Be-
pected (61). The background noise level and up to 30% for lactate [74]) of the cause only the total area under a reso-
should exhibit minimal variations if the true concentration can be caused by nance corresponds to the real peak in-
spectrometer has good long-term stabil- magnetization transfer effects if a wa- tensity and the contribution beyond the
ity (56). ter-suppression technique such as pre- lower and upper integration boundaries
Compartmentation.—Because in vivo saturation is used. However, most me- is neglected, the peak area will be un-
MR is performed on tissue at a macro- tabolites are not susceptible to magneti- derestimated (possibly by up to 40%
scopic level, various brain compartments zation transfer effects and show a signal [79]). Therefore, integration is an ade-
(gray matter, white matter, blood, cere- change only slightly above the limits of quate method only if the resonances are
brospinal fluid, or lesions) with different experimental error (75). Furthermore, well separated without any baseline
metabolite concentrations might contrib- in most measurement methods magne- fluctuations. Unfortunately, this is rarely
the case, since most in vivo spectra suf-
fer markedly from spectral overlap and
Table 3 baseline fluctuations. Therefore, the
Relative Merits and Drawbacks of Data Analysis Procedures area can hardly be attributed to a single
resonance, and, in addition, the base-
Preparation Sensitivity to Baseline Ease of
line will lead to an unknown contribu-
Procedure Time Imperfections Use Accuracy
tion.
Integration ⫹ ⫺ ⫹ ⫺
Peak fitting ⫹ ⫺ ⫹ ⫺ Peak Fitting
Peak fitting with prior knowledge 0 0 0 0 In this approach, all important peaks
Peak fitting with metabolite basis set ⫺ ⫹ ⫺ ⫹ are initially selected and coarse estima-
tions of the resonance frequency, line
Note.—⫹ ⫽ Merit, ⫺ ⫽ serious drawback, 0 ⫽ neutral aspect.
width, and peak intensity are per-

Radiology: Volume 240: Number 2—August 2006 323


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

formed, either by the operator or by an sequence. One has to take into account .uab.es/mrui/) (90), which includes the
algorithm. Subsequently, a fit is per- the type of imager, B0, pulse sequence, quantum mechanical simulation algo-
formed by using a least-squares optimi- echo time, and repetition time, as well rithm NMR-SCOPE (Laboratoire de
zation algorithm, which iteratively fits as the pH, ion composition, and temper- RMN, Universite Lyon I-CPE, Villeur-
all peaks to a line-shape model function, ature of the solution in which the indi- banne, France) (88); and the metabolite
so that the fitted spectrum resembles vidual metabolites are dissolved. In this basis set fitting routine QUEST (Labora-
the experimental spectrum as closely as way, the prior knowledge of each me- toire de RMN, Universite Lyon I-CPE)
possible (80). In general, this method tabolite—including chemical shifts, sig- (85) and the algorithm TDFDFit (Depart-
proves to be fairly robust with respect nal intensity (influenced by relaxation ment of MR Spectroscopy and Methodol-
to spectral overlap. However, if the ac- effects), amplitude ratios, splitting pat- ogy, University and Inselspital, Berne,
tual line shapes deviate substantially terns, and J evolution—matches the in Switzerland) (91). A detailed description
from Gaussian or lorentzian model func- vivo conditions. The in vivo MR spectra (92) and a critical assessment (7) of both
tions, the algorithm will not be able to fit are then analyzed as a linear combina- AMARES and LCModel can be found
the peaks accurately. To prevent this tion of the separately recorded in vitro elsewhere. Examples of LCModel output
problem, other line shapes have occa- spectra of the individual metabolites. and TDFDfit output are given in Figures 2
sionally been used, of which the Voigt There are two commonly used ap- and 3, respectively.
profile is the most common (81). How- proaches for obtaining a basis set: sim- One should note that the T1 and/or
ever, the use of a Voigt profile intro- ulation and in vitro measurement. In the T2 relaxation times are not always the
duces more degrees of freedom in the simulation approach (85,86), the re- same in the in vitro and the in vivo situ-
fitting procedure, which can lead to am- sponse is numerically simulated on the ations, and different resonances in the
biguous results. basis of molecular and quantum me- same metabolite might have different
chanical characteristics of each metabo- relaxation times. For example, the
Prior Knowledge lite (87,88). The in vitro approach (89) methylene protons of creatine, 2CH2,
This method allows the incorporation of requires that for each metabolite, a which resonate at around 3.90 ppm,
prior knowledge about the metabolites spectrum is acquired with exactly the display shorter T1 and T2 relaxation
that contribute to the 1H MR spectro- same conditions (eg, pulse sequence times (31% for T1 and 28% for T2,
scopic signal (82). All known signal pa- and timing parameters) as those used measured at 3 T) than do the methyl
rameters, such as relative frequencies, during the in vivo measurements. The protons of creatine, N(CH3), which res-
amplitude ratios, scalar coupling, and advantage of the simulation approach is onate at around 3.03 ppm (93). There-
phases of resonances that are charac- that, as long as the exact molecular fore, in pathologic conditions that are
teristics of a certain metabolite, can be structure is known, each metabolite can known to affect relaxation times, in
implemented as constraints in the fitting be included, whereas the in vitro ap- vitro prior knowledge cannot always be
routine. Prior knowledge is the only way proach requires carefully prepared me- applied to in vivo 1H MR spectra, which
of enhancing, by reducing the degrees of tabolite phantoms. The advantage of the may lead to systematic errors (see pre-
freedom, the accuracy of fitted model in vitro approach is that, as long as the vious discussion on echo time and repe-
parameters for a given data set. The phantoms are adequately prepared, the tition time in Important Considerations
calculation time is also reduced. An ex- response of the metabolites in the in for AQ). In that case, a separate relax-
ample of a method that enables incorpo- vitro measurement will be identical to ation measurement would be favorable.
ration of prior knowledge is the AM- the response of the metabolites in the in
ARES method (Department of Electrical vivo 1H MR spectroscopy examination. Important Considerations for Data
Engineering, Katholieke Univesiteit Leu- The high information content of 1H Analysis
ven, Leuven, Belgium) (83). MR spectra leads to an advantage regard- Quantification accuracy.—It is important
Advanced prior knowledge obtained ing the fitting accuracy, because overlap- to study and report the error estimates of
through metabolite basis set.—A de- ping resonances at one chemical shift po- the quantification method. Most of the
cade ago, algorithms were proposed sition can be directly related to other non- fitting routines present the so-called Cra-
(82,84) that implemented a strategy overlapping resonances from the same mer-Rao minimum variance bounds
where the maximum of prior knowledge metabolite. Currently used computer (CRMVBs), which reflect the theoretic
is used. Those methods were based on software includes the metabolite basis set standard of precision for the model pa-
the assumption that there are a limited fitting program LCModel (Stephen Prov- rameter estimates obtained from the data
number of 1H MR spectroscopically de- ender, PhD, http://s-provender.com (94). The parameter estimation must not
tectable metabolites present in the hu- /pages/lcmodel.shtml) (84), possibly in- contain systematic errors (eg, incorrect
man brain that had already been identi- corporating the molecular simulation prior knowledge), which may lead to un-
fied and analyzed in earlier studies. The library GAMMA (Department of Radiol- derestimation errors. It is important to
approach aimed to individually deter- ogy, University of California, San Fran- realize that the CRMVBs provide a mea-
mine the exact response of all metabo- cisco) (87); the quantification package sure of quality of the spectral fit and do
lites possibly present to a specific pulse jMRUI(TheMRUIProject,http://sermn02 not necessarily reflect the quality of the

324 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

original data. Furthermore, SNR degra- overlap completely with sharp metabo- Alternatively, a pure metabolite
dation and increases in line width, which lite resonances and, therefore, cannot spectrum can be obtained by subtract-
may lead to systematic errors, are not even be accounted for with conventional ing a macromolecules spectrum (simi-
1
necessarily reflected in CRMVB estimates H MR spectroscopy (65,96). Several larly obtained by exploiting the relax-
(95). methods can be used to separate the ation differences) from the acquired in
macromolecular baseline from the vivo spectrum (98). Unfortunately, in
Macromolecules wanted metabolite signal. vivo determination of the exact relax-
Macromolecules are compounds with a The best way to eliminate macromo- ation times for both macromolecules
relatively high molecular weight and in- lecular contamination is to use acquisi- and metabolites is complicated and time
clude proteins and polypeptides. Owing tion schemes that exploit the different consuming. Furthermore, neither me-
to their lower mobility, macromolecules relaxation properties of macromole- tabolites nor macromolecules necessar-
generally have a short T2 and are thus cules and small metabolites. Removal ily display a narrow distribution of re-
characterized by broad spectral lines on based on T1 relaxation can be achieved laxation times, which means that re-
1
H MR spectra. Macromolecules are the by so-called inversion-recovery (65) or moval based on relaxation times will not
main contributors to the fluctuating saturation-recovery (97) sequences (Fig always yield good results. During post-
baseline that generally hinders accurate 4), whereas removal based on T2 relax- processing, the macromolecular signal
quantification of small metabolites. ation can be obtained by increasing the can be estimated by using a spline or
Some macromolecular resonances even echo time (32). polynomial function, which can be sub-
tracted from the original spectrum to
improve the baseline (84). However,
Figure 2 since this approach relies on an estima-
tion, it is not flawless.
Recently, it has become popular to
include the macromolecules baseline in
the fitting routine (20,89,99). With the
LCModel approach, it is possible to ex-
tend the metabolite basis set with a
macromolecules spectrum. Since the
macromolecular contribution to in vivo
spectra cannot be simulated with simple
model solutions, one has to determine
the macromolecules baseline experi-
mentally. Both inversion recovery (89)
and saturation recovery (100) have
been used to obtain macromolecules
baseline spectra, which were averaged
over several subjects and then parame-
terized to be included in the basis set.
The incorporation of macromolecules
into the basis set generally improves
quantification accuracy and precision
(101).
Macromolecules themselves have
been the subject of recent research
(65,102). It has been reported that
several conditions, such as stroke
(103), brain tumors (101), and multi-
ple sclerosis (99), show an altered
Figure 2: 1H MR spectrum analyzed with LCModel (version 6.1-4) output of point-resolved spatially localized macromolecular profile. For example,
spectroscopy in a healthy adult subject. Spectra were recorded at 1.5 T (Intera; Philips Medical Systems, Best, the stroke (103) and multiple sclerosis
Netherlands) from the occipital lobe (repetition time msec/echo time msec, 2000/23; 128 signals acquired; voxel (99), respectively, can cause the signal
size, 8 cm3). In vivo spectrum (thin upper spectrum) was estimated with LCModel output (thick upper spectrum); the occurring from the macromolecules
difference between spectra is plotted at the bottom. Above the difference spectrum is the baseline spline estimate, resonance at 1.3 ppm to increase by
determined with LCModel. Inserted table at right displays estimated metabolite concentrations and Cramer-Rao
86% and 60%. Therefore, one should
minimum variance bounds. Cho ⫽ choline, Conc. ⫽ concentration, Cr ⫽ creatine, FWHM ⫽ full width at half
always carefully consider the incorpo-
maximum, Ins ⫽ myo-inositol, SD ⫽ standard deviation, tCr ⫽ total creatine.
ration of macromolecules spectra in

Radiology: Volume 240: Number 2—August 2006 325


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

Figure 3
the basis set when such a condition is
of concern.

Motion
Physiologic motion can have a large ef-
fect on 1H MR spectroscopic quantifica-
tion (104). For example, repeated small
body motions or pulsatile cerebral mo-
tion (eg, due to cardiac activity), can
substantially affect the characteristics of
the MR signal, even with motion-com-
pensated 1H MR spectroscopy se-
quences. These effects may include an
increase in line width, reduced peak in-
tensities, a diminished quality of water
suppression, or a doubling of all peaks.
Moreover, if quantification is based on
the summed signal of reference spectra
of the unsuppressed water peak, mo-
tion-induced phase effects can cause a
substantial overestimation of metabo-
lite concentrations, a problem that can
be circumvented by storing and phasing
all spectra individually (104). For chem-
ical shift imaging studies that include
subcutaneous lipid signals, which have
an enhanced sensitivity to subject mo-
tion, an in-plane motion correction can
be applied (105). The reduction of mo-
tion effects in 1H MR spectroscopy has
been achieved in several studies (eg, by
using cardiac triggering [104] or post-
processing [106]).

VOI Shape and Location


In the process of spectral quantification,
one should always keep in mind that the
true spatial voxel profile might deviate
from a perfect rectangular profile. The
voxel dimensions are, in principle, iden-
Figure 3: Fit results for point-resolved spatially localized 1H MR spectrum (3000/20; 128 signals ac-
tical for all resonances; however, the
quired; voxel size, 16 cm3) obtained at 1.5 T (Signa; GE Healthcare, Milwaukee, Wis) from white matter in
voxel position is dependent on the
centrum semiovale of an 18-year-old control subject. Parameterized fitting with metabolite basis set fitting
chemical shift of individual resonances.
algorithm TDFDfit was used. At top are experimental spectrum, fitted spectrum, and residuals. Below are
When a section-selective gradient is ap-
traces for the individual components that add up to the best fit (see reference 100 for definition of abbrevia-
plied to a volume containing metabolites tions). Some metabolites were split into two spectra, providing the freedom of differential T2 values for differ-
with different chemical shifts, there will ent protons in the same molecule. Baseline spectrum was obtained from a saturation recovery experiment.
be a displacement of the sensitive vol- (Reprinted, with permission, from reference 100.)
ume for each resonance of the metabo-
lite. The displacement is regulated by
the bandwidth of the RF pulse for voxel 1.22-mm displacement of the 2 voxels, creasing the strength of section-selec-
selection (38). For example, the methyl which results in an 82.8% overlap of the tive gradients. Obviously, safety mar-
protons of NAA and the methylene pro- 2 voxels. gins should not be exceeded. It should
tons of total creatine are separated by Chemical shift artifacts can be re- be noted that chemical shift imaging
1.90 ppm, or 122 Hz, at 1.5 T. Use of an duced by increasing the bandwidth of does not suffer from the chemical shift
RF bandwidth of 2000 Hz and a voxel the section-selective RF pulses and, to artifact for the spatial phase-encoding
size of 2 ⫻ 2 ⫻ 2 cm results in a keep the same voxel dimensions, by in- gradients, which are applied in the ab-

326 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

sence of RF pulses. However, if the duces time-varying eddy currents in the a high-pass filter (110) or an algorithm
chemical shift imaging study involves magnetic cryostat. As a result, the mag- such as the Hankel-Lanczos singular-
preselection of a VOI (eg, to exclude netic field is transiently distorted. Sev- values decomposition filter (111) prior
skull lipids) then the position of that eral methods have been developed to to data analysis (Fig 5).
VOI is again subject to the chemical correct irregular line shapes, including
shift artifact, while the chemical shift the QUALITY deconvolution (107) and
imaging voxels within the VOI are not eddy current correction procedure Sequence-Specific Issues
shifted with respect to each other. (108). These correction methods use a
Chemical shift imaging data are also in- single reference peak that was affected Spectral Editing
fluenced by the point-spread function, by the same distortions. The unsup- Lately, several clinical studies have been
which will be treated in the section enti- pressed water signal, acquired by using performed using the spectral editing ap-
tled Sequence-Specific Issues. the same spoiler gradients as the me- proach (112,113). Spectral editing en-
tabolite spectrum, is usually used for ables detection of metabolites that are
Line Shape and Eddy Currents that purpose. hard to detect with conventional 1H MR
Very often, the line shape of signals ob- spectroscopic techniques because these
tained from metabolites deviates sub- Water Suppression metabolites are present in lower con-
stantially from the ideal lorentzian line As indicated earlier, water-suppression centrations and their resonance peaks
shape. If left uncorrected, these line pulses might saturate parts of the me- overlap with those of other more abun-
shape deviations severely complicate tabolite spectrum and can thus influence dant metabolites (114). By using a spe-
peak fitting. Generally, line shape devia- the measured concentration of several cifically designed acquisition scheme
tions are caused by variations in B0, metabolites (a reduction of up to 21% that allows selective recording of signals
such as magnetic field inhomogeneities has been reported [109]). Numeric cal- only from desired metabolite(s), the in-
and eddy currents. Rapid switching of culations can be employed for accurate formation content of the acquired signal
magnetic field gradients (which occurs corrections for these effects (109). In is reduced. Preferably, only peaks of the
in every localization pulse sequence) in- addition, the residual water signal that desired metabolite(s) remain in the
is still present in water-suppressed spectrum, while the other metabolites
Figure 4 spectra can substantially degrade the are eliminated. Among the metabolites
performance of metabolite fitting rou- that are the target of the spectral editing
tines. Therefore, it is advisable to elimi- approach are ␥-aminobutyric acid, glu-
nate the unwanted water signal by using tamine, lactate, and glutathione.
It is important that the pulse se-
quence be carefully optimized and that
Figure 5
the specificity of the editing procedure
be critically assessed. If unwanted sig-
nals remain in the final spectrum, sim-
ple peak fitting will not suffice, although
one could resort to the advanced prior
knowledge approach (115).

Chemical Shift Imaging


Quantification of chemical shift imaging
data generally requires extra consider-
Figure 5: Water signal removal by means of ations, because chemical shift imaging
Hankel-Lanczos singular-values decomposition data can be different from single-voxel
filter. In vivo chemical shift imaging 1H MR spec- data in some respects. For example,
Figure 4: Point-resolved spatially localized 1H troscopy in the brain of a healthy adult. A, Region chemical shift imaging data usually have
MR spectra obtained at 1.5 T (Signa, GE Health- indicated by horizontal bar contains the water a lower SNR, since these imaging data
care) in a healthy adult. Spectra (6000/20; 128 peak. The peaks of interest, choline (Cho), total are subject to field inhomogeneities
signals acquired; voxel size, 12 cm3) were re- creatine (tCr), and NAA, are disturbed by right tail (due to the large VOI). In addition,
corded from the centrum semiovale. A, Metabo- of the water peak. B, Spectrum after subtraction of chemical shift imaging data are also sub-
lites-of-interest (MOI) spectrum. B, Macromole- water signal components, as retrieved and quanti- ject to artifacts such as imperfect water-
cules-only (MM) spectrum. C, Sum spectra calcu- fied with the Hankel-Lanczos singular-values suppression and lipid-contamination ar-
lated from series of saturation-recovery spectra. decomposition filter in region of the horizontal bar tifacts. On the other hand, the spatial
Cho ⫽ choline, Ins ⫽ myo-inositol, tCr ⫽ total in A. Components outside horizontal bar region
information of chemical shift imaging of-
creatine. (Reprinted, with permission, from refer- are not affected by the procedure. (Reprinted, with
fers several opportunities to improve
ence 97.) permission, from reference 111.)
postprocessing and analysis. Knowl-

Radiology: Volume 240: Number 2—August 2006 327


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

edge of the spatial dependence of pa- outer volume suppression, are used to ity values for the AQ of NAA, total crea-
rameters may yield extra information prevent lipid signals from contaminating tine, choline, and myo-inositol have
with regard to prior knowledge or con- brain spectra. been published for single-voxel spec-
straints. For instance, the notion that Other methods reduce lipid contam- troscopy and range from 7% to 16%
voxel-to-voxel variations in metabolite ination during postacquisition process- (27,100); for chemical shift imaging, re-
concentrations can be expected to ing (122). In an AQ strategy, coregis- producibility values range from 5% to
change smoothly can be used to set con- tered MR images are generally used for 21% (123,124). Therefore, to detect an
straints on the limits of these variations tissue segmentation (T1- or T2-weighted abnormality, the deviation from normal
(116). MR imaging [17]) or for water referenc- values should be larger than the repro-
Not all AQ strategies are necessarily ing (proton-density–weighted MR imag- ducibility of that particular measure-
compatible with chemical shift imaging ing [40,52]). It is important to note that ment.
data. For example, it is often too time apodization filters, used to improve the
consuming to obtain fully relaxed water point-spread function of the metabolite SNR Considerations
reference spectra with the same resolu- spectra, should also be applied to the The SNR can be used as a measure in
tion (117). A commonly used referenc- MR imaging data that are used for the the rejection of spectra (eg, the SNR
ing scheme for chemical shift imaging is segmentation procedure. should generally be at least 4). The SNR
the time-efficient reciprocity principle Fully automated spectral analysis.— is usually defined in the frequency do-
(21,61). An absolute reference can also Analysis of spectra from large chemical main as the ratio of the highest metabo-
be obtained by means of proton-den- shift imaging data sets is time consum- lite peak intensity to the standard devi-
sity–weighted MR imaging (40). ing, and because usually only spectra ation of the noise amplitude in a metab-
Point-spread function.—Quantifica- from a few selected voxels are analyzed, olite-free part of the spectrum (118).
tion of chemical shift imaging data is the complete data set is often underuti- The SNR can be improved by increasing
complicated by the point-spread func- lized. Therefore, automated methods the size of the VOI and the number of
tion, which describes the spread of sig- have been developed (87,116,117) that signal averages acquired. However, an
nal from one voxel to surrounding vox- enable analysis of large numbers of increase in the size of the VOI generally
els. The point-spread function origi- spectra and that are relatively insensi- degrades the line shape, increases the
nates from Fourier transformation of a tive to the low SNR and spectral distor- line width, and may decrease the sensi-
signal sampled in the time domain at a tions commonly associated with in vivo tivity to detect abnormalities, whereas
limited number of discrete points. Al- chemical shift imaging. Furthermore, an increase in the number of signals
though the point-spread function influ- the introduction of a standardized pro- acquired adds to the examination time.
ences all Fourier-transformed data, in cessing and analysis protocol makes
chemical shift imaging the effect may comparisons between different studies Line Width
become prominent since chemical shift possible. The line width is commonly defined as
imaging data are generally acquired the full width of the peak at half its
with a relatively low spatial resolution, maximum height, or full width at half
corresponding to a small number of Quality Assessment maximum, in the frequency domain
phase-encoding increments. Generally speaking, spectra with a large (118). A small line width generally im-
The point-spread function, and line width, low SNR, or obvious artifacts plies a high spectral resolution, which
therefore the spatial resolution, can be (ie, signal contributions from outside will improve the quality of the fitting
improved by applying apodization func- the VOI, phase distortions, and techni- routines. Better shimming, the use of a
tions in the spatial frequency or k-space cal failures) should be discarded. In ad- smaller VOI, and the choice of a VOI at
domain (118). One approach is to apply dition, when studying certain pathologic a sufficient distance from tissue inter-
different k-space sampling schemes to conditions, it is important to establish faces can improve the resolution. It has
improve the point-spread function and the limits of metabolite concentrations been recommended that the full width
the SNR (119,120). Another approach that represent abnormality. To obtain at half maximum value should always be
is k-space weighting during signal acqui- reliable absolute concentrations for the treated as a covariate in the statistical
sition, which also yields a better SNR assessment of pathologic changes, it is analysis, to help correct for single-sub-
(121). In chemical shift imaging of the necessary to take both the data acquisi- ject intersession variations (27).
brain, the point-spread function effect tion methods and the data processing
will typically lead to contamination of 1H methods into account. Overall repro- Criteria for Rejection of Data
MR spectra of voxels within the brain by ducibility depends on the variability at In a recent insightful review, Kreis
intense extracranial lipid signals. k-Space immediate repetition of an acquisition, (125) proposed the following accep-
weighting or application of apodization intraindividual variability at reexamina- tance criteria for spectral data and their
functions to improve the point-spread tion of the same subject at a subsequent fitting results: (a) The full width at half
function is usually insufficient in this acquisition, and interindividual variabil- maximum of the metabolites should be
case; therefore, other methods, such as ity. The achievable overall reproducibil- less than 0.1 ppm; (b) the Cramer-Rao

328 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

minimum variance bound should be matter and cerebral metabolite changes in Bartlett PA, Barker GJ, Duncan JS. A
smaller than 50%; (c) the fitting residual children undergoing treatment for acute short-echo-time proton magnetic reso-
lymphoblastic leukemia: longitudinal study nance spectroscopic imaging study of tem-
should not contain unexplained fea-
with MR imaging and 1H MR spectroscopy. poral lobe epilepsy. Epilepsia 2002;43:
tures; and (d) the spectra should not Radiology 2003;229:659 – 669. 1021–1031.
contain artifacts (eg, doubled peaks or
4. Vermathen P, Laxer KD, Schuff N, Matson 16. Fernando KT, McLean MA, Chard DT, et
asymmetric line shapes).
GB, Weiner MW. Evidence of neuronal in- al. Elevated white matter myo-inositol in
jury outside the medial temporal lobe in clinically isolated syndromes suggestive of
temporal lobe epilepsy: N-acetylaspartate multiple sclerosis. Brain 2004;127:1361–
Conclusions and Future Perspectives
concentration reductions detected with 1369.
The implementation of AQ in a clinical multisection proton MR spectroscopic im-
aging—initial experience. Radiology 2003; 17. Hetherington HP, Pan JW, Mason GF, et al.
routine has been greatly facilitated by Quantitative 1H spectroscopic imaging of
226:195–202.
the development of calibration strate- human brain at 4.1 T using image segmen-
gies and the availability of spectral fit- 5. Rai GS, McConnell JR, Waldman A, Grant tation. Magn Reson Med 1996;36:21–29.
ting routines. This is beneficial progress D, Chaudry M. Brain proton spectroscopy
in dementia: an aid to clinical diagnosis. 18. Auer DP, Schirmer T, Heidenreich JO,
because AQ has an added value for 1H Herzog J, Putz B, Dichgans M. Altered
Lancet 1999;353:1063–1064.
MR spectroscopic quantification. Fur- white and gray matter metabolism in
thermore, the introduction of phased- 6. Ruiz-Pena JL, Pinero P, Sellers G, et al. CADASIL: a proton MR spectroscopy and
array coils (126) and higher (⬎1.5-T) Magnetic resonance spectroscopy of nor- 1H-MRSI study. Neurology 2001;56:635–
mal appearing white matter in early relaps- 642.
field strengths (127) has been proved to ing-remitting multiple sclerosis: correla-
be advantageous with regard to SNR, tions between disability and spectroscopy. 19. Pohl C, Block W, Karitzky J, et al. Proton
quantification precision, reproducibil- BMC Neurol 2004;4:8. magnetic resonance spectroscopy of the
ity, and detection sensitivity. To obtain motor cortex in 70 patients with amyotro-
7. Kanowski M, Kaufmann J, Braun J, Bernar- phic lateral sclerosis. Arch Neurol 2001;58:
reliable absolute concentrations, how- ding J, Tempelmann C. Quantitation of sim- 729 –735.
ever, one has to consider potential com- ulated short echo time 1H human brain
plicating factors, with respect to both spectra by LCModel and AMARES. Magn 20. Auer DP, Gossl C, Schirmer T, Czisch M.
Reson Med 2004;51:904 –912. Improved analysis of 1H-MR spectra in the
the data acquisition method and the
presence of mobile lipids. Magn Reson Med
data processing method. For example, 8. Kreis R. Quantitative localized (1)H MR 2001;46:615– 618.
relaxation effects in data acquisition can spectroscopy for clinical use. J Prog NMR
be either corrected for or eliminated, 1997;31:155–195. 21. McLean MA, Woermann FG, Barker GJ,
Duncan JS. Quantitative analysis of short
whereas data fitting is complicated by 9. Cady E. Determination of absolute concen- echo time (1)H-MRSI of cerebral gray and
factors such as contribution of macro- trations of metabolites from NMR spectra. white matter. Magn Reson Med 2000;44:
molecules. Nevertheless, most of these NMR Basic Princ Prog 1992;26:249 –281. 401– 411.
problems have been critically addressed 10. Henriksen O. In vivo quantitation of metab- 22. Connelly A, Jackson GD, Duncan JS, King
and can be taken into account in a satis- olite concentrations in the brain by means MD, Gadian DG. Magnetic resonance spec-
fying manner. AQ is available and can of proton MRS. NMR Biomed 1995;8:139 – troscopy in temporal lobe epilepsy. Neurol-
improve the diagnostic utility of 1H MR 148. ogy 1994;44:1411–1417.
spectroscopy procedures. Therefore, 11. Tofts PS, Wray S. A critical assessment of
23. Lundbom N, Gaily E, Vuori K, et al. Proton
further progress in the development of methods of measuring metabolite concen-
spectroscopic imaging shows abnormalities
automated spectral analysis methods trations by NMR spectroscopy. NMR
in glial and neuronal cell pools in frontal
Biomed 1988;1:1–10.
and databases of normal regional and lobe epilepsy. Epilepsia 2001;42:1507–
age-dependent metabolite concentra- 12. Stoppe G, Bruhn H, Pouwels PJ, Hanicke 1514.
tions has to be encouraged to make the W, Frahm J. Alzheimer disease: absolute
24. Mathews VP, Barker PB, Blackband SJ,
quantification of cerebral metabolites in
AQ procedures more easily applicable Chatham JC, Bryan RN. Cerebral metabo-
vivo using localized proton magnetic reso-
in clinical routine. lites in patients with acute and subacute
nance spectroscopy. Alzheimer Dis Assoc
strokes: concentrations determined by
Disord 2000;14:112–119.
quantitative proton MR spectroscopy. AJR
13. Mueller SG, Laxer KD, Suhy J, Lopez RC, Am J Roentgenol 1995;165:633– 638.
References Flenniken DL, Weiner MW. Spectroscopic
25. Chang L, Ernst T, Tornatore C, et al. Me-
1. Ross B, Bluml S. Magnetic resonance spec- metabolic abnormalities in mTLE with and
tabolite abnormalities in progressive multi-
troscopy of the human brain. Anat Rec without MRI evidence for mesial temporal
focal leukoencephalopathy by proton mag-
2001;265:54 – 84. sclerosis using hippocampal short-TE
netic resonance spectroscopy. Neurology
MRSI. Epilepsia 2003;44:977–980.
2. Alger JR, Cloughesy TF. Structural and 1997;48:836 – 845.
functional imaging of cerebral neoplasia. In: 14. Savic I, Osterman Y, Helms G. MRS shows
26. Li BS, Wang H, Gonen O. Metabolite ratios
Mazziotta JC, Toga AW, Frackowiak RSJ, syndrome differentiated metabolite changes
to assumed stable creatine level may con-
eds. Brain mapping: the disorders. San Di- in human-generalized epilepsies. Neuroim-
found the quantification of proton brain MR
ego, Calif: Academic Press, 2000. age 2004;21:163–172.
spectroscopy. Magn Reson Imaging 2003;
3. Chu WC, Chik KW, Chan YL, et al. White 15. Simister RJ, Woermann FG, McLean MA, 21:923–928.

Radiology: Volume 240: Number 2—August 2006 329


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

27. Schirmer T, Auer DP. On the reliability of DesPres DJ, Armstrong MR. Absolute centrations in the frontal lobe. Magn Reson
quantitative clinical magnetic resonance quantitation of short TE brain 1H-MR spec- Med 1999;41:883– 888.
spectroscopy of the human brain. NMR tra and spectroscopic imaging data. J Com-
Biomed 2000;13:28 –36. put Assist Tomogr 1993;17:191–199. 52. Schuff N, Ezekiel F, Gamst AC, et al. Re-
gion and tissue differences of metabolites in
28. Bottomley PA, inventor; General Electric, 41. Webb P, Spielman D, Macovski A. Inhomo- normally aged brain using multislice 1H
assignee. Selective volume method for per- geneity correction for in vivo spectroscopy magnetic resonance spectroscopic imaging.
forming localized NMR spectroscopy. U.S. by high-resolution water referencing. Magn Magn Reson Med 2001;45:899 –907.
patent 4,480,228 0ctober 30, 1984. Reson Med 1992;23:1–11.
53. Laule C, Vavasour IM, Moore GR, et al.
29. Frahm J, Merboldt KD, Hanicke W. Local- 42. Li S, Williams GD, Frisk TA, Arnold BW, Water content and myelin water fraction in
ized proton spectroscopy using stimulated Smith MB. A computer simulation of the multiple sclerosis: a T2 relaxation study.
echoes. J Magn Reson 1987;72:502–508. static magnetic field distribution in the hu- J Neurol 2004;251:284 –293.
30. Brown TR, Kincaid BM, Ugurbil K. NMR man head. Magn Reson Med 1995;34:268 –
54. Helms G. Volume correction for edema in
chemical shift imaging in three dimensions. 275.
single-volume proton MR spectroscopy of
Proc Natl Acad Sci U S A 1982;79:3523– 43. Buchli R, Boesiger P. Comparison of meth- contrast-enhancing multiple sclerosis le-
3526. ods for the determination of absolute me- sions. Magn Reson Med 2001;46:256 –263.
31. Maudsley AA, Hilal SK, Perman WH, Si- tabolite concentrations in human muscles
55. Grasso G, Alafaci C, Passalacqua M, et al.
mon HE. Spatially resolved high resolution by 31P MRS. Magn Reson Med 1993;30:
Assessment of human brain water content
spectroscopy by “four-dimensional” NMR. J 552–558.
by cerebral bioelectrical impedance analysis:
Magn Reson 1983;51:147–152. a new technique and its application to cere-
44. Duc CO, Weber OM, Trabesinger AH,
32. Kreis R, Ernst T, Ross BD. Absolute quan- Meier D, Boesiger P. Quantitative 1H MRS bral pathological conditions. Neurosurgery
titation of water and metabolites in the hu- of the human brain in vivo based on the 2002;50:1064 –1072.
man brain. II. Metabolite concentrations. J stimulation phantom calibration strategy.
56. Hoult DI, Richards RE. The signal-to-noise
Magn Reson B 1993;102:9 –19. Magn Reson Med 1998;39:491– 496.
ratio of the nuclear magnetic resonance ex-
33. Fatouros PP, Heath DL, Beaumont A, et al. 45. Christiansen P, Henriksen O, Stubgaard M, periment. J Magn Reson 1976;24:71– 85.
Comparison of NAA measures by MRS and Gideon P, Larsson HB. In vivo quantifica- 57. Helms G. A precise and user-independent
HPLC. Acta Neurochir Suppl 2000;76:35– tion of brain metabolites by 1H-MRS using quantification technique for regional com-
37. water as an internal standard. Magn Reson parison of single volume proton MR spec-
Imaging 1993;11:107–118. troscopy of the human brain. NMR Biomed
34. Huppi PS, Fusch C, Boesch C, et al. Re-
gional metabolic assessment of human 46. Danielsen ER, Henriksen O. Absolute 2000;13:398 – 406.
brain during development by proton mag- quantitative proton NMR spectroscopy 58. Kreis R, Slotboom J, Pietz J, Jung B, Boe-
netic resonance spectroscopy in vivo and by based on the amplitude of the local water sch C. Quantitation of localized (31)P mag-
high-performance liquid chromatography/ suppression pulse: quantification of brain netic resonance spectra based on the reci-
gas chromatography in autopsy tissue. Pe- water and metabolites. NMR Biomed 1994; procity principle. J Magn Reson 2001;149:
diatr Res 1995;37:145–150. 7:311–318. 245–250.
35. Bottomley PA, Weiss RG. Noninvasive lo- 47. Knight-Scott J, Haley AP, Rossmiller SR, et 59. Kreis R, Hofmann L, Kuhlmann B, Boesch
calized MR quantification of creatine kinase al. Molality as a unit of measure for ex- C, Bossi E, Huppi PS. Brain metabolite
metabolites in normal and infarcted canine pressing 1H MRS brain metabolite concen- composition during early human brain de-
myocardium. Radiology 2001;219:411– 418. trations in vivo. Magn Reson Imaging 2003; velopment as measured by quantitative in
21:787–797. vivo 1H magnetic resonance spectroscopy.
36. Battistuta J, Bjartmar C, Trapp BD. Post-
mortem degradation of N-acetyl aspartate Magn Reson Med 2002;48:949 –958.
48. Thulborn KT, Ackerman JH. Absolute mo-
and N-acetyl aspartylglutamate: an HPLC lar concentrations by NMR in inhomoge- 60. Michaelis T, Merboldt KD, Bruhn H, Han-
analysis of different rat CNS regions. Neu- neous B1: a scheme for analysis of in vivo icke W, Frahm J. Absolute concentrations
rochem Res 2001;26:695–702. metabolites. J Magn Reson 1983;55:357– of metabolites in the adult human brain in
37. Burri R, Bigler P, Straehl P, Posse S, Co- 371. vivo: quantification of localized proton MR
lombo JP, Herschkowitz N. Brain develop- spectra. Radiology 1993;187:219 –227.
49. Barker PB, Soher BJ, Blackband SJ,
ment: 1H magnetic resonance spectroscopy Chatham JC, Mathews VP, Bryan RN. 61. Soher BJ, van Zijl PC, Duyn JH, Barker PB.
of rat brain extracts compared with chro- Quantitation of proton NMR spectra of the Quantitative proton MR spectroscopic im-
matographic methods. Neurochem Res human brain using tissue water as an inter- aging of the human brain. Magn Reson Med
1990;15:1009 –1016. nal concentration reference. NMR Biomed 1996;35:356 –363.
38. de Graaf RA, Rothman D. In vivo detection 1993;6:89 –94.
62. Jost G, Harting I, Heiland S. Quantitative
and quantification of scalar coupled (1)H
50. Ernst T, Kreis R, Ross BD. Absolute quan- single-voxel spectroscopy: the reciprocity
NMR resonances. Concepts Magn Reson
titation of water and metabolites in the hu- principle for receive-only head coils. J
2001;13:32–76.
man brain. I. Compartments and water. J Magn Reson Imaging 2005;21:66 –71.
39. Roth K, Hubesch B, Meyerhoff DJ, et al. Magn Reson B 1993;102:1– 8.
63. Ethofer T, Mader I, Seeger U, et al. Com-
Noninvasive quantitation of phosphorus
51. Brooks JC, Roberts N, Kemp GJ, Martin parison of longitudinal metabolite relax-
metabolites in human-tissue by NMR-spec-
PA, Whitehouse GH. Magnetic resonance ation times in different regions of the hu-
troscopy. J Magn Reson 1989;81:299 –311.
imaging-based compartmentation and its man brain at 1.5 and 3 tesla. Magn Reson
40. Alger JR, Symko SC, Bizzi A, Posse S, application to measuring metabolite con- Med 2003;50:1296 –1301.

330 Radiology: Volume 240: Number 2—August 2006


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

64. Rutgers DR, Kingsley PB, van der Grond J. 76. Vanhamme L, Sundin T, Hecke PV, Huffel ter R. Toward an in vivo neurochemical
Saturation-corrected T 1 and T 2 relaxation SV. MR spectroscopy quantitation: a re- profile: quantification of 18 metabolites in
times of choline, creatine and N-acetyl as- view of time-domain methods. NMR short-echo-time (1)H NMR spectra of the
partate in human cerebral white matter at Biomed 2001;14:233–246. rat brain. J Magn Reson 1999;141:104 –
1.5 T. NMR Biomed 2003;16:286 –288. 120.
77. Mierisova S, Ala-Korpela M. MR spectros-
65. Behar KL, Rothman DL, Spencer DD, copy quantitation: a review of frequency 90. Naressi A, Couturier C, Devos JM, et al.
Petroff OA. Analysis of macromolecule res- domain methods. NMR Biomed 2001;14: Java-based graphical user interface for the
onances in 1H NMR spectra of human 247–259. MRUI quantitation package. MAGMA
brain. Magn Reson Med 1994;32:294 –302. 2001;12:141–152.
78. Abildgaard F, Gesmar H, Led JJ. Quantita-
66. Kreis R, Slotboom J, Hofmann L, Boesch C. tive-analysis of complicated nonideal Fou- 91. Slotboom J, Boesch C, Kreis R. Versatile
Integrated data acquisition and processing rier-transform NMR-spectra. J Magn Re- frequency domain fitting using time domain
to determine metabolite contents, relax- son 1988;79:78 – 89. models and prior knowledge. Magn Reson
ation times, and macromolecule baseline in Med 1998;39:899 –911.
79. Meyer RA, Fisher MJ, Nelson SJ, Brown
single examinations of individual subjects.
TR. Evaluation of manual methods for inte- 92. in ⬘t Zandt H, van Der Graaf M, Heerschap
Magn Reson Med 2005;54:761–768.
gration of in vivo phosphorus NMR spectra. A. Common processing of in vivo MR spec-
67. Walker PM, Ben Salem D, Lalande A, Gir- NMR Biomed 1988;1:131–135. tra. NMR Biomed 2001;14:224 –232.
oud M, Brunotte F. Time course of NAA T2
80. Nelson SJ, Brown TR. A method for auto- 93. Traber F, Block W, Lamerichs R, Gieseke
and ADC(w) in ischaemic stroke patients:
matic quantification of one-dimensional J, Schild HH. 1H metabolite relaxation
1H MRS imaging and diffusion-weighted
spectra with low signal-to-noise ratio. J times at 3.0 tesla: measurements of T1 and
MRI. J Neurol Sci 2004;220:23–28.
Magn Reson 1987;75:229 –243. T2 values in normal brain and determina-
68. Hanstock CC, Cwik VA, Martin WR. Re- tion of regional differences in transverse
81. Marshall I, Higinbotham J, Bruce S, Freise
duction in metabolite transverse relaxation relaxation. J Magn Reson Imaging 2004;19:
A. Use of Voigt lineshape for quantification
times in amyotrophic lateral sclerosis. 537–545.
of in vivo 1H spectra. Magn Reson Med
J Neurol Sci 2002;198:37– 41.
1997;37:651– 657. 94. Cavassila S, Deval S, Huegen C, van Or-
69. Isobe T, Matsumura A, Anno I, et al. Quan- mondt D, Graveron-Demilly D. Cramer-
82. de Graaf AA, Bovee WM. Improved quanti-
tification of cerebral metabolites in glioma Rao bounds: an evaluation tool for quantita-
fication of in vivo 1H NMR spectra by opti-
patients with proton MR spectroscopy us- tion. NMR Biomed 2001;14:278 –283.
mization of signal acquisition and process-
ing T2 relaxation time correction. Magn Re-
ing and by incorporation of prior knowl- 95. Kreis R, Boesch C. Bad spectra can be bet-
son Imaging 2002;20:343–349.
edge into the spectral fitting. Magn Reson ter than good spectra [abstr]. In: Proceed-
70. Lynch J, Peeling J, Auty A, Sutherland GR. Med 1990;15:305–319. ings of the 11th Meeting of the International
Nuclear magnetic resonance study of cere- Society for Magnetic Resonance in Medi-
83. Vanhamme L, van den Boogaart A, Van
brospinal fluid from patients with multiple cine. Berkeley, Calif: International Society
Huffel S. Improved method for accurate
sclerosis. Can J Neurol Sci 1993;20:194 – for Magnetic Resonance in Medicine, 2003;
and efficient quantification of MRS data
198. 264.
with use of prior knowledge. J Magn Reson
71. Helms G. T2-based segmentation of 1997;129:35– 43. 96. Behar KL, Ogino T. Characterization of
periventricular paragraph sign volumes for macromolecule resonances in the 1H NMR
84. Provencher SW. Estimation of metabolite
quantification of proton magnetic para- spectrum of rat brain. Magn Reson Med
concentrations from localized in vivo pro-
graph sign resonance spectra of multiple 1993;30:38 – 44.
ton NMR spectra. Magn Reson Med 1993;
sclerosis lesions. MAGMA 2003;16:10 –16.
30:672– 679. 97. Hofmann L, Slotboom J, Boesch C, Kreis R.
72. Kruiskamp MJ, de Graaf RA, van Vliet G, Characterization of the macromolecule
85. Ratiney H, Coenradie Y, Cavassila S, van
Nicolay K. Magnetic coupling of creatine/ baseline in localized (1)H-MR spectra of hu-
Ormondt D, Graveron-Demilly D. Time-do-
phosphocreatine protons in rat skeletal man brain. Magn Reson Med 2001;46:855–
main quantitation of 1H short echo-time
muscle, as studied by (1)H-magnetization 863.
signals: background accommodation. MAGMA
transfer MRS. Magn Reson Med 1999;42:
2004;16:284–296. 98. Kassem MN, Bartha R. Quantitative proton
665– 672.
short-echo-time LASER spectroscopy of
86. Govindaraju V, Young K, Maudsley AA.
73. Kruiskamp MJ, de Graaf RA, van der normal human white matter and hippocam-
Proton NMR chemical shifts and coupling
Grond J, Lamerichs R, Nicolay K. Magnetic pus at 4 tesla incorporating macromolecule
constants for brain metabolites. NMR
coupling between water and creatine pro- subtraction. Magn Reson Med 2003;49:
Biomed 2000;13:129 –153.
tons in human brain and skeletal muscle, as 918 –927.
measured using inversion transfer (1)H- 87. Young K, Govindaraju V, Soher BJ, Mauds-
99. Mader I, Seeger U, Weissert R, et al. Pro-
MRS. NMR Biomed 2001;14:1– 4. ley AA. Automated spectral analysis I: for-
ton MR spectroscopy with metabolite-null-
mation of a priori information by spectral
74. Kotitschke K, Schnackerz KD, Dringen R, ing reveals elevated macromolecules in
simulation. Magn Reson Med 1998;40:812–
Bogdahn U, Haase A, von Kienlin M. Inves- acute multiple sclerosis. Brain 2001;124:
815.
tigation of the 1H NMR visibility of lactate 953–961.
in different rat and human brain cells. NMR 88. Graveron-Demilly D, Diop A, Briguet A,
100. Hofmann L, Slotboom J, Jung B, Maloca P,
Biomed 1994;7:349 –355. Fenet B. Product-operator algebra for
Boesch C, Kreis R. Quantitative 1H-mag-
strongly coupled spin systems. J Magn Re-
75. Leibfritz D, Dreher W. Magnetization netic resonance spectroscopy of human
son A 1993;101:233–239.
transfer MRS. NMR Biomed 2001;14:65– brain: influence of composition and param-
76. 89. Pfeuffer J, Tkac I, Provencher SW, Gruet- eterization of the basis set in linear combi-

Radiology: Volume 240: Number 2—August 2006 331


REVIEW: 1H MR Spectroscopy of the Brain Jansen et al

nation model-fitting. Magn Reson Med 110. Marion D, Ikura M, Bax A. Improved sol- schemes for multidimensional MR spectro-
2002;48:440 – 453. vent suppression in one- and two-dimen- scopic imaging. Magn Reson Med 1996;36:
sional NMR spectra by convolution of time- 469 – 473.
101. Seeger U, Klose U, Mader I, Grodd W,
domain data. J Magn Reson 1989;84:425–
Nagele T. Parameterized evaluation of mac- 120. Maudsley AA, Matson GB, Hugg JW,
430.
romolecules and lipids in proton MR spec- Weiner MW. Reduced phase encoding in
troscopy of brain diseases. Magn Reson 111. de Beer R, van Ormondt D. Analysis of spectroscopic imaging. Magn Reson Med
Med 2003;49:19 –28. NMR data using time domain fitting proce- 1994;31:645– 651.
dures. NMR Basic Princ Prog 1992;26:201–
102. Mader I, Seeger U, Karitzky J, Erb M, 121. Kuhn B, Dreher W, Norris DG, Leibfritz D.
248.
Schick F, Klose U. Proton magnetic reso- Fast proton spectroscopic imaging employ-
nance spectroscopy with metabolite nulling 112. Chang L, Cloak CC, Ernst T. Magnetic res- ing k-space weighting achieved by variable
reveals regional differences of macromole- onance spectroscopy studies of GABA in repetition times. Magn Reson Med 1996;
cules in normal human brain. J Magn Reson neuropsychiatric disorders. J Clin Psychia- 35:457– 464.
Imaging 2002;16:538 –546. try 2003;64(suppl 3):7–14.
122. Haupt CI, Schuff N, Weiner MW, Maudsley
103. Graham GD, Hwang JH, Rothman DL, Pri- 113. Mueller SG, Trabesinger AH, Boesiger P,
AA. Removal of lipid artifacts in 1H spec-
chard JW. Spectroscopic assessment of al- Wieser HG. Brain glutathione levels in pa-
troscopic imaging by data extrapolation.
terations in macromolecule and small-mol- tients with epilepsy measured by in vivo
Magn Reson Med 1996;35:678 – 687.
ecule metabolites in human brain after (1)H-MRS. Neurology 2001;57:1422–1427.
stroke. Stroke 2001;32:2797–2802. 123. Chard DT, McLean MA, Parker GJ, Mac-
114. Trabesinger AH, Meier D, Boesiger P. In
104. Felblinger J, Kreis R, Boesch C. Effects of vivo 1H NMR spectroscopy of individual hu- Manus DG, Miller DH. Reproducibility of in
physiologic motion of the human brain man brain metabolites at moderate field vivo metabolite quantification with proton
upon quantitative 1H-MRS: analysis and strengths. Magn Reson Imaging 2003;21: magnetic resonance spectroscopic imaging.
correction by retro-gating. NMR Biomed 1295–1302. J Magn Reson Imaging 2002;15:219 –225.
1998;11:107–114.
115. Terpstra M, Henry PG, Gruetter R. Mea- 124. Wiedermann D, Schuff N, Matson GB, et
105. Haupt CI, Kiefer AP, Maudsley AA. In- surement of reduced glutathione (GSH) in al. Short echo time multislice proton mag-
plane motion correction for MR spectro- human brain using LCModel analysis of dif- netic resonance spectroscopic imaging in
scopic imaging. Magn Reson Med 1998;39: ference-edited spectra. Magn Reson Med human brain: metabolite distributions and
749 –753. 2003;50:19 –23. reliability. Magn Reson Imaging 2001;19:
1073–1080.
106. Helms G, Piringer A. Restoration of mo- 116. Soher BJ, Young K, Govindaraju V, Mauds-
tion-related signal loss and line-shape dete- ley AA. Automated spectral analysis III: ap- 125. Kreis R. Issues of spectral quality in clinical
rioration of proton MR spectra using the plication to in vivo proton MR spectroscopy 1H-magnetic resonance spectroscopy and a
residual water as intrinsic reference. Magn and spectroscopic imaging. Magn Reson gallery of artifacts. NMR Biomed 2004;17:
Reson Med 2001;46:395– 400. Med 1998;40:822– 831. 361–381.
107. de Graaf AA, van Dijk JE, Bovee WM. 117. Ebel A, Soher BJ, Maudsley AA. Assess- 126. Natt O, Bezkorovaynyy V, Michaelis T,
QUALITY: quantification improvement by ment of 3D proton MR echo-planar spec- Frahm J. Use of phased array coils for a
converting lineshapes to the lorentzian troscopic imaging using automated spectral determination of absolute metabolite con-
type. Magn Reson Med 1990;13:343–357. analysis. Magn Reson Med 2001;46:1072– centrations. Magn Reson Med 2005;53:
1078. 3– 8.
108. Klose U. In vivo proton spectroscopy in
presence of eddy currents. Magn Reson 118. Ernst RR, Bodenhausen G, Wokaun A.
127. Srinivasan R, Vigneron D, Sailasuta N,
Med 1990;14:26 –30. Principles of nuclear magnetic resonance in
Hurd R, Nelson S. A comparative study of
one and two dimensions. Oxford, England:
109. Hsu AC, Gregory CD. Offset-dependent myo-inositol quantification using LCmodel
Oxford University Press, 1987.
partial saturation in binomial solvent sup- at 1.5 T and 3.0 T with 3 D 1H proton
pression sequences. J Magn Reson 1998; 119. Hugg JW, Maudsley AA, Weiner MW, Mat- spectroscopic imaging of the human brain.
131:46 –53. son GB. Comparison of k-space sampling Magn Reson Imaging 2004;22:523–528.

332 Radiology: Volume 240: Number 2—August 2006

You might also like