Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

JOURNAL OF MAGNETIC RESONANCE IMAGING 13:560 –567 (2001)

Original Research

Proton MR Spectroscopy in Clinical Routine


Isabella M. Burtscher, MD, PhD* and Stig Holtås, MD, PhD

In vivo magnetic resonance spectroscopy (MRS) addresses brain has by tradition, and based on the well-suited
metabolic pathways and their steady states in different MRS properties of brain tissue, been one of the organs
tissue types. The brain has by tradition, and due to tech- most studied by MRS. Furthermore, a number of dis-
nical limitations in other organs, been one of the tissues eases of the central nervous system (CNS) or systemic
most studied by MRS, and both 1H- and 31P-MRS have been
diseases showing CNS involvement can often not be
used. Although 31P-MRS is outstanding for the evaluation
diagnosed sufficiently by means of noninvasive imaging
of sources of metabolic energy in the brain, 1H-MRS has
become the major clinically applied method in neurospec- techniques, due to lack of or simply uncharacteristic
troscopy, as it provides information on markers of neuro- structural abnormalities. As the discussion of all or-
nal function, myelin, cell membranes, and metabolic ac- gans possibly evaluated by MRS in clinical routine
tive compounds. Furthermore, MR sensitivity is much would be beyond the scope of a single review, we focus
greater for protons than it is for phosphorus and 1H-MRS, on neurospectroscopy and diagnostic insights into di-
therefore allowing better spatial resolution. This review verse neurological problems provided by MRS applied
focuses on neurospectroscopy and diagnostic insights into as a clinical tool.
diverse neurological problems provided by 1H-MRS applied
as a clinical tool. J. Magn. Reson. Imaging 2001;13:
560 –567. © 2001 Wiley-Liss, Inc.
TECHNICAL BACKGROUND AND CHALLENGES
Index terms: proton magnetic resonance spectroscopy; brain;
disease; in vivo; human The results of an MRI examination are images repre-
senting the proton (1H) content of the examined tissue
INTRODUCED TO MEDICINE in the early 1980s, mag- displayed as slices of the body anatomy with a spatial
netic resonance (MR) has proven to be a valuable tool in resolution as low as approximately 1 mm3 and the sig-
the evaluation of anatomy, physiology, and biochemis- nal mainly arising from water and fat. In spectroscopy,
try of the human body. In contrast to MR imaging (MRI) MR signals corresponding to biochemical compounds
and MR angiography (MRA), both focusing on human investigated are presented as peaks (also called reso-
morphology, in vivo MR spectroscopy (MRS) addresses nances) in MR spectra illustrated in Fig. 1. Depending
metabolic pathways and their steady states in different on the addressed nucleus, different compounds can be
tissue types (1– 6). The fields of application for MRI detected. 1H, also used in MRI, and 31P are the most
expanded rapidly during the last decades and finally frequently used nuclei in clinical MRS; however, even
13
cover all major tissues and organs of the body. Al- C, 15N, 19F, and 23Na are assessable by MRS. Giving
though MRS can theoretically be applied to almost all insights into the energetic pathways, including, e.g.,
human organs, which is shown by the numerous re- ATP and phosphocreatine (PCr), 31P-MRS is outstand-
search publications in the field of MRS, the use of the ing for the evaluation of sources of metabolic energy in
technique is, in clinical routine, still restricted to a few the brain. Nevertheless, 1H-MRS has become the main
organs and a selection of diseases represented in the clinically applied tool in neurospectroscopy, as it pro-
studied tissues. Restrictions are mainly of a technical vides information on markers of neuronal function, my-
nature and will partly be overcome by the introduction elin, cell membranes, and metabolic active compounds,
of more powerful magnets and software solutions ap- as well as the fact that the MR sensitivity is much
plicable for clinical routine. greater for protons than it is for phosphorus and allows
The introduction of MRS to clinical neurospectros- for better spatial resolution.
copy went parallel to the tremendous development of Spatial resolution in in vivo 1H-MRS is limited, and
MRI applications in the field of neuroradiology. The spectra are usually acquired from a localized area ob-
taining single (single-voxel spectroscopy (SVS)) or mul-
tiple voxels (multivoxel spectroscopy, also referred to as
Department of Diagnostic Radiology, University Hospital Lund, Lund,
chemical shift imaging (CSI)) on the order of 1– 8 cm3.
Sweden. The acquisition of localized spectra is of special impor-
Part of the illustrations were presented in a lecture at the 6th Annual tance in focal or heterogeneous lesions where it allows
Meeting of ISMRM, Sydney, 1998, morning categorical course, by I.M.B. the separate evaluation of morphologically normal and
*Address reprint requests to: I.M.B., Department of Radiology, Univer-
sity Hospital, S-22185 Lund, Sweden.
pathologic-appearing parts in and adjacent to brain
E-mail: isabella.burtscher@drad.lu.se lesions, as well as the comparison to the contralateral
Received July 28, 2000; Accepted December 28, 2000. hemisphere.
© 2001 Wiley-Liss, Inc. 560
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Proton MR Spectroscopy in Clinical Routine 561

Figure 1. Metabolic compounds de-


tected by proton MRS at different echo
times. A: TE ⫽ 270 msec (PRESS se-
quence); reading from right to left, the
three main resonances NAA (2.0 ppm),
Cr (3.0 ppm), and Cho (3.2 ppm) are ob-
served. B: TE ⫽ 20 msec (STEAM se-
quence); with main additional reso-
nances at 3.6 and 4.0 ppm (mI) and at
2.6 ppm (additional NAA peak) and a
cluster of small peaks at 2.2–2.4 ppm,
consistent of Glx, which is mainly de-
tected in pathologically altered tissue,
e.g., in hepatic encephalopathy (not
shown).

To be able to acquire signals representing metabolic Total creatine (Cr), as well as phosphocreatine (PCr),
compounds of interest, the brain water resonance, the is a marker for brain energy metabolism and displayed
main signal source in 1H-MRS, has to be suppressed, at 3.0 ppm in the proton spectrum. Cr concentrations
as the water signal is much larger than the desired are comparable for different locations throughout the
signal of investigated brain compounds and their reso- brain and relatively resistant to changes. Therefore, Cr
nances would literally drown in the water peak. Special is used as the internal standard to which resonance
water suppression pulses are added to the pulse se- intensities of other compounds are normalized. How-
quences. ever, alterations in Cr levels occur, e.g., in trauma,
Besides localization and water suppression, shim tumors, and hypoxia. Cr, PCr, Cho, and myo-inositol
procedures represent one of the major challenges in (see below) are also recognized osmotically active me-
clinical MRS, as the separation of interesting reso- tabolites (7).
nances requires a field homogeneity exceeding stan- Lactate (Lac) is not seen in the spectrum of a healthy
dards known from conventional MRI. Although shim human. Pathologic accumulation (resonance at 1.3
procedures are often automated, manual shimming ppm) of this end product of anaerobe glycolysis indi-
might still be necessary in, e.g., short TE experiments, cates that oxidative metabolism is unable to meet en-
which allow the detection of compounds with short T2 ergy requirements. Neurological diseases accompanied
relaxation times, as discussed in the section below. by positive lactate findings in MRS are, e.g., hypoxia,
ischemia, epilepsy, tumors, and mitochondrial diseases
COMPOUNDS OBSERVABLE IN VIVO (1,7,8,14,20).
Potentially MR-visible lipids in normal brain tissue
Main resonances observed in a proton MR spectrum
are bound to macromolecules in membranes and mye-
(Fig. 1) are (5,7–12) N-acetyl-aspartate (NAA), and a
lin and therefore invisible. Due to severe pathologic
neuron marker observed at 2.02 ppm in the spectrum.
disruption of such structures, increased lipid turnover
A decrease of the compound is seen in processes involv-
leads to more mobile lipids, yielding detectable lipid
ing neuronal loss or damage, such as neoplasm, epi-
lepsy, multiple sclerosis (MS), stroke, hypoxia, and de- signals at 0.9 and 1.3 ppm (8).
mentia (13–16). Compared to the many diseases The glutamate (Glu) and glutamine (Gln) resonances
leading to decreased NAA, cases with a pathologically are often summarized as a multiplet, Glx, at 2.2–2.4
increased NAA resonance, such as Canavan’s disease ppm. The signal from normal brain tissue is weak, and
(17), occur rarely but are nonetheless well known. short echo time studies (10 –20 msec), studies at higher
Physiologically increasing NAA levels can be seen in field strength (ⱖ1.5 Tesla), or 13C or 15N studies are
brain development and maturation in the infant preferable to achieve a closer look at Glx turnover. Glu
(18,19), and they can represent axonal recovery follow- is a neurotransmitter, a redox marker, and, when oc-
ing neuronal damage (14). curring in pathologically increased concentrations, a
Choline (Cho), as well as choline-containing com- neurotoxin. Gln is an astrocyte marker (5,7).
pounds, has its main resonance at 3.2 ppm. It is often Myo-inositol (mI) is an astrocyte marker and os-
referred to as a cell membrane and myelin marker. The molyte, and its resonance, preferably evaluated with
major cerebral choline compound, phosphatidylcho- short echo times, appears at 3.6 ppm in the proton
line, is partly MR invisible, and stored in cell mem- spectrum (5,7).
branes, myelin, and other cerebral lipid pools. Its visi- Further metabolites, such as amino acids and ace-
ble form is released from them under diverse tate, will be referred to in the text below.
pathological conditions, such as acute myelin break- Although MR spectra, as any MR image, can be read
down and increased cellular density (5,7,15). visually, quantitative information on concentrations of
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
562 Burtscher and Holtås

observed compounds and metabolite (compound) ratios and complementary to that provided by MRI. Compared
permit a more precise and reproducible interpretation to space-occupying brain lesions, many patients pre-
of MRS data and should therefore be obtained (19,21– senting seizures do not show any MRI-detectable, mor-
23). Furthermore, clinical data and morphologic infor- phological alterations of brain parenchyma. Spectros-
mation based on computer tomography (CT), MRI, sin- copy provides the possibility of obtaining information
gle-photon-emission computer tomography (SPECT), on cerebral metabolism based on the evaluation of
and positron emission tomography (PET) have to be static metabolic abnormalities, primarily through the
taken into account when interpreting MR spectra, as NAA resonance, and on dynamic changes associated
pathologic entities easily differentiated on clinical or with the activation of anaerobic glycolysis through the
morphological bases can be impossible to differentiate lactate resonance (14,24,25).
by MRS alone. Also, age differences and concentration Temporal lobe epilepsy (TLE) is well studied by MRS,
differences for the observed compounds throughout the focusing on lateralization of the seizure (14,26 –29).
brain have to be considered (7,18,19). Measurements of NAA in epileptogenic regions have
consistently shown decreased NAA levels that represent
neuronal dysfunction and neuronal loss. MRS localizes
MAIN CLINICAL APPLICATIONS OF PROTON MRS unilateral seizure foci in concordance with EEG and
OF THE HUMAN BRAIN clinical recordings with high precision. However, MRS
In Europe, a developed part of the world with a high also demonstrates the presence of a high proportion of
standard of health care, several CNS diseases and sys- bilateral abnormalities (28), which can be difficult to
temic diseases with CNS involvement remain major observe with other techniques. MRS revealed both de-
causes of disability or death. More sophisticated diag- creased NAA levels to be present even in the absence of
nostic methods are solicited to identify, characterize, any MRI-visible abnormality and possible improvement
and classify these diseases to provide a diagnostic basis of NAA levels in the remaining temporal lobe after suc-
for new and/or standardized therapy regimens. cessful resection of the epileptogenic temporal lobe (30).
MRS is one of these sophisticated methods adjusted Based on these findings, NAA decrease is thought to
to the diagnostic challenges represented by diseases partly reflect neuronal dysfunction associated with the
like epilepsy, cerebral space-occupying lesions, MS, de- epileptic state rather than neuronal loss associated
generative diseases, and metabolic diseases. The im- with, e.g., hippocampal sclerosis. An increase in cho-
pact of these diseases on society both from economic line-containing compounds in the epileptogenic region
and humanitarian points of view can be imagined, con- might be associated with a reactive gliosis (24). In tissue
sidering some of the following incidence and prevalence directly involved in seizure activity, the metabolic de-
rates. mands may exceed the capacity of the tissue to provide
The mean incidence rate of epilepsy in Europe is the required energy by oxidative metabolism, leading to
50/100,000; however, regional differences occur, and regional accumulation of lactate as a result of activated
the prevalence is 6 –7/1000. Among space-occupying anaerobic glycolysis. This local increase in lactate can
lesions, brain tumors have an incidence rate of 15/ persist for several hours and may be used as a more
100,000 (prevalence of 60/100,000). Although brain direct marker of seizure activity (7,25,31). Figure 2 il-
tumors represent about 10% of neoplasm in adults lustrates spectral abnormalities and asymmetries in a
(higher percentage in children) and two-thirds of brain patient presenting with partial status epilepticus and
tumors are benign, highly malignant astrocytomas are electroencephalographically and clinically determined
the fourth leading cause of cancer mortality in men and unilateral epileptogenic focus.
women between 15 and 34 years of age and the fifth
leading cause in men between 35 and 54 years of age. Cerebral Space-Occupying Lesions
Other space-occupying lesions, such as brain ab-
scesses, are rare in developed countries. MS shows Cerebral space-occupying lesions can easily be de-
high regional differences in incidence (3/100,000) and tected and visualized by means of diagnostic tools, such
prevalence (between 50 –100/100,000; 70/100,000 in as MRI, CT, and SPECT. However, as visualization itself
central Europe) with the highest rates in the northern is often not sufficient for discrimination of different
parts of the United States, Canada, central and north- lesion types, MRS can add another dimension to differ-
ern Europe, southern Australia, and New Zealand. For ential diagnosis by providing biochemical profiles of the
degenerative diseases, exact prevalence and incidence lesions.
rates are often unknown. A total of 5%–7% of the pop- Comparable to the diversity of tumor types, a variety
ulation over 65 years of age suffers from Alzheimer’s of different spectroscopic methods has been suggested
disease (AD), the most common form of dementia in for differential diagnosis of brain tumors. Starting with
Western industrialized countries. Parkinson’s disease the interpretation of metabolite ratios and quantitative
shows highest prevalence rates in northern parts of MRS data acquired by means of single-volume spec-
America and Europe (200/100,000 in central Europe) troscopy measurements covering large and heteroge-
and incidence rates of 20/100,000. neous parts of the lesions, several spectral character-
istics could be associated to different tumor groups,
although large overlaps, questioning the specificity,
Epilepsy
were observed (8,32– 41). Consistent findings were the
Spectroscopy findings in seizures illustrate that MRS decrease and ultimately loss of NAA resonances in tu-
can provide valuable information that is different from mors derived from other-than-neuronal tissue and an
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Proton MR Spectroscopy in Clinical Routine 563

Figure 2. Partial status epilepticus. A: NAA is decreased and lactate slightly increased ipsilateral to the electroencephalographi-
cally determined epileptogenic focus (right, frontocentral) in an adult with partial status epilepticus. MRS localization scout
image (B) with measurement area and voxels represented by the spectra outlined in A and C. The contralateral brain tissue does
not show any spectral alterations (C).

increase in choline-containing compounds represent- gaged by T2-weighted lesions in MS patients, changes


ing increased cell turnover, cell density, and gliosis (Fig. in normal-appearing WM might be more significant
3A). Lactate, mobile lipids, choline, and NAA levels were for the chronic functional impairment of the patients
furthermore used for malignancy staging (41– 44), and than abnormalities in the lesions. Fu et al (58)
the occurrence of alanine was associated with menin- showed that changes in NAA in normal-appearing
giomas (34). More sophisticated evaluation of spectral WM correlated better with functional impairment
data using neuronal networks and automated pattern than those in T2-weighted lesions. MRS may be used
recognition showed promising results and, although for detection of axonal damage and demyelination in
their clinical implication still is limited to a few research MS and might be the only noninvasive method to
centers, will certainly contribute to an increased repro- evaluate future treatment targeting axonal injury and
ducibility of true differentiation of cerebral tumors in demyelination.
the future (45–54). CSI allows the observation of spec-
tral alterations in heterogeneous parts of space-occu- Degenerative Diseases
pying lesions based on increased resolution and more
efficient data acquisition regarding the number of vox- Although often diagnosed based on clinical findings,
els per examination time. The information provided several neurodegenerative diseases show MRS-detect-
can, as illustrated in Fig. 3, be used for differential able metabolic changes, which can contribute to the
diagnosis of focal vs. infiltrating tumors and ring-en- differentiation of different groups and subtypes of these
hancing tumors vs. abscesses (55,56). diseases, as well as to the monitoring of progression in
certain diseases or their response to therapy.
AD shows, as other types of dementia, decreased NAA
MS
levels (1,7,16,25). Differentiation might, however, be
Acute and chronic functional impairment in MS re- possible when utilizing information from short TE ex-
sults from multiple mechanisms, including demyeli- aminations revealing, e.g., increased mI in AD, com-
nation, conduction block, and axonal injury. The pared to increased Cho in multi-infarct dementia (7).
main spectroscopic finding in MS lesions is decreased Parkinson’s disease challenges MRS, due to technical
NAA. However, early increase in Cho and lactate, as limitations in spatial resolution necessary for the as-
well as early, transient decrease in Cr, can be ob- sessment of brain tissue involved in Parkinson’s dis-
served (15,25,35,57,58). In chronic lesions, Cr levels ease (e.g., substantia nigra) and in management of sus-
normalize while NAA levels stay low, representing in- ceptibility effects caused by excess of iron in these
complete recovery of axonal injury. Studies using regions. However, preliminary results from Holshouser
short echo times showed increased mI (59) and lipid et al (60) indicate a decreased NAA/Cho ratio in the
signals. Detecting decreased NAA levels even outside striatum in a treatment subset of patients.
lesions visualized using T2-weighted MRI, MRS In Huntington’s disease (HD), MRS reveals reduced
proved to be more sensitive in the detection of tissue NAA/Cho ratios and increased lactate in the cortex and
abnormalities in MS than conventional MRI. Consid- basal ganglia in symptomatic HD patients. Increased
ering the low percentage of white matter (WM) en- lactate levels detected by MRS guided the choice of
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License

Figure 3. CSI can contribute to differential diagnosis of ring-enhancing lesions, such as infiltrating brain tumors (high-grade
glioma in AA), focal metastases (adenocarcinoma metastasis in BA), and abscesses (CA) by the detection of spectral alterations
in heterogeneous parts of the space-occupying lesions. The spectra representing the cystic parts of the lesions allow the
differentiation of the abscess (CB) from the glioma (AB) and the metastasis (BB) based on the combination of the detected
resonances of amino acids (2.4 ppm; 1.4 ppm, alanine; and 0.9 ppm), acetate (1.9 ppm), lactate (1.3 ppm), and mobile lipids (0.9
ppm) in the spectrum of the abscess. In comparison, the spectra representing the cystic necrosis of the glioma (AB) are
dominated by a lactate peak, and the cystic part of the metastasis shows lactate and an unassigned peak at 2.0 ppm, not
consistent with NAA. Spectra from the boarders of the cysts show, due to partial volume effects, a combination of the resonances
representing the cystic components and surrounding contrast-enhancing tissue with increased Cho/NAA (AC, BC, and CC).
Normal-appearing tissue outside the lesions in metastases (BD) and abscesses (CD) do not show any major spectral alterations.
However, normal-appearing tissue surrounding a glioma might show increased Cho and decreased NAA (AD), representing the
infiltrative growth of the glioma.
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Proton MR Spectroscopy in Clinical Routine 565

Figure 4. Decreased NAA and a minor


lactate peak (inverted due to acquisition
with TE ⫽ 135 msec) following asphyxia
are seen in the MR spectrum (B) from a
voxel covering the basal ganglia (out-
lined in A) in a 2-day-old boy (gestational
age, 41 weeks).

treatment, and reversion of increased lactate following terns, and spectral information always has to be inter-
coenzyme Q treatment can be monitored (61). preted in conjunction with clinical findings and case
MRS utilized in other neurodegenerative diseases history. Some of the specific changes recognized as
showed decrease in NAA and increase in Cho and mI in characteristic for a particular metabolic disease when
amyotrophic lateral sclerosis (ALS) (7,62) and strong interpreted on the basis of the individual case are: ele-
lipid-triglyceride signal in central pontine myelinolysis vated lactate and sometimes decreased NAA in mito-
(CPM) consistent with histological evidence of lipid- chondrial disorders (Fig. 5) (5,7); elevation of Glu, de-
filled macrophages at the site of myelinolysis (7). pletion of mI, and reduction of Cho (in WM) in chronic
hepatic encephalopathy (66); reduced Cho and mI in
Hypoxia hypo-osmolar encephalopathy (7); increased NAA in
Canavan’s disease (17); increased glycine in nonketotic
General biochemical reactions underlie the major spec- hyperglycinemia; and elevated phenylalanine in phe-
tral alterations detected by 1H-MRS following hypoxia nylketonuria (5). MRS may also be used for monitoring
based on, e.g., asphyxia (Fig. 4), near-drowning, car- the therapeutic intervention on the abnormal metabo-
diac arrest, respiratory failure, stroke, TIA, or trauma lism in some of these disorders, as, e.g., in hepatic
(5,7,63– 65). At the expense of accumulation of lactate, encephalopathy with normalization of Glu, mI, and Cho
anaerobic glycolysis meets energy requirements when after liver transplantation (5,7).
O2 is insufficient, which also leads to increased Glx in
short echo time studies. Decrease in NAA reflects neu-
ron dysfunction and axonal injury, and NAA levels can CONCLUSIONS
continue to decline several days after the acute insult to
the CNS. Increased Cho levels can be observed in asso- MRS is an interesting and expanding branch of radiol-
ciation with diffuse axonal injury and myelin and mem- ogy and neuroradiology in particular, and acquisition of
brane breakdown/turnover. Also, changes in Cr may clinical MRS data has during the last decade become
occur after hypoxia, as O2 is insufficient. MRS may be convenient from a technical point of view. Interpreta-
used for prediction of impaired neurologic function tion of standard examinations has been made easier by
based on NAA and lactate levels and for monitoring the accessibility of extensive research reports on MRS
therapeutic intervention in, e.g., the acute phase of and separate sessions focusing on clinical MRS at in-
stroke. ternational and clinical, as well as technical, MR meet-
ings.
MRS has already great clinical impact in localization
Metabolic Diseases
of foci and brain damage in epilepsy and represents a
In metabolic diseases, inherited or acquired, the chal- valuable complement to conventional imaging tech-
lenge for MRS is most often not the detection of the niques, such as CT and MRI. MRS is superior to MRI in
primarily underlying metabolite, protein, enzyme, or the differential diagnosis of untreated bacterial ab-
transmitter, but the visualization of the secondary scesses vs. other space-occupying lesions and in the
changes caused by the pathophysiological mechanisms detection of tumor growth in morphologically normal
and expressed as neuronal dysfunction/damage, de- tissue. The possibility of a noninvasive evaluation of
myelination, gliosis, or accumulation of vast products. metabolic abnormalities provided by MRS allows, in
Only a limited number among the variety of metabolic comparison to MRI and CT, in many cases of metabolic
diseases evaluated by MRS show specific spectral pat- diseases, a more accurate differential diagnosis and
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
566 Burtscher and Holtås

Figure 5. MRS in a patient with a mitochon-


drial disorder (measurement area and voxel
outlined on a MRS scout image) (A) shows
increased lactate (B).

prediction of severity. MRS can also provide crucial 11. Luyten PR, den Hollander JA. Observation of metabolites in the
information for the evaluation of prognosis in asphyxia. human brain by MR spectroscopy. Radiology 1986;161:795–798.
12. Podo F, de Certaines J. Magnetic resonance spectroscopy in can-
Neurospectroscopy, however, still provides more
cer: phospholipid, neutral lipid and lipoprotein metabolism and
challenges and can be expected to expand to other clin- function. Anticancer Res 1996;16:1305–1316.
ical areas, such as therapy monitoring in, e.g., tumors, 13. Castillo M, Kwock L, Scatliff J, Mukherji SK. Proton MR spectros-
metabolic and degenerative diseases, or MS. Whether copy in neoplastic and non-neoplastic brain disorders. MRI Clin N
other diagnostic challenges can be found where MRS is Amer 1998;6:1–20.
14. Jackson GD, Connelly A. New NMR measurements in epilepsy. T2
useful above and beyond MRI, SPECT, PET, and CT will
relaxometry and magnetic resonance spectroscopy. Adv Neurol
depend on further clinical studies and future develop- 1999;79:931–937.
ments. Main technical challenges are the optimization 15. Arnold DL. Magnetic resonance spectroscopy: imaging axonal dam-
of pulse sequences for clinical settings, e.g., multislice age in MS. J Neuroimmunol 1999;98:2– 6.
sequences, the development of more user-friendly post- 16. Rose SE, De Zubicaray GI, Wang D, et al. A 1H MRS study of
probable Alzheimer’s disease and normal aging: implications for
processing protocols, and the introduction of neural
longitudinal monitoring of dementia progression. Magn Reson Im-
network analysis, automated pattern recognition, and aging 1999;17:291–299.
other mathematical or statistical methods for data 17. Grodd W, Kragelah-Mann I, Klose U, Sauter R. Metabolic and de-
analysis. structive brain disorders in children: findings with localized proton
MR spectroscopy. Radiology 1991;181:173–181.
18. van der Knaap MS, van der Grond J, van Rijen PC, Faber JA, Valk
REFERENCES J, Willemse K. Age-dependent changes in localized proton and
phosphorus MR spectroscopy of the brain. Radiology 1990;176:
1. Jenkins BG, Chen YI, Kuestermann E, et al. An integrated strategy
509 –515.
for evaluation of metabolic and oxidative defects in neurodegenera-
19. Kreis R, Ernst T, Ross BD. Development of the human brain: in vivo
tive illness using magnetic resonance techniques. Ann N Y Acad Sci
quantification of metabolite and water content with proton mag-
1999;893:214 –242.
netic resonance spectroscopy. Magn Reson Med 1994;30:424 – 437.
2. McCully K, Mancini D, Levine S. Nuclear magnetic resonance spec-
20. De Stefano N, Matthews PM, Ford B, Genge A, Karpati G, Arnold
troscopy. Its role in providing valuable insight into diverse clinical
DL. Short-term dichloroacetate treatment improves indices of ce-
problems. Chest 1999;116:1434 –1441.
rebral metabolism in patients with mitochondrial disorders. Neu-
3. Kurhanewicz J, Vigneron DB, Males RG, Swanson MG, Yu KK,
rology 1995;45:1193–1198.
Hricak H. The prostate: MR imaging and spectrsocopy. Present and
21. Ala-Korpela M, Usenius JP, Keisala J, et al. Quantification of me-
future. Radiol Clin North Am 2000;38:115–138.
4. Vion-Dury J, Meyerhoff DJ, Cozzone PJ, Weiner MW. What might tabolites from single-voxel in vivo 1H NMR data of normal human
be the impact on neurology of the analysis of brain metabolism by brain by means of time-domain data analysis. MAGMA 1995;3:
in vivo magnetic resonance spectroscopy? J Neurol 1994;241:354 – 129 –136.
371. 22. Michaelis T, Merboldt K-D, Bruhn H, Hänicke W, Frahm J. Abso-
5. Ross B, Michaelis T. Clinical applications of magnetic resonance lute concentrations of metabolites in the adult human brain in vivo:
spectroscopy. MR Quarterly 1994;10:191–247. quantification of localized proton MR spectra. Radiology 1993;187:
6. Castillo M, Kwock L, Mukherji SK. Clinical applications of proton 219 –227.
MR spectroscopy. AJNR Am J Neuroradiol 1996;17:1–15. 23. Barker PB, Soher BJ, Blackband SJ, Chatham JC, Mathews VP,
7. Rubaek Danielsen E, Ross B, editors. Magnetic resonance spectros- Bryan RN. Quantitation of proton NMR spectra of the human brain
copy diagnosis of neurological diseases. New York: Marcel Dekker, using tissue water as an internal concentration reference. NMR
Inc.; 1999. Biomed 1993;6:89 –94.
8. Sijens PE, Levendag PC, Vecht CJ, vanDijk P, Oudkerk M. 1H MR 24. Gadian DG, Connelly A, Duncan JS, et al. 1H magnetic resonance
spectroscopy detection of lipids and lactate in metastatic brain spectroscopy in the investigation of intractable epilepsy. [General
tumors. NMR Biomed 1996;9:65–71. discussion, p 122–126.] Acta Neurol Scand 1994;152(Suppl):116 –
9. Constantinidis I. Magnetic resonance spectroscopy and the prac- 121.
ticing neurologist. The Neurologist 1998;4:77–98. 25. Rudkin TM, Arnold DL. Proton magnetic resonance spectroscopy
10. Ross BD. The biochemistry of living tissues: examination by MRS. for the diagnosis and management of cerebral disorders. Arch Neu-
NMR Biomed 1992;5:215–219. rol. 1999;56:919 –926.
15222586, 2001, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.1079 by Republic of Macedonia Hinari NPL, Wiley Online Library on [28/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Proton MR Spectroscopy in Clinical Routine 567

26. Achten E, Boon P, Van de Kerckhove T, Caemaert J, De Reuck J, magnetic resonance image properties, spectroscopy, and neural
Kunnen M. Value of single-voxel proton MRS in temporal lobe networks. J Neurosurg 1997;86:755–761.
epilepsy. AJNR Am J Neuroradiol 1997;18:1131–1139. 46. Laws ER. Imaging brain tumors— beyond three dimensions. Nat
27. Cendes F, Andermann F, Preul MC, Arnold DL. Lateralization of Med 1996;2:271–272.
temporal lobe epilepsy based on regional metabolic abnormalities 47. Preul MC, Caramanos Z, Collins DL, et al. Accurate, noninvasive
in proton magnetic resonance spectroscopic images. Ann Neurol diagnosis of human brain tumors by using proton magnetic reso-
1994;35:211–216. nance spectroscopy. Nat Med 1996;2:323–325.
28. Connelly A, Jackson GD, Duncan JS, King MD, Gadian DG. Mag- 48. Tate AR, Crabb S, Griffiths JR, et al. Lipid metabolite peaks in
netic resonance spectroscopy in temporal lobe epilepsy. Neurology pattern recognition analysis of tumour in vivo MR spectra. Anti-
1994;44:1411–1417. cancer Res 1996;16:1575–1580.
29. Vermathen P, Laxer KD, Matson GB, Weiner MW. Hippocampal 49. Preul MC, Caramanos Z, Leblanc R, Villemure JG, Arnold DL.
structures: anteroposterior N-acetylaspartate differences in pa- Using pattern analysis of in vivo proton MRSI data to improve the
tients with epilepsy and control subjects as shown with proton MR diagnosis and surgical management of patients with brain tumors.
spectroscopic imaging. Radiology 2000;214:403– 410. NMR Biomed 1998;11:192–200.
30. Hugg JW, Kuzniecky RI, Gilliam FG, Morawetz RB, Faught RE, 50. Tate AR, Griffiths JR, Martinez-Pérez I, et al. Towards a method for
Hetherington HP. Normalization of contralateral metabolic function automated classification of 1H MRS spectra from brain tumors.
following temporal lobectomy demonstrated by 1H magnetic reso- NMR Biomed 1998;11:177–191.
nance spectroscopic imaging. Ann Neurol 1996;40:236 –239. 51. Hagberg G. From magnetic resonance spectroscopy to classifica-
31. Najm IM, Wang Y, Shedid D, Luders HO, Ng TC, Comair YG. MRS tion of tumors. A review of pattern recognition methods. NMR
metabolic markers of seizures and seizure-induced neuronal dam- Biomed 1998;11:148 –156.
age. Epilepsia 1998;39:244 –250. 52. Soltanian-Zadeh H, Peck DJ, Windham JP, Mikkelsen T. Brain
32. Taylor JS, Ogg RJ, Langston JW. Proton MR spectroscopy of pedi- tumor segmentation and characterization by pattern analysis of
atric brain tumors. Neuroimaging Clin N Amer 1998;8:753–779. multispectral NMR images. NMR Biomed 1998;11:201–208.
33. Ott D, Henning J, Ernst T. Human brain tumors: assessment with 53. Nikulin AE, Dolenko B, Bezabeh T, Somorjai RL. Near-optimal
in vivo proton MR spectroscopy. Radiology 1993;186:745–752. region selection for feature space reduction: novel preprocessing
34. Demaerel P. In vivo localized single-voxel proton magnetic reso- methods for classifying MR spectra. NMR Biomed 1998;11:209 –
nance spectroscopy of intracranial tumors. Int J Neuroradiol 1997;
216.
3:94 –110.
54. Tate AR, Howells S. Pattern recognition analysis. [Editorial.] NMR
35. De Stefano N, Caramanos Z, Preul MC, Francis G, Antel JP, Arnold
Biomed 1998;11:147.
DL. In vivo differentiation of astrocytic brain tumors and isolated
55. Burtscher IM, Holtås S. In vivo 1H-MR spectroscopy in untreated
demyelinating lesions of the type seen in multiple sclerosis using
1 and treated brain abscesses. AJNR Am J Neuroradiol 1999;20:
H magnetic resonance spectroscopic imaging. Ann Neurol 1998;
1049 –1053.
44:273–278.
56. Martı́nez-Pérez I, Moreno Á, Alonso J, et al. Diagnosis of brain
36. Carpinelli G, Carapella CM, Palombi L, Raus L, Caroli F, Podo F.
abscess by magnetic resonance spectroscopy. J Neurosurg 1997;
Differentiation of glioblastoma multiforme from astrocytomas by in
86:708 –713.
vitro 1H MRS analysis of human brain tumors. Anticancer Res
57. Richards TL, Bowen JD, Alvord EC, et al. Magnetic resonance brain
1996;16:1559 –1564.
spectroscopy. Basic concepts with emphasis on multiple sclerosis.
37. Go KG, Kamman RL, Mooyaart EL, et al. Localised proton spectros-
Int J Neuroradiol 1996;2:123–133.
copy and spectroscopic imaging in cerebral gliomas, with compar-
58. Fu L, Matthews PM, De Stefano N, et al. Imaging axonal damage of
ison to positron emission tomography. Neuroradiology 1995;37:
normal appearing white matter in multiple sclerosis. Brain 1998;
198 –206.
121:103–113.
38. Florian CL, Preece NE, Bhakoo KK, Williams SR, Noble M. Charac-
teristic metabolic profiles revealed by 1H NMR spectroscopy for 59. Davie CA, Hawkins CP, Barker GJ, et al. Serial proton magnetic
three types of human brain and nervous system tumours. NMR resonance spectroscopy in acute multiple sclerosis lesions. Brain
Biomed 1996;9:253–264. 1994;117(Pt 1):49 –58.
39. Broniscer A, Gajjar A, Bhargava R, et al. Brain stem involvement in 60. Holshouser BA, Komu M, Moller HE, et al. Localized proton NMR
children with neurofibromatosis type 1: role of magnetic resonance spectroscopy in the striatum of patients with idiopathic Parkin-
imaging and spectroscopy in the distinction from diffuse pontine son’s disease: a multicenter pilot study. Magn Reson Med 1995;33:
glioma. Neurosurgery 1997;40:331–337. [Comments: Neurosur- 589 –594.
gery 1997;40:337–338.] 61. Koroshetz WJ, Jenkins BG, Rosen BR, Beal MF. Energy metabo-
40. Castillo M, Kwock L. Proton MR spectroscopy of common brain lism defects in Huntington’s disease and effects of coenzyme Q10.
tumors. Neuroimaging Clin N Amer 1998;8:733–752. Ann Neurol 1997;41:160 –165.
41. Burtscher IM, Skagerberg G, Geijer B, Englund E, Ståhlberg F, 62. Bowen BC, Pattany PM, Bradley WG, et al. MR imaging and local-
Holtås S. Proton MR spectroscopy and preoperative diagnostic ac- ized proton spectroscopy of the precentral gyrus in amyotrophic
curacy: an evaluation of intracranial mass lesions characterized by lateral sclerosis. AJNR Am J Neuroradiol 2000;21:647– 658.
stereotactic biopsy findings. AJNR Am J Neuroradiol 2000;21:84 – 63. Rutgers DR, Klijn CJ, Kappelle LJ, van der Grond J. Cerebral
93. metabolic changes in patients with a symptomatic occlusion of the
42. Shimizu H, Kumabe T, Tominaga T, et al. Noninvasive evaluation of internal carotid artery: a longitudinal 1H magnetic resonance spec-
malignancy of brain tumors with proton MR spectroscopy. AJNR troscopy study. J Magn Reson Imaging 2000;11:279 –286.
Am J Neuroradiol 1996;17:737–747. 64. Kreis R, Arcinue E, Ernst T, Shonk TK, Flores R, Ross BD. Hypoxic
43. Tedeschi G, Lundbom N, Raman R, et al. Increased choline signal encephalopathy after near-drowning studied by quantitative 1H-
coinciding with malignant degeneration of cerebral gliomas: a serial magnetic resonance spectroscopy. J Clin Invest 1996;97:1142–
proton magnetic resonance spectroscopy imaging study. J Neuro- 1154.
surg 1997;87:516 –524. 65. Pendlebury ST, Lee MA, Blamire AM, Styles P, Matthews PM. Cor-
44. Speck O, Thiel T, Henning J. Grading and therapy monitoring of relating magnetic resonance imaging markers of axonal injury and
astrocytomas with 1H-spectroscopy: preliminary study. Anticancer demyelination in motor impairment secondary to stroke and mul-
Res 1996;16:1581–1586. tiple sclerosis. Magn Reson Imaging 2000;18:369 –378.
45. Arle JE, Morriss C, Wang ZJ, Zimmerman RA, Phillips PG, Sutton 66. McConnell JR, Antonson DL, Ong CS, et al. Proton spectroscopy of
LN. Prediction of posterior fossa tumor type in children by means of brain glutamine in acute liver failure. Hepatology 1995;22:69 –74.

You might also like