You are on page 1of 13

Review

The measurement and clinical relevance of brain atrophy in


multiple sclerosis
Robert A Bermel, Rohit Bakshi

Lancet Neurol 2006; 5: 158–70 Brain atrophy has emerged as a clinically relevant component of disease progression in multiple sclerosis.
Department of Neurology, Progressive loss of brain tissue bulk can be detected in vivo in a sensitive and reproducible manner by MRI.
Cleveland Clinic Foundation, Clinical studies have shown that brain atrophy begins early in the disease course. The increasing amount of data
Cleveland, OH, USA
linking brain atrophy to clinical impairments suggest that irreversible tissue destruction is an important
(R A Bermel MD); and Center for
Neurological Imaging, Partners determinant of disease progression to a greater extent than can be explained by conventional lesion assessments.
Multiple Sclerosis Center, In this review, we will summarise the proposed mechanisms contributing to brain atrophy in patients with
Departments of Neurology and multiple sclerosis. We will critically discuss the wide range of MRI-based methods used to quantify regional and
Radiology, Brigham and
whole-brain-volume loss. Based on a review of current information, we will summarise the rate of atrophy among
Women’s Hospital, Harvard
Medical School, Boston, MA, phenotypes for multiple sclerosis, the clinical relevance of brain atrophy, and the effect of disease-modifying
USA (Rohit Bakshi MD) treatments on its progression.
Correspondence to:
Dr Rohit Bakshi, Brigham & Historical background Focal-tissue loss within overt white-matter lesions is
Women’s Hospital, Harvard
Brain-volume loss was reported as a component of probably a major contributor to brain atrophy due to loss
Medical School, 77 Avenue Louis
Pasteur, HIM 730 Boston, multiple sclerosis in early descriptions of the pathology. of myelin and axonal density.10,14 But is there a relation
MA 02115, USA In 1938, Robert Carswell discussed multiple sclerosis in between lesion load and brain atrophy, even remote to
rbakshi@bwh.harvard.edu an article on atrophy in his Atlas of Pathology.1 In 1963, lesion development? One group studied 29 patients with
German pathologist Eduard Rindfleisch reported focal a clinically isolated demyelinating syndrome over
atrophy of brain tissue in his description of the 14 years.15 Progression of T2-hyperintense lesion load in
perivascular nature of multiple sclerosis lesions.2 With the first 5 years after disease onset was the best predictor
advances in technology,3 structural neuroimaging of whole-brain atrophy 14 years later, suggesting that
provides increasingly sensitive methods of monitoring lesions partly contribute to the development of global
brain atrophy in vivo. In this review we will focus on atrophy. However, most studies examining the relation
brain atrophy, referring the readers to separate reviews between T2-hyperintense lesions and whole-brain
on atrophy of the optic nerve and spinal cord in multiple atrophy reported that lesion load accounts for at most
sclerosis.4–8 10% of the variance in atrophy in patients with
relapsing-remitting multiple sclerosis, and has limited
Pathogenesis predictive value for the subsequent development of
Although brain atrophy is probably an endpoint of atrophy.16–18 An increasing amount of data show that
irreversible tissue loss in multiple sclerosis, it is not whole-brain grey-matter volume is moderately
pathologically specific. The underlying mechanisms for correlated with lesion volume, suggesting that remote
brain atrophy are diverse and complex. Some proposed effects of white-matter lesions are partly responsible for
mechanisms are directly related to the multifocal grey-matter degeneration.16,19–23 However, this
inflammatory disease process, whereas others are association might not extend to atrophy in deep grey-
indirectly related to or are independent from traditional matter nuclei, in which atrophy is poorly related to such
measures of overt lesions.9 lesions.24
Gadolinium-enhancing lesions partly predict the
Overt lesions development of whole-brain atrophy in early relapsing-
Multiple sclerosis lesions are dynamic and progress remitting multiple sclerosis in some studies.14,25–33 One
through distinct stages on MRI scans. Lesions that are study26 reported that the number of enhancing lesions
enhanced with intravenous-gadolinium contrast on T1- was moderately correlated with ventricular enlargement
weighted images represent active areas of inflammation in 16 patients with relapsing-remitting multiple
and dysfunction of the blood–brain barrier.10,11 Most sclerosis. Three other studies of patients with similar
gadolinium-enhancing lesions become permanent characteristics lend support to this association.27–29 Two
hyperintense lesions on T2-weighted images and groups showed that ring-enhancing lesions might be
represent a range of pathological changes (eg, oedema, particularly predictive of brain atrophy.26,30 However,
gliosis, inflammation, demyelination, remyelination, results from two studies17,31 suggest that gadolinium-
and axonal loss). About half of gadolinium-enhancing enhancing lesions do not predict whole-brain atrophy.
lesions will ultimately persist as severe hypointensity on Overall, the relation between enhancing lesion volume
T1-weighted images (T1 black holes).11 These persistent and brain atrophy seems to disappear in later disease
lesions represent irreversible tissue destruction and stages.32 Even with near complete pharmacological
axon loss.10,12,13 suppression of gadolinium-enhancing lesions in

158 http://neurology.thelancet.com Vol 5 February 2006


Review

patients with secondary progressive multiple sclerosis, parenchymal loss in 42 patients with relapsing-remitting
brain atrophy continues to progress.33 multiple sclerosis.39 Another group, measuring whole-
The association between T1-black holes and brain brain N-acetyl aspartate and diffusivity, showed these
atrophy however, is unclear.14,17,34,35 One study34 recorded a markers (but not any lesion-load volumes) were
relation between T1-black holes and supratentorial brain associated with whole-brain atrophy.40 These studies
atrophy (r=0·48) in a cross-sectional study of patients provide evidence to show that in addition to overt lesion
with relapsing-remitting multiple sclerosis (n=55). formation, global dysfunction or degeneration of neurons
Results from our group supported this finding in 78 might be occuring, and perhaps preceding brain atrophy.
patients with multiple sclerosis (r=0·35),35 and we also
reported an association between subcortical atrophy and Alternative mechanisms
volume of T1-black holes.36 This finding contrasts with The absence of a strong association between lesion load
studies showing that the volume of T1-black holes at and brain atrophy suggests that other mechanisms
baseline does not predict the development of contribute to atrophy. There has been evidence of a
longitudinal whole-brain atrophy in relapsing-remitting genetic predisposition to brain atrophy or
multiple sclerosis.17 neurodegeneration in multiple sclerosis. Specifically,
Are multiple sclerosis lesions and brain atrophy linked, patients who carry the APOE 4 allele have up to five
or are they unique results of different pathological times greater annual rate of brain atrophy than do
processes? On the basis of the above results, there seems patients who do not have the allele.41 Patients with this
to be a partial link between lesions and brain atrophy. genotype are also at risk for developing persistent T1-
One key mechanism linking MRI lesion load to brain black holes.41 Brain atrophy has also been associated
atrophy is the remote effect of axonal injury on neuronal with damage in grey matter, as measured by T2
loss (Wallerian degeneration). A study of the pathology37 hypointensity of subcortical structures, and is thought to
has shown the abundance of transected axons in the brain represent pathological iron deposition18,42,43 (figure 1).
of patients with multiple sclerosis (more than 11 000 per
mm3 of lesional tissue). Wallerian degeneration can be Localisation of brain atrophy
detected at the earliest stage of multiple sclerosis (in Brain atrophy seems to be widespread in multiple
patients with a clinically isolated demyelinating sclerosis, affecting all brain regions, including all the
syndrome).38 However, the slight predictive value of cerebral lobes, the compact white-matter tracts,
lesion volumes suggests that other mechanisms might be brainstem, and cerebellum.44 Both grey-matter and
contributing to volume loss.14 There has been a recent white-matter compartments are affected. Several groups
effort to correlate diffuse damage in the normal- have assessed grey-matter versus white-matter volume
appearing brain tissue with brain atrophy. One group loss using various segmentation tools, and selective
used MR spectroscopy to quantify N-acetyl aspartate as a grey-matter atrophy has been documented in patients
marker of neuronal integrity and showed that decreased with a clinically isolated demyelinating syndrome or
whole-brain N-acetyl aspartate preceded global- relapsing-remitting multiple sclerosis.16,20,21,23,24,45–47

Patient A Patient B

Baseline T2 intensity values: Baseline T2 intensity values:


Mean thalamus=0·44 Mean thalamus=0·59
Mean caudate=0·48 Mean caudate=0·63
Left putamen=0·42 Left putamen=0·59
Right putamen=0·42 Right putamen=0·59
Left globus pallidus=0·28 Left globus pallidus=0·44
Right globus pallidus=0·32 Right globus pallidus=0·44
Mean red nucleus=0·39 Mean red nucleus=0·46

Baseline BPF=0·82 Baseline BPF=0·85

Year 2 BPF=0·77 Year 2 BPF=0·85

Figure 1: Midthalamic T2-weighted axial images from two placebo patients at baseline
We did a post-hoc analysis of patients with MS from a 2-year clinical trial of interferon beta-1a (30g IM weekly) or placebo.18 We determined deep grey-matter T2
hypointensity, brain parenchymal fraction (BPF), and total T2-, gadolinium-enhancing-, and T1-lesion volumes. T2 hypointensity was chosen in regression modeling
as the best predictor of BPF change at the 1 year (R2=0·23, p=0·002) and 2 year (R2=0·33, p0·001) time points after accounting for all MRI variables. Mid-thalamic
T2-weighted axial images from two placebo patients at baseline are shown, relating visual to quantitative findings, showing a range of T2 intensity and BPF values.
Patient A (left) had relative baseline T2 hypointensity and atrophy, with marked atrophy progression. Patient B (right) had higher baseline T2 intensity and BPF
values, with no atrophy progression. Thus, grey matter T2-hypointensity predicts the progression of brain atrophy in patients with early relapsing-remitting multiple
sclerosis. This predictive effect is seen as early as the first year. We hypothesise that brain atrophy may involve iron deposition as a mediator of neurotoxicity or as a
disease epiphenomenon. Reproduced with permission from the American Medical Association.18

http://neurology.thelancet.com Vol 5 February 2006 159


Review

matter volume loss, which could have been the result of


8000
atrophy, was offset by inflammation or oedema-related
increases in tissue bulk. These data suggest that
decreases in grey-matter volume can identify progressive
7000 neurodegeneration more sensitively than can decreases
in white matter or whole-brain volumes.
Atrophy of the deep grey nuclei also seems to happen
Caudate volume (mm3)

6000 in multiple sclerosis (figures 2 and 3). Tissue loss from


the thalamus52,53 and the caudate nucleus24 has been
measured with MRI segmentation. There was
5000 substantial neuron loss, with neuronal density decreased
by 22%, in the thalamus of patients with multiple
sclerosis.52 This pathological evidence for
neurodegeneration in the grey matter highlights the
4000
underlying causes of damage detected non-invasively by
imaging studies, such as hypometabolism,54 decreased
neuronal activation,55,56 increased diffusivity,57 and
3000
Normal MS decreased N-acetyl aspartate.52
n=10 n=24
Methods used to measure brain atrophy
Figure 2: Box plot of normalised caudate volumes Qualitative measures
We tested whether caudate atrophy occurs in MS, and whether it correlates with
Clinically, brain atrophy can be identified from
conventional MRI or clinical markers of disease progression. Caudate nuclei of
24 patients with MS and ten age-matched healthy controls were traced, qualitative images by the recognition of an increase in
normalised, reconstructed, and visualised from high-resolution MRI scans. cerebrospinal fluid spaces or a reduction in size of
Normalised bicaudate volume was 19% lower in MS vs controls (p0·001), an parenchymal structures compared with the normal
effect that persisted after adjusting for whole-brain atrophy (p0·008).
Caudate volume did not correlate with total brain T2 hyperintense or
appearance for age (figure 4). Upon review of serial
T1-hypointense lesion load (both p0·05). Reproduced with permission from studies, progressive atrophy can be detected by
Lippincott, Williams & Wilkins.24 comparison of images (figure 5). Such relatively simple
determinations are easy to implement in routine patient
Cortical atrophy happens early in the course of multiple care or for semiquantitative analysis,44 and can help to
sclerosis45,48,49 and can be localised to specific regions of assess disease severity and disease progression.
the brain. One group measured cortical thickness in
20 patients with multiple sclerosis and noted the Quantitative two-dimensional measures
presence of focal atrophy in the bilateral frontal and Because of the limited reproducibility and precision of
temporal cortices early in the disease course, and visually based atrophy measures, quantitative
additionally in the motor cortex in patients with more
advanced disease.49 Evidence from other groups, I cm
however, suggests that atrophy similarly affects both
grey and white-matter compartments20,50 or preferentially
affects the white-matter compartment.19 The similarity of
the patient populations, small sample sizes, and
differences in techniques might explain, in part, the
conflicting conclusions of these studies.
Data show that the grey matter, although affected by
the disease, has relatively less inflammation51 compared
with white matter, which is more prominently affected.
This process could potentially cause increases in white-
matter volume and mask ongoing atrophy. Consistent
with this hypothesis, in a 3 year longitudinal study,16
patients with early-onset multiple sclerosis had large
decreases in grey-matter volume but small increases in
white-matter volume. Additionally, those with a Figure 3: Deep grey-matter damage might be an important component of
clinically isolated demyelinating syndrome had loss of the MS disease process
grey-matter volume, but there were no changes in white- Superior view of three-dimensional caudate reconstructions from a patient with
MS (green) and a normal control (pink), both age 50 years old, normalised for
matter volume. Both groups of patients had large head size. Surfaces were reconstructed from volume acquisition coronal MR
increases in white-matter lesions during the observation images using a manual parcellation technique, normalised for head size, and co-
period, prompting the authors to speculate that white- registered using Visualization Toolkit.24

160 http://neurology.thelancet.com Vol 5 February 2006


Review

Healthy control RRMS RRMS SPMS

BPF 0·89 BPF 0·84 BPF 0·80 BPF 0·70

Figure 4: Whole brain atrophy in MS as measured by a normalised proportional method


Representative midventricular axial non-contrast T1-weighted MRI scans are shown from age-matched individuals in the sixth decade. Note the progressive decrease
in brain parenchyma, increase in cerebrospinal fluid spaces, and decrease in brain parenchymal fraction (BPF) among the scans from left to right. The first patient with
relapsing-remitting multiple sclerosis (RRMS) has an EDSS score of 1·5 and disease duration of 5 years. The next patient with relapsing-remitting multiple sclerosis
has an EDSS score of 4·0 and disease duration of 10 years. The patient with secondary-progressive multiple sclerosis has an EDSS score of 6·5 and disease duration of
18 years. Reproduced with permission from the American Society for Experimental NeuroTherapeutics.11

techniques are preferred. Quantitative two-dimensional hardcopy films).29,35,36,58–63 Although two-dimensional


measures of atrophy include linear measures, which can measures have the advantage of relative ease of
be quantified on a single-image section with a distance implementation in the clinical setting, the main
tool on a computer workstation (or even a ruler on disadvantage is the absence of reproducibility compared

Figure 5: Progressive brain atrophy in a 41-year-old man with RRMS imaged at baseline and 4 years later
Non-contrast T1-weighted images show progressive enlargement of the ventricles and subarachnoid spaces consistent with diffuse brain volume loss. Top row is at
baseline, bottom row is at 4 year follow-up. Reproduced with permission from the American Society for Experimental NeuroTherapeutics.11

http://neurology.thelancet.com Vol 5 February 2006 161


Review

Figure 6: Two-dimensional measures of brain atrophy


Third ventricle width (A), maximum lateral ventricle width (B; arrows), brain width (C; arrows), corpus callosum area (D), and bicaudate ratio (E; minimum intercaudate distance [solid line] divided by
the brain width along the same line [dashed line]). Measurement of third ventricular width is done by measuring the width along the anteriorposterior midpoint of the third ventricle. Lateral ventricular
width is determined along a plane corresponding to the anteroposterior midpoint of the ventricle on an anatomical level from an axial slice at which the septum pellucidum remains thin. Brain width is
the distance between two points on the cortical surface, measured at the same level and along the same line as the lateral ventricle width. Corpus callosum area is determined by outlining the margins
of the structure from the best available midsagittal section The bicaudate ratio, also known as the intercaudate nucleus ratio, is measured from an axial section on which the heads of the caudate
nuclei are most visible and closest together. These two dimensional measures of brain atrophy have shown longitudinal sensitivity to disease progression,29 meaningful correlations with clinical
findings,29,36 and strong associations with three dimensional measures of whole brain atrophy.64 Both the third ventricle width and bicaudate ratio show associations with cognitive impairment, even
after accounting for other MRI biomarkers including whole-brain atrophy and lesion load.36,60 A–D reproduced with permission from Lippincott, Williams & Wilkins29

with three-dimensional measures. Figure 6 shows statistical parametric mapping (figure 7),23,64,67,82,83
examples of two-dimensional measures. template-driven segmentation,84 and brain boundry shift
integral.79 The advantages of registration-based methods
Quantitative three-dimensional measures are that they are sensitive to longitudinal changes and
Quantitative measures of whole-brain atrophy, acquired (because they are mostly automated) need little operator
by automated or semiautomated methods,64 have input and time. The main disadvantage is the need for
become the most powerful methods for assessing additional steps in image processing.
patients with multiple sclerosis over time because of Thus, there are a multitude of techniques now
their reproducibility, sensitivity, and potential to capture available to measure brain atrophy. However, the main
global disease effects.65,66 limitation of the use of brain atrophy in clinical trials is
Segmentation-based techniques include separation of that different measures can produce conflicting results.
intracranial contents into parenchymal and non- Differing results might be valid,85 but as a result of the
parenchymal classes, thereby arriving at an expression low specificity of brain atrophy, techniques can quantify
of what proportion of space is occupied by brain tissue at different physiological observations.
any given time. The most popular terminology for this
“proportional method” is brain parenchymal fraction Atrophy and disease course
(BPF) (figures 4 and 7).17,35,64,67,68 Other examples of Clinically isolated syndrome
techniques which use segmentation-based methods There is increasing evidence that brain atrophy is not
include 3DVIEWNIX,69 SIENAX,70, index of brain restricted to the later progressive stages, but begins in
atrophy,71 whole brain ratio,72 brain to intracranial cavity the earliest stages of multiple sclerosis.6 In patients with
ratio,73 brain to intracranial volume ratio,74 fuzzy clinically isolated demyelinating syndrome, whole-brain
connectedness,75 the Alfano method,76 MIDAS,77 and atrophy is detectable in those who go on to develop
ILAB4.78 Segmentation-based techniques can also multiple sclerosis.86,87 Patients who meet the
separately quantify the volume of grey matter, white international panel criteria for multiple sclerosis88 (but
matter, and cerebrospinal fluid compartments who have had only a single demyelinating event) had
(figure 7).23 The abnormal signal intensity of multiple detectable ventricular enlargement over the subsequent
sclerosis lesions presents a potential source of voxel year (median enlargement=0·3 cm3, n=27) compared
misclassification. Lesions in the white matter tend to be with those who do not meet the criteria (median
misclassified as grey matter,20 and misclassified volumes enlargement=0·05 cm3, n=28, p=0·03).77 In another
should be corrected to avoid confounding clinical study of patients with clinically isolated demyelinting
correlations.23 syndrome followed up over 3 years, the group that
Registration-based techniques are designed to follow developed multiple sclerosis during the study period
patients longitudinally over time.79 These methods align had detectable grey-matter atrophy, but no white-matter
two serial scans from a patient and find areas of signal- atrophy.16 In the placebo group of a randomised
intensity change after accounting for changes in head controlled trial of subcutaneous interferon beta-1a in
position or slice plane. The result is a number such as clinically isolated demyelinating syndrome,
percent brain volume change. Examples of such investigators measured a mean decrease in brain-
measures include SIENA,80 voxel-based morphometry,81 parenchymal volume of 1·68% over 2 years.89 In

162 http://neurology.thelancet.com Vol 5 February 2006


Review

Figure 7: Representative slices from a T1-weighted gradient echo series and obtained grey, white, and CSF tissue compartments after automated skull-
stripping and segmentation by SPM99 for a normal control (top row) and a patient with MS (bottom row)
Images from left to right are: raw T1-weighted image; white matter only (bright areas); grey matter only (bright areas); CSF only (bright areas) with background brain
parenchyma (inner dark areas). Note the misclassification of white-matter lesions that have been classified as grey matter. This procedure can be used to quantify the
volumes separately, or determine residual or proportion-based measures of normalised whole-brain atrophy. BPF=total parenchymal volume divided by total
intracranial volume. Applying this method we studied 41 patients with MS and 18 age and sex-matched healthy controls. We also measured lesion load (total T1-
hypointense and FLAIR hyperintense lesion volume), third ventricular width and bicaudate ratio. Disability was assessed by the EDSS and timed 25-foot walk. Patients
with MS had lower grey matter (3·9%, p=0·003) and total parenchymal volume (3·8%, p=0·003), but only a trend for lower white-matter volume (3·7%,
p=0·052) relative to normal controls. Grey-matter atrophy was most closely related to clinical status as compared to all other MRI measures (see figure 8 and table for
more information). Reproduced with permission from Academic Press.23

summary, when brain atrophy occurs in clinically Secondary-progressive multiple sclerosis


isolated demyelinating syndrome it seems to be related Studies which have assessed both patients with
to the underlying multiple sclerosis disease process, as relapsing-remitting multiple sclerosis and those with
pathological brain atrophy is more common in patients secondary-progressive multiple sclerosis have reported
who go on to develop multiple sclerosis than in those that the annual rates of atrophy are similar between the
who do not. two groups, with rates between 0·6% and 0·8% per
year.6,79,91,98 In a clinical trial of an immunomodulatory
Relapsing-remitting multiple sclerosis drug in secondary-progressive multiple sclerosis, the
Brain atrophy has been well documented in relapsing- placebo group (n=31) had a mean brain-volume loss of
remitting multiple sclerosis, and seems to happen 3·86% over 3 years.32 Other studies showed that brain
rapidly in early stages of the disease.6 Overall, the rate of atrophy occurred at a lower rate in secondary-progressive
brain-parenchymal volume loss in relapsing-remitting multiple sclerosis than in relapsing-remitting multiple
multiple sclerosis is 0·6–1·35% per year. In the pivotal sclerosis,92,101 suggesting that the most aggressive rates of
trial of weekly intramuscular interferon-beta 1a, patients atrophy happen early on in the disease course,
with early relapsing-remitting multiple sclerosis (mean coincident with rapid accumulation of lesion burden.33
disease duration 6·2 years) and only mild disability
(mean expanded disability status scale (EDSS] 2·3) had a Primary-progressive multiple sclerosis
lower whole-brain volume than age-matched normal In a multicentre study of progressive multiple
controls (5 SD below the normal control group) and a sclerosis,102 137 patients with primary-progressive
higher annual rate of brain-volume loss than controls disease averaged 1·3% loss of brain-parenchymal
over 2 years (0·6% per year vs 0·1% per year for volume over 1 year. Another group103 assessed
healthy controls).17 Another study measured brain 41 patients with primary-progressive multiple sclerosis
parenchymal volume in 53 patients with early untreated and showed a mean reduction in brain-parenchymal
relapsing-remitting multiple sclerosis (disease duration volume of 3·7% over 5 years. In the placebo group of a
1–5 years, EDSS5) and reported that volume loss therapeutic trial in this type of multiple sclerosis,
averaged 2·7% over 2 years.90 Other groups have ventricular volume increased over 2 years.104 Therefore
observed the rates of brain atrophy in either untreated brain atrophy happens in patients with primary-
patients31,68,79,91–95 or those in clinical trials.74,89,96–100 progressive multiple sclerosis at a rate greater than in

http://neurology.thelancet.com Vol 5 February 2006 163


Review

those with secondary-progressive multiple sclerosis, but Neuropsychological impairment


not as aggressively as reported in some studies of Multiple neuropsychological domains are affected by
relapsing-remitting multiple sclerosis.6 multiple sclerosis, including cognition, mood,
personality, fatigue, and quality of life.108–117 An early
Clinical correlations study118 identified an association between third
Physical disability ventricular width and cognition. Multiple subsequent
Lesion-load measures on MRI are associated only weakly studies have confirmed the link between brain atrophy
with physical disability in cross-sectional studies, which and neurobehavioural status.36,48,60,62,74,90,108,110,119–125 Our
has led to the traditional “clinical-imaging paradox” of group60 reported that third ventricular width was the
multiple sclerosis.105 Lesions can appear on MRI without most significant MRI predictor of cognitive dysfunction
any progression in physical disability, and conversely, in patients with multiple sclerosis. In this same study,
patients can progress in disability without the appearance whole-brain atrophy (ie, brain parenchymal fraction)
of new lesions. Whole-brain atrophy has a stronger, yet was the next significant predictor, and notably both
moderate, imaging association with physical disability, atrophy measures predicted more variance in
and is a stronger predictor of future disability than T1- neuropsychological function than did MRI-lesion
hypointense and T2-hyperintense lesion load.6 Many measures. Another study90 noted a strong correlation
groups have tested the relation between whole-brain between whole-brain parenchymal volume (but not
atrophy and EDSS score. One study showed that the rate lesion loads) and cognitive function in early relapsing-
of progression of brain atrophy over 18 months was remitting multiple sclerosis (r=0·51, p=0·0003). Many
significantly heightened in patients with deterioration of studies suggest that subcortical atrophy is well
disability, whereas only trends were seen between correlated with cognition, to a greater extent than whole-
disability change and progression of T2-hyperintense brain atrophy or lesion load.36,60,124 Grey-matter atrophy48
lesions.96 Another group reported significant correlations and lobar atrophy125 have also shown correlations with
between EDSS score and ventricular volume, but no cognitive dysfunction, to a greater extent than seen with
association between lesion loads and EDSS.106 Our group lesion-load measures.
and others have recorded similar associations between From the studies on brain atrophy and neuropsycho-
brain atrophy and measures of physical disability logical impairment we conclude that subcortical atrophy
(figure 7, figure 8, table).22,23,35 has a stronge association with general cognitive
Early atrophy seems to predict the subsequent dysfunction, which can be explained by a disruption of
development of physical disability to a better extent than frontal-subcortical circuits. Regional or cortical atrophy
lesion-load measures. A longitudinal study107 reported measures might provide insight into impairment of
that patients with more atrophy during the first 2 years more specific functional domains in some patients.
had greater disability 8 years later. Furthermore, the
change in brain parenchymal fraction over the first Effect of technical or other factors on volume
2 years was the best MRI predictor of 8 year disability measurement
after accounting for the 2 year change in all MRI-lesion A prerequisite for the use of brain volume as a surrogate
measures. At 8 years, patients with the largest amount of marker for irreversible tissue damage and neuro-
brain atrophy were about four times more likely to have a degeneration in patients with multiple sclerosis is the
disability level needing assistance with walking, or worse. need to understand the potential effect of other factors
Thus, brain atrophy is emerging as a clinically relevant such as MRI-related technical error and biological
measure of multiple sclerosis disease progression, factors that can affect brain volume independent of
supported by its association with physical disability. tissue destruction.
Spinal cord atrophy seems to show particularly strong
relations with physical disability, as reviewed separately.4 Effect of MRI protocol and analysis approach
Changes in the position of the patient’s head or slice
plane between scans are potential sources of error when
25-foot timed walk (residual)

measuring serial changes in brain volume. Either a


EDSS score (residual)

normalisation or co-registration procedure should


address these concerns. Normalised measures of whole-
brain volume (eg, expressed as a ratio to head size) show
a higher degree of reproducibility and sensitivity than do
those expressed as absolute volumes and have about
Whole grey-matter volume (residual) Whole grey-matter volume (residual)
twice the statistical power.65 This degree of difference
might improve the ability to detect treatment effects in
Figure 8: Correlation of whole-brain normalised grey-matter volume with physical disability
clinical trials.
Left: corrected whole grey-matter volume with EDSS scores (partial r=0·46, p=0·004). Right: corrected whole-
grey matter volume with the 25-foot timed walk (partial r=0·52, p=0·002). Reproduced with permission from In studying the longitudinal reproducibility of brain
Academic Press.23 parenchymal fraction, investigators did two serial MRI

164 http://neurology.thelancet.com Vol 5 February 2006


Review

n Parenchymal Grey matter White matter Third ventricle Bicaudate T1 black hole FLAIR lesion
volume volume volume width ratio lesion volume volume
EDSS 40 0·34* 0·46† 0·06 0·20 0·32* 0·41† 0·34*
25-foot timed walk 35 0·45† 0·52† 0·20 0·43* 0·32 0·46† 0·38*
Disease duration 41 0·46† 0·44† 0·31 0·48† 0·44† 0·34* 0·32*
Third ventricle width 41 0·80§ 0·71§ 0·60§ .. 0·80‡ 0·50† 0·47†
Bicaudate ratio 41 0·75§ 0·66§ 0·56§ .. .. 0·56‡ 0·49†
T1 black hole lesion volume 41 0·45† 0·46† 0·27 .. .. .. 0·89‡
FLAIR lesion volume 41 0·39* 0·43† 0·18 .. .. .. ..

Partial correlations (df=n4) are based on whole-brain data and express associations after adjusting for intracranial volume and age. FLAIR=fluid-attenuated inversion-recovery and
ellipses. *p0·05 †p0·01 ‡p0·001 §p0·0001. Note that grey-matter atrophy correlates most highly with measures of physical disability (more so than lesion load or white-matter-
volume measures). Reproduced with permission Academic Press.23

Table: Correlations between MRI measures (atrophy and lesion loads) and physical disability in patients with multiple sclerosis

scans (30 min apart) on 30 patients with multiple patients with secondary-progressive multiple sclerosis
sclerosis and calculated their brain parenchymal but not in those with relapsing-remitting multiple
fraction. The authors reported that a change of brain sclerosis. This decrease peaked at about a 1% change 2
parenchymal fraction of 0·0056% carries 95% certainty weeks after the steroids were first given and only in part
that this measure has actually changed (given a standard resolved over 8 weeks in patients with secondary-
error of 0·0020%).126 Thus, brain parenchymal fraction is progressive multiple sclerosis.
a sensitive measure and is an appealing method to serve By contrast, steroids might have a protective effect on
as a supportive outcome measure in clinical trials. brain atrophy in the long term in patients with multiple
The ability to pool results from different imaging sclerosis. In a phase II placebo-controlled study of
hardware is essential to help with multicentre clinical patients with relapsing-remitting multiple sclerosis,
trials. One group127 studied the effect of brain intravenous pulsed steroids reduced cerebral atrophy
parenchymal fraction acquired on different scanners over 5 years.133 Furthermore, the effect of one disease-
and different pulse sequences. They showed that this modifying therapy on brain atrophy measured in a
quantity can be accurately measured using a variety of clinical trial of patients with relapsing-remitting
MRI pulse sequences where there is a marked contrast multiple sclerosis seemed to be unaffected by steroid
between cerebrospinal fluid and brain parenchyma. Our dose near the time of MRI.134 Another study135 showed
group64 reported no effect of pulse sequences (spin echo that pulses of intravenous methylprednisolone injected
vs gradient echo) on the ability to accurately measure every 4–6 weeks did not accelerate (but rather slowed)
brain parenchymal fraction using both semiautomated the rate of brain atrophy in 11 patients with primary-
and automated methods; both protocols yielded robust progressive multiple sclerosis. In conclusion,
clinical correlations. Another group identified that good intravenous steroids seem to have a dual effect on brain
reproducibility of brain parenchymal fraction can be volume, but in the long term might protect against brain
achieved across scanners when using the same pulse atrophy in relapsing-remitting forms of the disease.
sequence at the same field strength.128
Effect of disease-modifying treatments
Reversible biological effects on brain volume Brain atrophy, because of its clinical relevance and
Another factor thought to confound brain volume ability to serve as a sensitive, reproducible quantitative
measures in multiple sclerosis is acute inflammation measure of irreversible tissue destruction, is now
and oedema in white matter that might transiently commonly used as a secondary therapeutic outcome
increase brain volume and offset decreases related to measure in clinical trials for multiple sclerosis.5,136
tissue destruction.16 Corticosteroids have been associated
with reduced brain volume in patients treated for Interferon beta-1a
neurological and non-neurological diseases.129 Proposed In the pivotal phase III trial of once-weekly intramuscular
mechanisms include steroid-induced protein catabolism interferon beta-1a,17 whole-brain atrophy (by brain
and reduction of water volume in the brain as a result of parenchymal fraction) was measured in patients with
decreased vascular permeability. Corticosteroids also relapsing-remitting multiple sclerosis randomised to
affect brain volume in patients with multiple sclerosis, interferon beta-1a and patients randomised to placebo.17
although the relation is complex. An acute effect of There was a 55% reduction in brain atrophy with
intravenous corticosteroids on brain volume has been interferon therapy in the second year of treatment
shown by several groups.130–132 One group132 looked at the (0·233% reduction in brain parenchymal fraction in
effects of a typical course of intravenous interferon beta-1a vs 0·521% in placebo), but no effect
methylprednisolone. The researchers identified a during the first year of treatment. In a larger study of
significant decrease in brain parenchymal fraction in patients who were treated with the same medication,

http://neurology.thelancet.com Vol 5 February 2006 165


Review

rates of decline in brain parenchymal fraction were “pseudoatrophy”, such as that due to treatment-related
0·686% in the first year, 0·377% in the second year, and resolution of brain oedema and inflammation.142
0·378% in the third year, again suggesting, delayed, but Whether interferon beta three times a week or every-
significant slowing of the rate of brain atrophy.137 other-day limits the rate of brain atrophy is unclear.
A large-scale randomised placebo-controlled Proposed mechanisms by which interferon might limit
prospective trial of once-weekly subcutaneous interferon brain atrophy include: increasing nerve growth factors;143
beta-1a showed a beneficial effect on whole-brain limiting immune-mediated destructive inflammation;144
atrophy after 2 years of treatment in patients with or by limiting toxic mechanisms such as pathological
clinically isolated demyelinating syndrome.89 The rate of iron deposition.18
brain-parenchymal volume decrease for patients on
placebo was 0·83% during the first year and 0·67% Glatiramer acetate
during the second year. Respective values for treated Trials of daily subcutaneous glatiramer acetate suggest
patients were 0·62% and 0·61%, with a treatment effect that it also limits brain atrophy. One group97 recorded a
seen after 2 years (p=0·0031). This is in contrast with a reduction in the amount of whole-brain volume loss
post-hoc analysis of the initial large phase III trial of the between 14 patients treated with glatiramer acetate and
same medication dosed at three times a week in patients 13 treated with placebo over 2 years (0·6% per year vs
with relapsing-remitting multiple sclerosis which 1·8% per year, p=0·0078). In a larger scale trial of
reported no effect of treatment on the loss of brain shorter duration, brain volume was measured in
volume (measured as whole brain ratio) over 2 years.72 113 patients treated with glatiramer acetate for
Another group100 did a post-hoc analysis of an open label, 18 months and 114 patients on placebo for the first
randomised dose comparison trial of subcutaneous 9 months (then switched to active treatment in the
interferon beta-1a (11 g vs 33 g subcutaneously three second 9 months) using a central seven-slice brain-
times per week) in 52 patients with relapsing-remitting atrophy absolute-volume measurement technique.28
multiple sclerosis, and showed that there was a Despite a reduction in inflammatory lesions there was
significant decline in brain volume in both groups no detecatble difference in brain-volume loss between
during the 2 years of the study, and no difference in groups. However, in a post-hoc analysis of the same
change in brain volume between doses. Two subset study with a more reproducible normalised whole-
analyses of 38 patients (with a mix of patients with brain volume measurement technique, the rate of brain
relapsing-remitting or secondary-progressive multiple atrophy was lower in the fully treated group in the last
sclerosis) in two trials138,139 showed no significant 9 months than in the placebo.65 Thus, glatiramer
treatment effect on upper cervical spinal cord area,140 but acetate might have a partial and delayed, but significant
a slowing in the rate of brain atrophy (measured as brain effect on limiting the rate of brain atrophy in relapsing-
parenchymal volume) over 18 months.98 Thus data are remitting multiple sclerosis.
conflicting for the effects of subcutaneous interferon
beta-1a on CNS atrophy. Conclusions
Data obtained over 3 years in an open-label study of Brain atrophy has emerged as a clinically relevant
interferon beta-1b in 30 patients with relapsing- component of the multiple sclerosis disease process.
remitting multiple sclerosis showed a significant but Progressive loss of brain tissue bulk can be identified
delayed reduction in the rate of cerebral atrophy.141 in-vivo in a sensitive and reproducible manner by MRI.
Brain volumes at baseline decreased by 1·35% during Brain atrophy begins early in the disease course. The
the first year of the study, then remained relatively increasing amount of data linking brain atrophy to
stable during the second (1·48% lower than at baseline) clinical impairments suggest that irreversible tissue
and third (1·68% lower than baseline) years. A trial of destruction is a major determinant of disease
interferon beta-1b in 95 patients with secondary- progression to a greater extent than can be explained by
progressive multiple sclerosis32 showed no significant conventional lesion assessments. Whole-brain atrophy
treatment effect overall on brain volumes; however, the is an appealing method to monitor patients for the
authors identified a significant effect in the subgroup of destructive aspects of the disease in clinical trials.
patients who had gadolinium-enhancing lesions at Measures of regional brain atrophy might be
study entry. particularly useful in helping to understand
When interferon beta is given once a week it seems to neuropsychological dysfunction or other specific
have a partial delayed beneficial effect on brain atrophy, clinical findings. Confounding factors must be
irrespective of formulation. The treatment effect is considered when assessing whether loss of brain
delayed by at least several months. This delay might volume directly indicates tissue atrophy. Although the
indicate the fact that the atrophy occurring in the first mechanisms that contribute to brain atrophy are
months is the culmination of a cascade of events that unclear, degenerative processes that underlie brain
began before the onset of therapy. Alternatively, the atrophy will hopefully provide novel therapeutic targets
ongoing loss of brain volume might be the result of to help patients.

166 http://neurology.thelancet.com Vol 5 February 2006


Review

15 Chard DT, Brex PA, Ciccarelli O, et al. The longitudinal relation


Search strategy and selection criteria between brain lesion load and atrophy in multiple sclerosis: a 14 year
follow up study. J Neurol Neurosurg Psychiatry 2003; 74: 1551–54.
References for this review were identifed by searches of 16 Dalton CM, Chard DT, Davies GR, et al. Early development of
PubMed from 1985 to October 2005 with the terms “brain multiple sclerosis is associated with progressive grey matter
atrophy in patients presenting with clinically isolated syndromes.
atrophy” or “spinal cord atrophy” and “multiple sclerosis”. Brain 2004; 127: 1101–07.
Articles resulting from that search as well as references cited 17 Rudick RA, Fisher E, Lee JC, Simon J, Jacobs L. Use of the brain
in those articles were considered for this review of brain parenchymal fraction to measure whole brain atrophy in relapsing-
remitting MS. Multiple sclerosis collaborative research group.
atrophy. Articles were also identifed through searches of the Neurology 1999; 53: 1698–1704.
authors’ own files. Only papers published in English were 18 Bermel RA, Puli SR, Rudick RA, et al. Prediction of longitudinal
reviewed. brain atrophy in multiple sclerosis by gray matter magnetic
resonance imaging T2 hypointensity. Arch Neurol 2005;
62: 1371–76
Acknowledgments 19 Ge Y, Grossman RI, Udupa JK, Babb JS, Nyul LG, Kolson DL.
RB was supported by research grants from the National Institutes of Brain atrophy in relapsing-remitting multiple sclerosis: fractional
volumetric analysis of gray matter and white matter. Radiology
Health (NIH-NINDS 1 K23 NS42379-01), National Multiple Sclerosis
2001; 220: 606-610.
Society (RG 3258A2/1; RG 3574A1), and National Science Foundation
20 Chard DT, Griffin CM, Parker GJ, Kapoor R, Thompson AJ,
(DBI-0234895). We thank Sophie Tamm for assistance with manuscript
Miller DH. Brain atrophy in clinically early relapsing-remitting
preparation. multiple sclerosis. Brain 2002; 125: 327–37.
Authors’ contributions 21 Quarantelli M, Ciarmiello A, Morra VB, et al. Brain tissue volume
RAB contributed to the literature search, and both RB and RAB changes in relapsing-remitting multiple sclerosis: correlation with
contributed to the overall paper design, writing, and editing. lesion load. Neuroimage 2003; 18: 360–366.
22 Tedeschi G, Lavorgna L, Russo P, et al. Brain atrophy and lesion
Conflicts of interest load in a large population of patients with multiple sclerosis.
RB discloses the following relationships in the past 3 years with Neurology 2005; 65: 280–85.
companies involved in the production or sales of pharmaceutical 23 Sanfilipo MP, Benedict RH, Sharma J, Weinstock-Guttman B,
treatments for multiple sclerosis: consultancies (Biogen Idec, Serono, Bakshi R. The relationship between whole brain volume and
Teva Neuroscience), speaker’s fees (Berlex, Biogen Idec, Serono, Teva disability in multiple sclerosis: a comparison of normalized gray vs
Neuroscience), and research funding (Biogen Idec, Pfizer, Serono, Teva white matter with misclassification correction. Neuroimage 2005;
Neuroscience, Pepgen). RAB has no conflicts of interest. 26: 1068–77.
24 Bermel RA, Innus MD, Tjoa CW, Bakshi R. Selective caudate
References
atrophy in multiple sclerosis: A 3D MRI parcellation study.
1 Carswell R. Pathological anatomy: illustrations of the elementary
Neuroreport 2003; 14: 335-339.
forms of disease. London: Longman, Orme, Browne, Green, and
Longman 1838. 25 Zivadinov R, Zorzon M. Is gadolinium enhancement predictive of
the development of brain atrophy in multiple sclerosis? A review of
2 Rindfleisch E. Histologisches detail zu der grauen degeneration
the literature. J Neuroimaging 2002; 12: 302–09.
von gehirn und ruckenmark. Arch of Pathol Anatomy Physiol 1863;
26: 474–83. 26 Leist TP, Gobbini MI, Frank JA, McFarland HF. Enhancing
magnetic resonance imaging lesions and cerebral atrophy in
3 Gross KR, Tomberg TA, Kokk AA, Kaasik AE. The prognosis of
patients with relapsing multiple sclerosis. Arch Neurol 2001;
multiple sclerosis: computed tomographic comparisons.
58: 57–60.
Zh Nevropatol Psikhiatr Im S S Korsakova 1993; 93: 32–35.
27 Luks TL, Goodkin DE, Nelson SJ, et al. A longitudinal study of
4 Lin X, Tench CR, Evangelou N, Jaspan T, Constantinescu CS.
ventricular volume in early relapsing-remitting multiple sclerosis.
Measurement of spinal cord atrophy in multiple sclerosis.
Mult Scler 2000; 6: 332–37.
J Neuroimaging 2004; 14 (3 suppl): 20–26.
28 Rovaris M, Comi G, Rocca MA, Wolinsky JS, Filippi M.
5 Rudick RA. Impact of disease-modifying therapies on brain and
European/Canadian Glatiramer Acetate Study Group. Short-term
spinal cord atrophy in multiple sclerosis. J Neuroimaging 2004;
brain volume change in relapsing-remitting multiple sclerosis:
14 (3 suppl): 54–64.
effect of glatiramer acetate and implications. Brain 2001;
6 Zivadinov R, Bakshi R. Central nervous system atrophy and clinical 124: 1803–12.
status in multiple sclerosis. J Neuroimaging 2004; 14 (3 suppl): 27–35.
29 Simon JH, Jacobs LD, Campion MK, et al. A longitudinal study of
7 Rocca MA, Hickman SJ, Bo L, et al. Imaging the optic nerve in brain atrophy in relapsing multiple sclerosis. Neurology 1999;
multiple sclerosis. Mult Scler 2005; 11: 537–41. 53: 139–48.
8 Simon JH. Brain and spinal cord atrophy in multiple sclerosis. 30 Zivadinov R, Bagnato F, Nasuelli D, et al. Short-term brain atrophy
Neuroimaging Clin N Am 2000; 10: 753–70. changes in relapsing-remitting multiple sclerosis. J Neurol Sci 2004;
9 Minagar A, Toledo EG, Alexander JS, Kelley RE. Pathogenesis of 223: 185–93.
brain and spinal cord atrophy in multiple sclerosis. J Neuroimaging 31 Saindane AM, Ge Y, Udupa JK, Babb JS, Mannon LJ, Grossman RI.
2004; 14 (3 suppl): 5–10. The effect of gadolinium-enhancing lesions on whole brain atrophy
10 Bruck W, Bitsch A, Kolenda H, Bruck Y, Stiefel M, Lassmann H. in relapsing-remitting MS. Neurology 2000; 55: 61–65.
Inflammatory central nervous system demyelination: correlation of 32 Molyneux PD, Kappos L, Polman C, et al. The effect of interferon
magnetic resonance imaging findings with lesion pathology. beta-1b treatment on MRI measures of cerebral atrophy in
Ann Neurol 1997; 42: 783–93. secondary progressive multiple sclerosis: European study group on
11 Bakshi R, Minagar A, Jaisani Z, Wolinsky JS. Imaging of multiple interferon beta-1b in secondary progressive multiple sclerosis.
sclerosis: role in neurotherapeutics. NeuroRx 2005; 2: 277–303. Brain 2000; 123: 2256–63.
12 Bitsch A, Kuhlmann T, Stadelmann C, Lassmann H, Lucchinetti C, 33 Paolillo A, Coles AJ, Molyneux PD, et al. Quantitative MRI in
Bruck W. A longitudinal MRI study of histopathologically defined patients with secondary progressive MS treated with monoclonal
hypointense multiple sclerosis lesions. Ann Neurol 2001; 49: 793–96. antibody campath 1H. Neurology 1999; 53: 751–57.
13 van Walderveen MA, Kamphorst W, Scheltens P, et al. 34 Paolillo A, Pozzilli C, Gasperini C, et al. Brain atrophy in relapsing-
Histopathologic correlate of hypointense lesions on T1-weighted remitting multiple sclerosis: Relationship with ‘black holes’, disease
spin-echo MRI in multiple sclerosis. Neurology 1998; 50: 1282–88. duration and clinical disability. J Neurol Sci 2000; 174: 85–91.
14 Meier DS, Weiner HL, Khoury SJ, Guttmann CR. Magnetic 35 Bermel RA, Sharma J, Tjoa CW, Puli SR, Bakshi R.
resonance imaging surrogates of multiple sclerosis pathology and A semiautomated measure of whole-brain atrophy in multiple
their relationship to central nervous system atrophy. sclerosis. J Neurol Sci 2003; 208: 57–65.
J Neuroimaging 2004; 14 (3 suppl): 46–53.

http://neurology.thelancet.com Vol 5 February 2006 167


Review

36 Bermel RA, Bakshi R, Tjoa C, Puli SR, Jacobs L. Bicaudate ratio as 59 Simon JH, Schiffer RB, Rudick RA, Herndon RM. Quantitative
a magnetic resonance imaging marker of brain atrophy in multiple determination of MS-induced corpus callosum atrophy in vivo
sclerosis. Arch Neurol 2002; 59: 275–80. using MR imaging. AJNR Am J Neuroradiol 1987; 8: 599–604.
37 Trapp BD, Peterson J, Ransohoff R, Rudick R, Al E. Axonal 60 Benedict RH, Weinstock-Guttman B, Fishman I, Sharma J,
transection in the lesions of multiple sclerosis. N Engl J Med 1998; Tjoa CW, Bakshi R. Prediction of neuropsychological impairment
338: 278. in multiple sclerosis: Comparison of conventional magnetic
38 Simon JH, Kinkel RP, Jacobs L, Bub L, Simonian N. A Wallerian resonance imaging measures of atrophy and lesion burden.
degeneration pattern in patients at risk for MS. Neurology 2000; Arch Neurol 2004; 61: 226–30.
54: 1155–60. 61 Caon C, Zvartau-Hind M, Ching W, Lisak RP, Tselis AC, Khan OA.
39 Ge Y, Gonen O, Inglese M, Babb JS, Markowitz CE, Grossman RI. Intercaudate nucleus ratio as a linear measure of brain atrophy in
Neuronal cell injury precedes brain atrophy in multiple sclerosis. multiple sclerosis. Neurology 2003; 60: 323–25.
Neurology 2004; 62: 624–27. 62 Berg D, Maurer M, Warmuth-Metz M, Rieckmann P, Becker G.
40 De Stefano N, Iannucci G, Sormani MP, et al. MR correlates of The correlation between ventricular diameter measured by
cerebral atrophy in patients with multiple sclerosis. J Neurol 2002; transcranial sonography and clinical disability and cognitive
249: 1072–77. dysfunction in patients with multiple sclerosis. Arch Neurol 2000;
41 Enzinger C, Ropele S, Smith S, et al. Accelerated evolution of brain 57: 1289-1292.
atrophy and “black holes” in MS patients with APOE-epsilon 4. 63 Kallmann BA, Sauer J, Schliesser M, et al. Determination of
Ann Neurol 2004; 55: 563–69. ventricular diameters in multiple sclerosis patients with
42 Bakshi R, Dmochowski J, Shaikh ZA, Jacobs L. Gray matter T2 transcranial sonography (TCS): a two year follow-up study. J Neurol
hypointensity is related to plaques and atrophy in the brains of 2004; 251: 30–34.
multiple sclerosis patients. J Neurol Sci 2001; 185: 19–26. 64 Sharma J, Sanfilipo MP, Benedict RH, Weinstock-Guttman B,
43 Bakshi R, Benedict RH, Bermel RA, et al. T2 hypointensity in the Munschauer FE 3rd, Bakshi R. Whole-brain atrophy in multiple
deep gray matter of patients with multiple sclerosis: a quantitative sclerosis measured by automated versus semiautomated MR
magnetic resonance imaging study. Arch Neurol 2002; 59: 62–68. imaging segmentation. AJNR Am J Neuroradiol 2004; 25:
985–996.
44 Bakshi R, Benedict RH, Bermel RA, Jacobs L. Regional brain
atrophy is associated with physical disability in multiple sclerosis: 65 Sormani MP, Rovaris M, Valsasina P, Wolinsky JS, Comi G,
Semiquantitative magnetic resonance imaging and relationship to Filippi M. Measurement error of two different techniques for brain
clinical findings. J Neuroimaging 2001; 11: 129–36. atrophy assessment in multiple sclerosis. Neurology 2004;
62: 1432–34.
45 De Stefano N, Matthews PM, Filippi M, et al. Evidence of early
cortical atrophy in MS: relevance to white matter changes and 66 Pelletier D, Garrison K, Henry R. Measurement of whole-brain
disability. Neurology 2003; 60: 1157–62. atrophy in multiple sclerosis. J Neuroimaging 2004; 14 (3 suppl):
11–19.
46 Tiberio M, Chard DT, Altmann DR, et al. Gray and white matter
volume changes in early RRMS: a 2-year longitudinal study. 67 Sanfilipo MP, Benedict RH, Zivadinov R, Bakshi R. Correction for
Neurology 2005; 64: 1001–07. intracranial volume in analysis of whole brain atrophy in multiple
sclerosis: the proportion vs. residual method. Neuroimage 2004;
47 Sastre-Garriga J, Ingle GT, Chard DT, Ramio-Torrenta L,
22: 1732–43.
Miller DH, Thompson AJ. Grey and white matter atrophy in early
clinical stages of primary progressive multiple sclerosis. 68 Ge Y, Grossman RI, Udupa JK, et al. Brain atrophy in relapsing-
Neuroimage 2004; 22: 353–59. remitting multiple sclerosis and secondary progressive multiple
sclerosis: longitudinal quantitative analysis. Radiology 2000;
48 Sanfilipo MP, Benedict RHB, Weinstock-Guttman B, Bakshi R.
214: 665–70.
Gray and white matter brain atrophy and neuropsychological
impairment in multiple sclerosis. Neurology (in press). 69 Udupa JK, Odhner D, Samarasekera S, et al. 3DVIEWNIX: An
open, transportable, multidimensional, multimodality,
49 Sailer M, Fischl B, Salat D, et al. Focal thinning of the cerebral
multiparametric imaging software system. SPIE Proc 1994;
cortex in multiple sclerosis. Brain 2003; 126: 1734–44.
2164: 58–73.
50 Chard DT, Griffin CM, Rashid W, et al. Progressive grey matter
70 Smith SM, Zhang Y, Jenkinson M, et al. Accurate, robust, and
atrophy in clinically early relapsing-remitting multiple sclerosis.
automated longitudinal and cross-sectional brain change analysis.
Mult Scler 2004; 10: 387-391.
Neuroimage 2002; 17: 479–89.
51 Peterson JW, Bo L, Mork S, Chang A, Trapp BD. Transected
71 Pelletier D, Nelson SJ, Oh J, et al. MRI lesion volume
neurites, apoptotic neurons, and reduced inflammation in cortical
heterogeneity in primary progressive MS in relation with axonal
multiple sclerosis lesions. Ann Neurol 2001; 50: 389–400.
damage and brain atrophy. J Neurol Neurosurg Psychiatry 2003;
52 Cifelli A, Arridge M, Jezzard P, Esiri MM, Palace J, Matthews PM. 74: 950–52.
Thalamic neurodegeneration in multiple sclerosis. Ann Neurol
72 Jones CK, Riddehough A, Li DKB, Zhao G, Paty DW. MRI cerebral
2002; 52: 650.
atrophy in relapsing-remitting MS: results from the PRISMS trial.
53 Taylor I. Serial MRI in multiple sclerosis: A prospective pilot study Neurology 2001; 56: A379.
of lesion load, whole brain volume and thalamic atrophy.
73 Collins DL, Montagnat J, Zijdenbos AP, Evans AC, Arnold DL.
J Clin Neurosci 2004; 11: 153–58.
Automated estimation of brain volume in multiple sclerosis with
54 Bakshi R, Miletich RS, Kinkel PR, Emmet ML, Kinkel WR. High- BICCR. Lect Notes Comput Sci 2001; 141–47.
resolution fluorodeoxyglucose positron emission tomography
74 Hohol MJ, Guttmann CR, Orav J, et al. Serial neuropsychological
shows both global and regional cerebral hypometabolism in
assessment and magnetic resonance imaging analysis in multiple
multiple sclerosis. J Neuroimaging 1998; 8: 228–34.
sclerosis. Arch Neurol 1997; 54: 1018–25.
55 Filippi M, Rocca MA, Colombo B, et al. Functional magnetic
75 Udupa JK, Wei L, Samarasekera S, Miki Y, van Buchem MA,
resonance imaging correlates of fatigue in multiple sclerosis.
Grossman RI. Detection and quantification of MS lesions using
Neuroimage 2002; 15: 559–67.
fuzzy topological principles. Proc SPIE 1996; 2710: 81–91.
56 Inglese M, Ge Y, Filippi M, Falini A, Grossman RI, Gonen O.
76 Alfano B, Brunetti A, Larobina M, et al. Automated segmentation
Indirect evidence for early widespread gray matter involvement in
and measurement of global white matter lesion volume in patients
relapsing-remitting multiple sclerosis. Neuroimage 2004; 21:
with multiple sclerosis. J Magn Reson Imaging 2000; 12: 799–807.
1825–829.
77 Dalton CM, Brex PA, Jenkins R, et al. Progressive ventricular
57 Fabiano AJ, Sharma J, Weinstock-Guttman B, et al. Thalamic
enlargement in patients with clinically isolated syndromes is
involvement in multiple sclerosis: a diffusion-weighted magnetic
associated with the early development of multiple sclerosis.
resonance imaging study. J Neuroimaging 2003; 13: 307–14.
J Neurol Neurosurg Psychiatry 2002; 73: 141–47.
58 Simon JH. Linear and regional measures of brain atrophy in
78 Lukas C, Hahn HK, Bellenberg B, et al. Sensitivity and
multiple sclerosis. In: Zivadinov R, Bakshi R (eds). Brain and
reproducibility of a new fast 3D segmentation technique for clinical
spinal cord atrophy in multiple sclerosis. Hauppauge: Nova
MR-based brain volumetry in multiple sclerosis. Neuroradiology
Science, 2004: 15–27.
2004; 46: 906–15.

168 http://neurology.thelancet.com Vol 5 February 2006


Review

79 Fox NC, Jenkins R, Leary SM, et al. Progressive cerebral atrophy in 101 Filippi M, Rovaris M, Iannucci G, Mennea S, Sormani MP, Comi G.
MS: A serial study using registered, volumetric MRI. Neurology Whole brain volume changes in patients with progressive MS
2000; 54: 807–12. treated with cladribine. Neurology 2000; 55: 1714–18.
80 Smith SM, De Stefano N, Jenkinson M, Matthews PM. 102 Stevenson VL, Miller DH, Leary SM, et al. One year follow up study
Normalized accurate measurement of longitudinal brain change. of primary and transitional progressive multiple sclerosis.
J Comput Assist Tomogr 2001; 25: 466–75. J Neurol Neurosurg Psychiatry 2000; 68: 713–18.
81 Ashburner J, Friston KJ. Voxel-based morphometry: the methods. 103 Ingle GT, Stevenson VL, Miller DH, Thompson AJ. Primary
Neuroimage 2000; 11: 805–21. progressive multiple sclerosis: a 5-year clinical and MR study.
82 Chard DT, Parker GJ, Griffin CM, Thompson AJ, Miller DH. Brain 2003; 126: 2528–36.
The reproducibility and sensitivity of brain tissue volume 104 Leary SM, Miller DH, Stevenson VL, Brex PA, Chard DT,
measurements derived from an SPM-based segmentation Thompson AJ. Interferon beta-1a in primary progressive MS:
methodology. J Magn Reson Imaging 2002; 15: 259–67. an exploratory, randomized, controlled trial. Neurology 2003;
83 Kassubek J, Tumani H, Ecker D, Kurt A, Ludolph AC, 60: 44–51.
Juengling FD. Age-related brain parenchymal fraction is 105 Barkhof F. MRI in multiple sclerosis: correlation with expanded
significantly decreased in young multiple sclerosis patients: disability status scale (EDSS). Mult Scler 1999; 5: 283–86.
a quantitative MRI study. Neuroreport 2003; 14: 427–30. 106 Dastidar P, Heinonen T, Lehtimaki T, et al. Volumes of brain
84 Guttmann CRG, Benson R, Warfield SK, et al. White matter atrophy and plaques correlated with neurological disability in
abnormalities in mobility-impaired older persons. Neurology 2000; secondary progressive multiple sclerosis. J Neurol Sci 1999;
54: 1277–83. 165: 36–42.
85 Zivadinov R, Grop A, Sharma J, et al. Reproducibility and accuracy 107 Fisher E, Rudick RA, Simon JH, et al. Eight-year follow-up study of
of quantitative magnetic resonance imaging techniques of whole- brain atrophy in patients with MS. Neurology 2002; 59: 1412–20.
brain atrophy measurement in multiple sclerosis. J Neuroimaging 108 Benedict RH, Carone DA, Bakshi R. Correlating brain atrophy with
2005; 15: 27–36. cognitive dysfunction, mood disturbances, and personality disorder
86 Paolillo A, Piattella MC, Pantano P, et al. The relationship between in multiple sclerosis. J Neuroimaging 2004; 14 (3 suppl): 36–45.
inflammation and atrophy in clinically isolated syndromes 109 Janardhan V, Bakshi R. Quality of life and its relationship to brain
suggestive of multiple sclerosis: a monthly MRI study after triple- lesions and atrophy on magnetic resonance images in 60 patients
dose gadolinium-DTPA. J Neurol 2004; 251: 432–39. with multiple sclerosis. Arch Neurol 2000; 57: 1485–91.
87 Brex PA, Jenkins R, Fox NC, et al. Detection of ventricular 110 Bakshi R, Czarnecki D, Shaikh ZA, et al. Brain MRI lesions and
enlargement in patients at the earliest clinical stage of MS. atrophy are related to depression in multiple sclerosis. Neuroreport
Neurology 2000; 54: 1689–91. 2000; 11: 1153–58
88 McDonald WI, Compston A, Edan G, et al. Recommended 111 Amato MP, Ponziani G, Pracucci G, Bracco L, Siracusa G,
diagnostic criteria for multiple sclerosis: guidelines from the Amaducci L. Cognitive impairment in early-onset multiple
international panel on the diagnosis of multiple sclerosis. sclerosis: pattern, predictors, and impact on everyday life in a
Ann Neurol 2001; 50: 121–27. 4-year follow-up. Arch Neurol 1995; 52: 168–72.
89 Filippi M, Rovaris M, Inglese M, et al. Interferon beta-1a for brain 112 Rao SM, Leo GJ, Bernardin L, Unverzagt F. Cognitive dysfunction
tissue loss in patients at presentation with syndromes suggestive of in multiple sclerosis: frequency, patterns, and prediction. Neurology
multiple sclerosis: a randomised, double-blind, placebo-controlled 1991; 41: 685–91.
trial. Lancet 2004; 364: 1489–96. 113 Kujala P, Portin R, Ruutiainen J. The progress of cognitive decline
90 Zivadinov R, Sepcic J, Nasuelli D, et al. A longitudinal study of in multiple sclerosis: a controlled 3-year follow-up. Brain 1997;
brain atrophy and cognitive disturbances in the early phase of 120: 289–97.
relapsing-remitting multiple sclerosis. J Neurol Neurosurg Psychiatry 114 Beatty WW, Goodkin DE, Monson N, Beatty PA. Cognitive
2001; 70: 773–80. disturbances in patients with relapsing remitting multiple
91 Kalkers NF, Ameziane N, Bot JC, Minneboo A, Polman CH, sclerosis. Arch Neurol 1989; 46: 1113–19.
Barkhof F. Longitudinal brain volume measurement in multiple 115 Litvan I, Grafman J, Vendrell P, et al. Multiple memory deficits in
sclerosis: rate of brain atrophy is independent of the disease patients with multiple sclerosis: exploring the working memory
subtype. Arch Neurol 2002; 59: 1572–76. system. Arch Neurol 1988; 45: 607–10.
92 Redmond IT, Barbosa S, Blumhardt LD, Roberts N. Short-term 116 Grant I, McDonald WI, Trimble MR, Smith E, Reed R. Deficient
ventricular volume changes on serial MRI in multiple sclerosis. learning and memory in early and middle phases of multiple
Acta Neurol Scand 2000; 102: 99–105. sclerosis. J Neurol Neurosurg Psychiatry 1984; 47: 250–55.
93 Inglese M, Rovaris M, Giacomotti L, Mastronardo G, Comi G, 117 Demaree HA, DeLuca J, Gaudino EA, Diamond BJ. Speed of
Filippi M. Quantitative brain volumetric analysis from patients with information processing as a key deficit in multiple sclerosis:
multiple sclerosis: a follow-up study. J Neurol Sci 1999; 171: 8–10. implications for rehabilitation. J Neurol Neurosurg Psychiatry
94 Zivadinov R, Bakshi R. Role of MRI in multiple sclerosis II: brain 1999; 67: 661–63.
and spinal cord atrophy. Front Biosci 2004; 9: 647–64. 118 Rao SM, Glatt S, Hammeke TA, et al. Chronic progressive multiple
95 Lin X, Blumhardt LD, Constantinescu CS. The relationship of brain sclerosis. relationship between cerebral ventricular size and
and cervical cord volume to disability in clinical subtypes of neuropsychological impairment. Arch Neurol 1985; 42: 678–82.
multiple sclerosis: a three-dimensional MRI study. 119 Rovaris M, Filippi M. MRI correlates of cognitive dysfunction in
Acta Neurol Scand 2003; 108: 401–06. multiple sclerosis patients. J Neurovirol 2000; 6 (suppl 2): 172–75.
96 Losseff NA, Wang L, Lai HM, et al. Progressive cerebral atrophy in 120 Huber SJ, Bornstein RA, Rammohan KW, Christy JA,
multiple sclerosis: a serial MRI study. Brain 1996; 119: 2009–19. Chakeres DW, McGhee RB. Magnetic resonance imaging
97 Ge Y, Grossman RI, Udupa JK, et al. Glatiramer acetate (Copaxone) correlates of neuropsychological impairment in multiple sclerosis.
treatment in relapsing-remitting MS: quantitative MR assessment. J Neuropsychiatry Clin Neurosci 1992; 4: 152–58.
Neurology 2000; 54: 813–17. 121 Pelletier J, Suchet L, Witjas T, et al. A longitudinal study of callosal
98 Turner B. Cerebral atrophy and disability in relapsing-remitting and atrophy and interhemispheric dysfunction in relapsing-remitting
secondary progressive multiple sclerosis over four years. Mult Scler multiple sclerosis. Arch Neurol 2001; 58: 105–11.
2003; 9: 21–27. 122 Zorzon M, Zivadinov R, Nasuelli D, et al. Depressive symptoms
99 Wolinsky JS, Narayana PA, Johnson KP, Multiple Sclerosis Study and MRI changes in multiple sclerosis. Eur J Neurol 2002;
Group and the MRI Analysis Center. United states open-label 9: 491–96.
glatiramer acetate extension trial for relapsing multiple sclerosis: 123 Feinstein A, Roy P, Lobaugh N, Feinstein K, O’Connor P, Black S.
MRI and clinical correlates. multiple sclerosis study group and the Structural brain abnormalities in multiple sclerosis patients with
MRI analysis center. Mult Scler 2001; 7: 33–41. major depression. Neurology 2004; 62: 586–90.
100 Gasperini C, Paolillo A, Giugni E, et al. MRI brain volume changes 124 Christodoulou C, Krupp LB, Liang Z, et al. Cognitive performance
in relapsing-remitting multiple sclerosis patients treated with and MR markers of cerebral injury in cognitively impaired MS
interferon beta-1a. Mult Scler 2002; 8: 119–123. patients. Neurology 2003; 60: 1793–98.

http://neurology.thelancet.com Vol 5 February 2006 169


Review

125 Benedict RH, Zivadinov R, Carone DA, et al. Regional lobar 136 Bakshi R, Dandamudi VSR, Neema M, De C, Bermel RA.
atrophy predicts memory impairment in multiple sclerosis. Measurement of brain and spinal cord atrophy by magentic
AJNR Am J Neuroradiol 2005; 26: 1824–31. resonance imaging as a tool to monitor multiple sclerosis.
126 Wei X, Guttmann CR, Warfield SK, Eliasziw M, Mitchell JR. Has J Neuroimaging 2005; 15 (4 suppl): 30S–45S.
your patient’s multiple sclerosis lesion burden or brain atrophy 137 Hardmeier M, Wagenpfeil S, Freitag P, et al. Rate of brain atrophy
actually changed? Mult Scler 2004; 10: 402–06. in relapsing MS decreases during treatment with IFNbeta-1a.
127 Horsfield MA, Rovaris M, Rocca MA, et al. Whole-brain atrophy in Neurology 2005; 64: 236–40.
multiple sclerosis measured by two segmentation processes from 138 Randomised double-blind placebo-controlled study of interferon
various MRI sequences. J Neurol Sci 2003; 216: 169–77. beta-1a in relapsing/remitting multiple sclerosis: PRISMS
128 Gasperini C, Rovaris M, Sormani MP, et al. Intra-observer, inter- (prevention of relapses and disability by interferon beta-1a
observer and inter-scanner variations in brain MRI volume subcutaneously in multiple sclerosis) study group. Lancet 1998;
measurements in multiple sclerosis. Mult Scler 2001; 7: 27–31. 352: 1498–504.
129 Gordon N. Apparent cerebral atrophy in patients on treatment with 139 Secondary Progressive Efficacy Clinical Trial of Recombinant
steroids. Dev Med Child Neurol 1980; 22: 502–06. Interferon-beta-1a in MS (SPECTRIMS) Study Group: randomized
130 Hoogervorst EL, Polman CH, Barkhof F. Cerebral volume changes controlled trial of interferon- beta-1a in secondary progressive
in multiple sclerosis patients treated with high-dose intravenous MS—clinical results. Neurology 2001; 56: 1496–504.
methylprednisolone. Mult Scler 2002; 8: 415–19. 140 Lin X, Tench CR, Turner B, Blumhardt LD, Constantinescu CS.
131 Rao AB, Richert N, Howard T, et al. Methylprednisolone effect on Spinal cord atrophy and disability in multiple sclerosis over four
brain volume and enhancing lesions in MS before and during years: application of a reproducible automated technique in
IFNbeta-1b. Neurology 2002; 59: 688–94. monitoring disease progression in a cohort of the interferon beta-
1a (rebif) treatment trial. J Neurol Neurosurg Psychiatry 2003;
132 Fox RJ, Fisher E, Tkach J, Lee JC, Cohen JA, Rudick RA. Brain
74: 1090–94.
atrophy and magnetization transfer ratio following
methylprednisolone in multiple sclerosis: short-term changes and 141 Frank JA, Richert N, Bash C, et al. Interferon-beta-1b slows
long-term implications. Mult Scler 2005; 11: 140–45. progression of atrophy in RRMS: Three-year follow-up in NAb
and NAb patients. Neurology 2004; 62: 719–25.
133 Zivadinov R, Rudick RA, De Masi R, et al. Effects of IV
methylprednisolone on brain atrophy in relapsing-remitting MS. 142 Miller DH. Biomarkers and surrogate outcomes in
Neurology 2001; 57: 1239–47. neurodegenerative disease: lessons from multiple sclerosis.
NeuroRx 2004; 1: 284–94
134 Rudick RA, Fisher E, Lee JC, Duda JT, Simon J. Brain atrophy in
relapsing multiple sclerosis: Relationship to relapses, EDSS, and 143 Biernacki K, Antel JP, Blain M, Narayanan S, Arnold DL, Prat A.
treatment with interferon beta-1a. Mult Scler 2000; 6: 365–72. Interferon beta promotes nerve growth factor secretion early in the
course of multiple sclerosis. Arch Neurol 2005; 62: 563–68.
135 Pato-Pato A, Prieto JM, Lema M, Dapena-Bolano D, Abella-Corral J,
Pumar JM. Cerebral atrophy in multiple sclerosis patients treated 144 Yong VW, Chabot S, Stuve O, Williams G. Interferon beta in the
periodically with boluses of methylprednisolone. Rev Neurol 2003; treatment of multiple sclerosis: Mechanisms of action. Neurology
37: 501–06. 1998; 51: 682–89.

170 http://neurology.thelancet.com Vol 5 February 2006

You might also like