You are on page 1of 12

REvIEwS

Chronic inflammation in multiple


sclerosis — seeing what was always
there
Paul M. Matthews   1,2
Abstract | Activation of innate immune cells and other compartmentalized inflammatory cells
in the brains and spinal cords of people with relapsing–remitting multiple sclerosis (MS) and
progressive MS has been well described histopathologically. However, conventional clinical MRI is
largely insensitive to this inflammatory activity. The past two decades have seen the introduction
of quantitative dynamic MRI scanning with contrast agents that are sensitive to the reduction in
blood–brain barrier integrity associated with inflammation and to the trafficking of inflammatory
myeloid cells. New MRI imaging sequences provide improved contrast for better detection of
grey matter lesions. Quantitative lesion volume measures and magnetic resonance susceptibility
imaging are sensitive to the activity of macrophages in the rims of white matter lesions. PET and
magnetic resonance spectroscopy methods can also be used to detect contributions from innate
immune activation in the brain and spinal cord. Some of these advanced research imaging
methods for visualization of chronic inflammation are practical for relatively routine clinical
applications. Observations made with the use of these techniques suggest ways of stratifying
patients with MS to improve their care. The imaging methods also provide new tools to support
the development of therapies for chronic inflammation in MS.

Multiple sclerosis (MS) is a chronic, inflammatory, T cells10. Chronic CNS inflammation seems to play a
demyelinating neurodegenerative disease1. The diagno- major role in initiating the neurodegenerative process11,12
sis and monitoring of relapsing–remitting MS (RRMS) in RRMS and progressive forms of MS13,14. However,
was transformed by the discovery in 1981 that MRI is conventional clinical imaging is largely insensitive to this
sensitive to the white matter lesions of MS2 together with chronic inflammation, making it difficult to characterize
subsequent more detailed pathological correlations of and monitor in living patients.
the MRI signal with the acute inflammatory oedema, The focus of this Review is advanced imaging meth-
demyelination and gliosis associated with these MS ods for assessing chronic inflammation in MS. I first
lesions throughout the white matter of the CNS3,4. The review the major neuropathological features of these
use of MRI as an outcome measure in early-​phase clin- inflammatory processes, emphasizing that they have
ical trials reduced the risks of drug development and long been a well-​recognized aspect of MS neuropath­
accelerated the development of new treatments5,6. ology. I then discuss advanced imaging methods and
Radiological correlates of the inflammatory episodes describe how they can be used to visualize this inflam-
that are particularly characteristic of RRMS have been mation. These newer research imaging methods have
the major focus for clinical MRI studies to date, but the the potential to define relationships between chronic
1
Department of Brain neuropathology of MS also highlights chronic inflam- inflammatory activity, measures of neurodegenera-
Sciences, Department of mation as a feature of all clinical stages of the disease. tion and clinical progression in life. They also promise
Medicine, Imperial College This chronic inflammation arises from activation of to enable better clinical management of MS through
London, London, UK.
innate and adaptive immune responses that are relatively patient stratification for treatment and by facilitating the
2
UK Dementia Research compartmentalized in the CNS. Many cell types con- development of new therapies.
Institute, Imperial College
London, London, UK.
tribute to the innate immune response in the CNS, but
microglia, macrophages and astrocytes are numerically Chronic inflammation in MS
e-​mail: p.matthews@
imperial.ac.uk most important7. Subtypes of adaptive immune cells can Most of what has been described as the neuropathology
https://doi.org/10.1038/ become compartmentalized in the CNS as lymphoid-​like of MS comes from studies on brain or spinal cord tissue
s41582-019-0240-y leptomeningeal follicles8,9 or as tissue-​resident memory from patients with secondary progressive MS (SPMS).

Nature Reviews | Neurology


Reviews

Key points molecule 1. Inflammatory cells can also enter the brain
through the choroid plexus, where expression levels of
• Advanced MRI and PET methods enable visualization of features related to chronic VCAM-1 are particularly high in brains from people
inflammation in progressive and relapsing–remitting forms of multiple sclerosis (MS). with progressive MS, even those with later progressive
• Quantitative analysis of uptake of gadolinium contrast agent and ultra-​small disease28.
paramagnetic particles provide in vivo evidence of chronic, low-​grade inflammation Soluble forms of the adhesion molecules E-selectin,
in people with progressive or relapsing–remitting MS (RRMS). L-selectin, intracellular adhesion molecule 1 and
• Lesions associated with activated macrophages/microglia (slowly expanding T2 VCAM-1 can be measured in the blood of people with
hyperintense lesions and lesions with high susceptibility-​weighted MRI signals at progressive MS29 and are presumed to reflect their
their rims) are more common in progressive MS than in RRMS.
relative endo­thelial expression. Measurement of solu­
• Persistent focal leptomeningeal inflammation, detectable with gadolinium contrast-​ ble VCAM-1 has shown that levels in the blood of
enhanced T2 fluid attenuation inversion recovery MRI in many people with MS
patients with PPMS are higher than those in patients
(particularly progressive MS), is associated with cortical lesions and accelerated
cortical atrophy.
with RRMS29.
Postmortem data also suggest that the integrity of the
• Translocator protein PET can detect increased innate immune activation in brains of
people with MS; typically, activation is greater in secondary progressive MS than
BBB is chronically impaired throughout the course of
in RRMS. MS30. For example, tight junction abnormalities are only
• Indirect evidence suggests that magnetic resonance spectroscopy measures of
moderately less frequent in lesions that exhibit some
myo-​inositol and some recently introduced PET measures can reflect contributions inflammatory activity but appear to be chronic than in
of astrocyte activation to brain innate immune responses. lesions that exhibit active inflammation that appears to
be of recent onset31. Evidence of chronic BBB impair-
ment, such as abnormal tight junctions, can also be
The neuropathology of MS has traditionally been found in normal-​appearing grey matter32. Furthermore,
considered to involve neuroinflammation and demy- fibrinogen can be seen around vessels at the edges of
elination with relative neuroaxonal sparing15. The char- mixed active–inactive lesions, an observation that, in
acteristic, multifocal white matter demyelination lesions association with histopathological evidence of ongoing
are perivenular, with inflammatory activity that is asso- demyelination and inflammation, suggests that blood
ciated with leukocytes, reactive astrocytes, microg­lia and proteins leak into the CNS and help to sustain chronic
myelin-​laden macrophages in various combinations16,17. inflammation33,34.
The leukocyte infiltrates are dominated by T cells, As important as understanding the pathways by
accompanied by a variable number of B cells, mono- which peripheral inflammatory cells enter the brain is
cytes and occasional plasma cells. However, in addition defining how antigen-​presenting cells leave the CNS
to these classical features, substantial involvement of the to activate a peripheral immune response. Important
grey matter is now recognized18–20, as is clinically signifi- insights into mechanisms of this process came with
cant neuroaxonal injury and loss from the earliest stages the discovery of a rudimentary CNS lymphatic system
of the disease21–23. associated with the dural venous sinuses that allows
migration of dendritic cells and some T cells for antigen
The blood–brain barrier presentation in deep cervical lymph nodes35,36. Memory
Interactions between the peripheral immune system and B cells trafficking between cervical lymph nodes and
any compartmentalized CNS immune responses depend the CNS parenchyma probably also use this route,
on interactions between inflammatory cells or circulat- which might become more functionally developed as a
ing signalling factors and the blood–brain barrier (BBB). consequence of chronic CNS inflammation26. Reverse
The endothelial cells at the BBB form tight junctions and flow in the paravascular glymphatic system could also
have low pinocytotic activity, enabling them to control contribute to the transport of brain antigenic proteins
the movement of peripheral inflammatory cells and to the blood37,38.
molecules from the blood into the CNS24. The classical
pathway of T cell migration into the CNS involves direct Leptomeningeal inflammation
interactions between the T cells and the endothelium of Meningeal B cells, which can form ectopic lymphoid-​
postcapillary blood vessels. like aggregates, could play major roles in cortical
In the brains of people with primary progressive MS inflammatory demyelination and neurodegeneration39
(PPMS) or SPMS, inflammatory markers and adhesion (Fig. 1). Serum and cerebrospinal fluid levels of CXCL13,
molecules are substantially upregulated in the microvas- which is believed to support the aggregation of the
cular endothelium25. This upregulation marks activation B cells 40, are elevated in patients with RRMS and
of endothelial cells at the BBB, which is a major mech- SPMS41,42. Leptomeningeal inflammation observed in
anism that facilitates infiltration of peripheral immune MS ranges from disorganized collections of immune
cells into the CNS26. This complex process involves coor- cells in the meninges of patients with RRMS or PPMS
dinated expression of multiple cell adhesion molecules to well-​organized ectopic lymphoid follicles observed
on the luminal surface of endothelial cells27. Expression postmortem in brains from some people with SPMS39.
of the inflammatory activation marker human leukocyte Ectopic leptomeningeal lymphoid tissue is found at
antigen DR isotype and vascular cell adhesion molecule 1 autopsy in the brains of ~40% of people with SPMS9. Its
(VCAM-1) can occur in as many as half of the cerebral presence is associated with aggressive disease and more
microvessels in patients with MS; only moderately fewer extensive cortical demyelination and neurodegenera-
of these microvessels express intracellular adhesion tion8,43. In addition to the almost complete demyelination

www.nature.com/nrneurol
Reviews

lesion display a predominantly pro-​inflammatory phe-


notype52,53 (with expression of markers such as CD68,
CYBA, major histocompatibi­lity complex class II anti-
gens and ferritin). During active demyelination, myelin
breakdown products, including iron, accumulate in these
macrophages54. Microglia can also be found in the cen-
tres of mixed active–inactive lesions, but these microg­
lia, activity of which might facilitate remyelination55,
are relatively sparse and adopt a predominantly anti-​
inflammatory phenotype56. Activated astrocytes, which
have functions intimately related to those of microglia,
are also prominent57 in mixed active–inactive lesions.
Chronic inflammation persists in white matter lesions
throughout the course of MS. Although brains from
100 μm patients with progressive forms of MS have fewer active
lesions in the white matter than those from patients
Fig. 1 | leptomeningeal inflammation in multiple sclerosis. A photomicrograph that with RRMS, a range of active, mixed active–inactive
shows binding of a B cell immunohistochemical marker (anti-​CD20 antibody). A dense and inactive lesions can be seen in postmortem brains
meningeal cluster of B cells (arrows) overlies the cortical grey matter (arrow heads). This of almost all people with MS58. Mixed active–inactive
cluster is an example of the lymphoid-​like meningeal B cell foci that appear to be chronic plaques are most abundant in brains from people whose
and that are associated with demyelination and neuronal injury and loss in the adjacent
disease duration was >10 years and who were >50 years
cortex. Image courtesy of R . Reynolds, Imperial College London, UK.
of age at death59,60. Even in patients with the longest dis-
ease duration (42–64 years), one study reported that
in affected regions, loss of neurons in adjacent cortex can just over one-​third of the lesions had an active or mixed
also be substantial. The reported postmortem neuronal active–inactive phenotype60.
density in the cortex is almost 40% lower in brains from Microglia that express tumour microenvironment of
people with MS than in those from matched controls metastasis 119 (TMEM119), which are neurodevelop-
(14.9 billion neurons per section in MS versus 24 billion mentally distinct from macrophages because they are
per section in brains without evidence of neurological derived from yolk sac progenitors61, are found in the
disease)44. Loss of synapses is even more marked than nonlesional white matter of brains from people with
the loss of neurons45,46. RRMS or progressive forms of MS. In the late stages
of MS, these microglia have a phenotype that is inter-
White and grey matter mediate between homeostatic and pro-​inflammatory
Lymphocytes in the brain parenchyma contribute (which is sometimes described as ‘primed’), defined
to chronic inflammation47,48. Most of the T cells that by coexpression of CD68 and major histocompatibility
are recruited to inflammatory foci have an activated complex class II antigen56. They are often found in small
(cytotoxic) phenotype and are presumed to be short-lived. clusters that are diffusely scattered in the white matter62.
However, some convert into tissue-​resident effector These clusters of activated microglia are sometimes
memory T cells10 that persist owing to loss of expres- found in association with complement and degenerat-
sion of the surface receptors that normally facilitate the ing axons, suggesting that the microglia have adopted
egress of cells (the spingosine-1-phosphate receptor 1 a pro-​inflammatory state63,64, although microglia in
(S1P1) and CCR7). Upregulation of the integrin CD103 the nonlesional white matter might also clear damaged
might also contribute to compartmentalization of these neurons and other cellular debris65.
cells in the CNS49. An inflammatory trigger can cause
these memory T cells to differentiate into cytotoxic Ageing and inflammation
T cells. B cells that can differentiate into plasmablasts The potential changes in the immunopathological
and plasma cells are also found in the brain parenchyma mechanisms of MS with ageing are wide-​ranging66.
of people with MS. These chronically tissue-​resident A comprehensive discussion of this complex and poorly
leukocytes might be able to initiate a new inflammatory understood area is beyond the scope of this Review, but
focus when inflammatory tissue injury exposes antigens it is important to highlight two general and seemingly
that activate these cells10. contradictory aspects. One is immunosenescence, which
Despite these contributions of lymphocytes, white has been attributed, at least in part, to decreases in T cell
matter lesions in MS are classified neuropathologi- responsiveness with ageing67,68. However, the other is
cally on the basis of the nature and extent of microglia/ an increase in inflammatory function with ageing; for
macrophage-​mediated inflammation and demyelina- example, microglia in the healthy human brain assume a
tion50. Lesions with signs of ongoing demyelination and more pro-​inflammatory activation state with increasing
with macrophages and microglia throughout their vol- age69. Broadly consistent with this observation, epide-
ume are classified as active lesions. Lesions with central miological evidence indicates an age-​related increase
demyelination, or thin myelin that is associated with in incident progressive disease and greater severity of
remyelination51, and with macrophages/microglia pre- disease with later onset70.
dominantly at their borders are classified as mixed active– The apparent contradiction between immunosenes-
inactive lesions (Fig. 2). The macrophages in both types of cence and relative inflammatory activation might be

Nature Reviews | Neurology


Reviews

evidence for persistent inflammation discussed above


and also neglects early longitudinal observations with
gadolinium contrast MRI, which identified an average
of more than one enhancing lesion per person per year
among patients with SPMS, although fewer among
patients with PPMS81. Although the frequency of gad-
olinium enhancing lesions among people with SPMS
is substantially lower than among those with RRMS, it is
high enough that clinical studies of highly effective treat-
ments that suppress inflammation are plausible82. These
imaging studies might not fully represent observations
for the most commonly encountered patients today, but
they highlight the potential for inflammatory lesion
activity even in progressive forms of MS.
Conventional clinical MRI has unquestionably rev-
olutionized the diagnosis and monitoring of MS3,4, but
is relatively insensitive to the chronic inflammation in
200 µm grey and white matter that is so prominent on postmor-
tem neuropathology. The past two decades have seen
Fig. 2 | Macrophages in a mixed active–inactive the introduction of new research imaging methods to
multiple sclerosis lesion. A photomicrograph that address this challenge. Quantitative dynamic MRI with
shows a mixed active–inactive lesion from a patient with contrast agents is sensitive to the reduction of BBB integ-
secondary progressive multiple sclerosis. A dense rim of rity associated with inflammation and to trafficking of
highly activated major histocompatibility complex class II-
inflammatory myeloid cells into the brain parenchyma
positive microglia (yellow arrows) can be seen around a
central demyelinated core (white arrows). Image courtesy from the blood. MRI imaging sequences83–85 provide
of R . Reynolds, Imperial College London, UK. greater contrast for grey matter lesions. Quantitative
lesion volume measures, T2* MRI and susceptibility
imaging are sensitive to the activity of macrophages in
attributed to age-​related changes in immune response the rims of white matter lesions. PET and magnetic res-
network interactions. Circulating, adaptive immune onance spectroscopy (MRS) methods can also be used to
cells that are activated by CNS dendritic cells and that assess innate immune activation in the brain (or spinal
are consequently trafficked into the CNS enhance cord). Together, these methods are beginning to make
CNS compartmentalized immune responses56,71. This the major neuropathological features of chronic CNS
process might be facilitated even by normal ageing of inflammation observable in vivo (Box 1). By using them
the immune system, such as decreased systemic regu- in large numbers of patients, direct correlations can be
latory T cell function and age-​related lymphopenia72. made between changes in chronic inflammation and
Leukocyte trafficking to the CNS might be increased clinical states.
further by environmental exposures or comorbidities
(for example, neurovascular inflammation associated Impairment of the blood–brain barrier
with small vessel disease)73,74 that lead to endothelial Gadolinium contrast enhancement (of acute and active
activation75–77. white matter lesions) is usually assessed qualitatively in
Repeated systemic inflammatory episodes (for exam- the clinic but, in a research setting, quantitative meas-
ple, severe infections) increase the relative abundance ures of change over time after injection of the contrast
of pro-​inflammatory microglia78. Experiments with agent can be made from serial, rapidly acquired images86.
a rodent model have provided novel evidence for a Quantitative analyses enable detection of the disruption
long-​lasting, epigenetically mediated microglial mem- to BBB integrity that accompanies chronic, low-​grade
ory of recurrent systemic inflammation79. Interactions inflammation, which is less severe than that associated
between microglia and astrocytes might contribute to with acute inflammation. After careful spatial align-
this phenom­enon, as functional changes in astrocytes ment of the serially acquired images, small brain signal
relevant to their roles in innate immune responses are changes over time after the injection of contrast agent
regulated by microRNA changes with ageing80. One goal into the blood can be fitted to a diffusion model that
in the use of advanced imaging in MS is to monitor not quantitatively reflects the permeability of the BBB to
just the extent of chronic brain immune activation and the contrast agent. These measures are sensitive to small
the associated cellular phenotypes, but also the traffick- impairments of venule barrier function that are believed
ing of inflammatory cells from the blood into the CNS to lead to the leakage of fibrinogen and other proteins
that contribute to modulation of this immune activation. seen in neuropathological studies33,34. In this way, multi-
ple lesions in a single brain can be differentiated on the
Imaging of chronic inflammation in MS basis of differences in permeability to gadolinium con-
The absence or low frequency of clinical relapses in trast agent86. Chronic inflammation in non-​enhancing
SPMS and PPMS has often been interpreted clini­cally lesions in people with either RRMS or progressive MS
as evidence that CNS inflammation is relatively absent. is associated with persistently increased permeability
However, this view does not reflect the neuropathological to gadolinium contrast agent87. Similar small, chronic

www.nature.com/nrneurol
Reviews

increases in contrast agent permeability can be found in that contain iron (which is released inside the cells on
the deep grey matter88. phagocytosis of myelin)93,94. Consequently, magnetic
Quantitative gadolinium measures are not yet useful resonance susceptibility imaging provides an indirect
or practical as clinical tools for assessing chronic inflam- approach to their identification in vivo95. Changes in
mation in MS. To become useful, clinically meaningful myelin and iron content alter the local magnetic sus-
differences in contrast agent permeability and how they ceptibility and, as a result, the so-​called T2* relaxation
reflect changes in the associated inflammatory patho­l­ time. On R2* (1/T2*) images, the core of most white
ogy need to be defined. Another important considera- matter lesions is hypointense, predominantly reflecting
tion is the benefit of gadolinium contrast relative to its the loss of iron-​rich myelin, which normally appears as
potential harm; concerns among doctors and regulators enhancement on R2* images. Following myelin break-
include the rare adverse event of nephrogenic systemic down, paramagnetic iron that is phagocytosed by mac-
sclerosis, which can occur in people with reduced renal rophages/microglia at the borders of demyelinating
function, and the potential long-​term adverse effects lesions increases the local R2* signal at the borders.
of gadolinium accumulation in tissues (including the Lesions with an isointense core and a hyperintense bor-
brain) with repeated contrast administration89,90. der on R2* images are found more commonly in the
brains of people with progressive MS than in those with
Lesion-​associated inflammation RRMS96.
Quantitative measurements of longitudinal changes in With serial imaging, lesions that have hyperintense
the sizes of individual white matter lesions provide borders on R2* images are more likely to expand over
in vivo evidence for chronic inflammation that could be time than are lesions without such borders. These lesions
more widely available for routine clinical assessments with border-​associated magnetic susceptibility changes
than quantitative gadolinium enhancement measures. have been characterized best with 7 T MRI, as the dif-
Precise assessment of lesion volumes with sensitive, ferences in magnetic susceptibility are easier to detect
computer-​based analyses of paired, serial MRI scans with higher magnetic fields, but smaller magnetic sus-
enables monitoring of the slow expansion of some T2 ceptibility effects can also be detected with clinically
hyperintense lesions91 (Fig. 3). These slowly expanding available 3 T MRI95. Detection of R2* hyperintense
lesions do not typically show conventional gadolinium lesion borders enables identification of patients with
enhancement and tend to have a lower intensity than active inflammation defined in this way, even with a
non-​expanding lesions on T1 images, probably reflect- single scan. Identification of R2* hyperintense borders
ing greater parenchymal rarefaction owing to axonal and slowly expanding lesions91 provide (at least partially)
loss92. These lesions are more common in PPMS than independent markers of chronic inflammation. When
in RRMS91. used together, they could improve sensitivity for chronic
The neuropathological correlates of these expanding inflammation and enable stratification of people with
lesions have rims enriched with macrophages/microglia progressive MS on the basis of this evidence for active,
chronic inflammation.
A different MRI-​based approach to identification of
Box 1 | Methods for in vivo imaging of chronic inflammation in multiple sclerosis
chronic lesion-​associated inflammation involves detec-
For each of the chronic inflammatory neuropathologies specified below, the imaging tion of brain MRI signal enhancement after intravenous
methods that can be used to visualize the chronic inflammation in vivo are listed. administration of ultra-​small paramagnetic iron oxide
New lesion activity nanoparticles (USPIO)97. The nanoparticles are phago-
• Gadolinium-​enhanced T1 MRI3,4,98 cytosed by activated tissue-​resident phagocytes and
• Longitudinal T2-weighted or fluid attenuated inversion recovery (FLAIR) MRI3,4
circulating myeloid effector cells that migrate from the
blood to white matter lesions. Enhancing lesions then
Mixed active–inactive lesions show high signal intensities on T1-weighted images
• Enhancement with dynamic gadolinium contrast MRI86–88 (and low signal intensities on T2-weighted images)
• Quantitative longitudinal measures of T2 hyperintense lesion growth91 with gadolinium and USPIO if both contrast agents are
• T2* or susceptibility-​weighted MRI93–96 used. However, gadolinium and iron oxide nanoparticles
• Ultra-​small paramagnetic iron particle contrast (USPIO) enhancement97–100 reveal different types of BBB impairment; lesions can be
found that enhance with only gadolinium, only USPIO,
Cortical demyelinating lesions or both97. Lesions that enhance with both contrast agents
• 3D double inversion recovery or phase-​sensitive inversion recovery MRI83,85,112 are larger and more likely to persist in a 6-month follow-​
• 3D magnetization-​prepared rapid gradient echo MRI84 up scan98,99. These lesions might, therefore, be those in
• Coregistration and multiplication of conventional 3D-​T2 weighted and 3D-​FLAIR107 which BBB impairment is most severe and myeloid
• Ultra-​high field 3D-​FLAIR108 macrophage infiltration — which is associated with pro-​
inflammatory macrophages that directly attack myelin
leptomeningeal inflammation — is greatest100.
• Leptomeningeal enhancement on T2-FLAIR MRI with gadolinium contrast117,119,121 USPIO contrast enhancement thus provides another
Innate immune inactivation novel potential tool for assessment of chronic inflam-
• Translocator protein (TSPO) PET125,126,128,129,131 matory activity in MS. Uniquely among the imaging
methods for the monitoring of MS, USPIO contrast
• 11C-​acetate PET138
enhancement also provides a measure of inflammatory
• Myo-​inositol proton magnetic resonance spectroscopy143–145
cell trafficking from the blood into the CNS. However,

Nature Reviews | Neurology


Reviews

several reports have described serious, life-​threatening lesion detection can be improved by use of 3D double
anaphylactic reactions to USPIO administered for iron inversion recovery83, the related 3D magnetization-​
supplementation101,102 and the nanoparticles are taken prepared rapid gradient echo method84 or phase sensi-
up and retained in several organs, the long-​term health tive inversion recovery MRI85, as they provide greater
risks of which cannot be assessed easily. The FDA has contrast between lesions and healthy-​appearing grey
issued a black box warning regarding these risks103. matter. Cortical lesions can also be detected with sim-
Consequently, the current generation of USPIO agents ilar sensitivity by coregistration and multiplication of
does not offer a favourable risk–benefit balance for rou- conventional 3D T2-weighted and 3D-​fluid attenua-
tine clinical use as an approach for stratification based tion inversion recovery (FLAIR; for cerebrospinal fluid
on disease activity in MS. However, potentially safer, less suppression) images107. However, the major challenge
immunogenic USPIO nanoparticles and related agents is spatial resolution; the higher resolution of 7 T MRI
are under development104. promises to provide future opportunities for improved
cortical lesion detection108.
Cortical MS lesions Despite these limitations, the numbers of cortical
Cortical MS lesions that have been seen with use of lesions visible with 3 T MRI can be counted and related
postmortem histopathological methods can be iden- to the clinical course of patients with MS. Generally,
tified with MRI, but they cannot be defined with high cortical lesions identified with MRI seem to be most
sensitivity because the cortex is thin relative to conven- frequent in the hippocampus and the temporal and
tional imaging voxel dimensions and the contrast of the frontal lobes (particularly the motor and cingulate
lesions relative to normal-​appearing cortical grey matter cortices)109,110. These cortical lesions progress inde-
is low83,105. An early study suggested that <10% of cor- pendently of white matter lesions111 and cross-​sectional
tical lesions that can be seen histopathologically could data indicate that they are associated with greater local
be identified with clinical MRI — juxtacortical lesions cortical atrophy112, more severe clinical presentations,
were more frequently detected106. Even with side-​by-side more rapid progression and greater disability (particu-
review of the magnetic resonance images and postmor- larly disability related to cognitive performance)113,114.
tem histopathology, many of the intrinsic cortical lesions Nevertheless, on average, the rates at which new cor-
could not be identified on the images105. Intracortical tical lesions develop are similar in people with RRMS
and people with SPMS115, and the rate in PPMS is also
comparable116.
a T1-weighted b T1-weighted
Postmortem studies have identified leptomeningeal
inflammation that is associated with extensive subpial
cortical demyelinating lesions and neurodegenera-
tion9,117. Related observations have been made with
MRI in vivo. One patient with MS who died soon after a
routine MRI scan that showed persistent leptomeningeal
enhancement had a pattern of demyelination that was
typical for a type III subpial lesion in the adjacent cor-
tex105. In patients with RRMS, focal leptomeningeal con-
trast enhancement is associated with reduced thickness
of the adjacent cortex118. Long-​lasting, focal gadolinium
contrast enhancement of the meninges can be seen with
MRI in a considerable proportion of patients with MS119.
T2-weighted T2-weighted Although this enhancement is not diagnostically specific
for MS119, its presence in people with MS does provide
evidence for active inflammatory pathology. Reliable
detection of this phenomenon is challenging because
the contrast enhancement is relatively small and needs
to be distinguished from the transient increase in the
signal in meningeal veins as the contrast bolus transits
through. With 3 T MRI, detection is improved by use
of a T2-weighted FLAIR (T2-FLAIR) MRI acquisition
sequence, which can increase sensitivity by as much as
an order of magnitude relative to T1-weighted imag-
ing119. In one study, multifocal leptomeningeal enhance-
ment was seen in a greater proportion (86%) of people
Fig. 3 | A slowly expanding multiple sclerosis lesion. with SPMS than of people with RRMS (18%)120. A higher
Coregistered T1-weighted (top panels) and T2-weighted
frequency of leptomeningeal contrast enhancement in
(bottom panels) axial brain MRI images were acquired from
the same person with multiple sclerosis at baseline (part a) scans of people with progressive MS relative to those
and 12 months later (part b). The lesion in the red box with RRMS has been confirmed in larger studies, which
shows slow growth over the period of observation. The have also provided evidence for an association between
T1-weighted image intensity also decreases over this leptomeningeal contrast enhancement and long-​term
period, consistent with progressive tissue rarefaction. cortical atrophy120.

www.nature.com/nrneurol
Reviews

Most TSPO-​expressing inflammatory cells in active MS


1 2.2 lesions or at the borders of mixed active–inactive lesions
seem to be microglia124.
Use of the ‘first-​generation’ TSPO radioligand
11
C-​PK11195 first identified an association between
TSPO radioligand binding that suggested foci of inflam-
mation and some T2 hyperintense lesions in the white
matter of people with MS125,126. Ex vivo autoradiography
and histology provided confidence in the relative speci­
ficity of the TSPO radioligand binding to microglia/
L R
macrophages126. Subsequent studies have extended these
observations across different patient groups and demon-
strated associations of increased TSPO PET radioligand
uptake with brain atrophy in progressive MS127. Similar
findings have been produced with the use of two second-​
generation TSPO PET radioligands128. In people with
MRI PET SPMS, increased TSPO PET radiotracer uptake is seen
in the nonlesional white matter, as well as in lesions129.
Fig. 4 | PeT imaging of microglial activation in secondary progressive multiple Greater global brain uptake of the second-​generation
sclerosis. The axial 3D T1 MRI image on the left shows multiple T1-hypointense ‘black TSPO radioligand 11C-​PBR28 has been associated with
holes’. The MRI image is overlaid with a coregistered parametric 11C-​PK11195 PET image a longer duration of disease, and uptake is greater in
from the same person on the right. The relative 11C-​PK11195 binding is illustrated as the brains of people with SPMS than in brains of people
distribution volume ratio in each voxel (colour scale bar). Red arrows indicate chronic
with RRMS128.
lesions with increased microglial activation and higher 11C-​PK11195 binding at the lesion
edge (mixed active–inactive lesions), and white arrows indicate chronic lesions without
TSPO PET can also be used to characterize heterogen­
notable microglial activation at the lesion edge (chronic inactive lesions). Images eity of radioligand uptake between individual lesions,
courtesy of L. Airas, University of Turku, Turku PET Centre, Finland. which is broadly consistent with the differences in
microglial/macrophage density between lesions that
have been described histopathologically50. TSPO PET
With the important caveat that the use of gadolinium reveals considerable heterogeneity in radioligand uptake
contrast agent must be justified by the clinical benefits between lesions in the same person. Heterogeneity is
of the study (because of the potential adverse effects of apparent even among T2 hyperintense lesions that are
gadolinium contrast described above), T2-FLAIR with not distinguishable with conventional clinical MRI.
contrast is practical to include in a clinical MS scanning Classification of T2 hyperintense brain lesions according
assessment120,121. Postprocessing, in which the precon- to their radioligand uptake shows that the proportion of
trast image is subtracted from the postcontrast image, lesions in which TSPO radioligand uptake is low relative
can minimize false-​positive interpretations of the scans to surrounding nonlesional white matter (which are con-
and increase the accuracy of detection of leptomeningeal sidered to be relatively noninflammatory and presumed
contrast enhancement. Use of T2-FLAIR contrast lepto­ to be inactive) is higher in people with SPMS (35%) than
meningeal enhancement in conjunction with counts in those with RRMS (23%), but lesions in which TSPO
of MRI-​visible cortical lesions could provide a way of radioligand uptake is high (which are presumed to
assessing new inflammatory activity that complements be active) are found in both groups. Lesions defined
assessment of white matter lesions, and might better as active on the basis of their high TSPO radioligand
predict future disability122. uptake can be numerous even in people who are receiv-
ing the disease-​modifying treatments that are currently
CNS innate immune activation most effective for RRMS (for example, natalizumab
Gadolinium contrast-​based methods assess inflamma- or alemtuzumab)128,129. Diffuse white matter uptake of
tion through secondary effects of the inflammation on TSPO radioligand that is consistent with widespread
BBB permeability. Chronic brain inflammation can be microglial activation in the nonlesional white matter can
evaluated more directly by imaging molecules that are also be seen in RRMS and SPMS129. TSPO radioligand
associated with microglia and astrocytes of the innate uptake increases with greater disability measured with
immune system. This can be done with PET using radio­ the Multiple Sclerosis Severity Scale130. The magnitude
ligands that bind to molecules that are specifically of the diffuse uptake in the nonlesional white matter can
expressed in these cells. explain much of the variance in disability127.
The potential to see chronic inflammatory changes
PET of microglia. In vivo evidence for chronic microglial in living patients with MS by use of TSPO PET allows
activation in MS has come from translocator protein the clinical significance of these changes to be assessed
(TSPO) PET studies (Fig. 4). TSPO is relatively highly prospectively. For example, in a group of people who
expressed in microglia (although, in the brain, it is also were followed-​up over 1 year after PET imaging, TSPO
found in endothelial cells and in some activated astro- radiotracer uptake in the normal-​appearing white mat-
cytes), although it cannot be used to distinguish between ter (which reflects the diffuse microglial activation seen
microglia with pro-​inflammatory and homeostatic in postmortem studies) at baseline correlated well with
activation phenotypes as it is associated with both123. the number of enlarging T2 hyperintense lesions that

Nature Reviews | Neurology


Reviews

Box 2 | Translocator protein radioligands activated cortical microglia in the cortex is that lympho-
cytes express lower levels of TSPO than do microglia,
A wide range of translocator protein (TSPO) PET radioligands are available; several have making even clusters of meningeal lymphocytes less
been validated for human use159, and at least six (11C-​PK11195, 11C-​PBR28, 11C-DPA713, likely to be a major source of the signal observed. Further
18
F-​PBR111, 18F-​DPA714 and 18F-​GE180) have been used for studies in multiple sclerosis ex vivo studies are needed to explore this question.
(MS). The most important advantages of the newer, so-​called second-​generation
The various TSPO radioligands available each have
TSPO radioligands are an increased affinity for TSPO relative to the first-​generation
11
C-​PK11195 (refs160,161) and a higher target specificity162. A higher radioligand affinity specific advantages and disadvantages (Box 2), but the
increases the binding signal and, in some contexts, increases the sensitivity of PET to overall value of any TSPO radioligand is limited for
inflammatory neuropathology and clinically relevant differences between people with some applications by the lack of specificity of TSPO as
MS and healthy controls128. The choice between the second-​generation radioligands a marker of microglia; TSPO is expressed in vascular
depends on practical issues159; for example, some are fluorinated, meaning their endothelia, and high levels are expressed in some acti-
half-​lives are long enough for them to be distributed from a central manufacturing site vated astrocytes132. Interpretation of the TSPO PET sig-
to users working elsewhere. However, these ligands can have limitations; for example, nal therefore depends on the neuropathological context.
18
F-​PBR111 is rapidly de-​fluorinated in the body and the released fluoride binds to the To date, the neuropathological information available for
skull, confounding accurate assessment of cortical uptake130. MS has been limited; more quantitative immunohisto-
A limitation of many of the second-​generation radioligands is that they bind with
logical data are needed, particularly to facilitate inter-
lower affinity to TSPO in people who carry one genetic polymorphism at the TSPO locus
that substantially reduces binding of some other radioligands; this is common in pretation of differences in radioligand uptake between
Northern European populations160. Other TSPO radioligands that have been used to lesions with different radiological and neuropatholog-
study MS (for example, 18F-​GE180 (refs163–165)) have an advantage in that they bind ical features128. Another general problem with use of
similarly to TSPO in all people. The largest difference in binding as a result of the TSPO PET is that TSPO expression does not clearly
polymorphism is seen with 11C-​PBR28, but the impact of this difference can be managed distinguish between microglia with pro-​inflammatory,
in studies by excluding people who are homozygous for the ‘low-​binding’ allele and by anti-​inflammatory or homeostatic phenotypes. In
analysing those who are heterozygous or homozygous for the ‘high-​binding’ allele human microglia (unlike in rodent microglia), expres-
separately. Stratification of the population by genotype can also be used for 18F-​DPA714 sion of TSPO is not upregulated in the transition from a
PET, but the dynamic range of the signal with this radioligand in MS is lower than that homeostatic to an activated state123.
for 11C-​PBR28, which limits its practical application to people who are homozygous for
These limitations of the use of TSPO as a radioligand
the ‘high-​binding’ TSPO allele166.
target have stimulated the search for alternative,
microglia-specific PET radioligands. Preclinical stud-
developed over the subsequent year127. In people with ies published in 2019 have suggested potential for
SPMS, microglial activation in the normal-​appearing 11
C-​CPPC, a high-​affinity ligand that binds specifically
white matter at baseline was strongly correlated with to the macrophage colony-​stimulating factor 1 receptor
subsequent progression of brain atrophy127. (CSF1R), expression of which is limited to microglia in
Fewer data have been collected from PET imaging the brain133; results in rodents and nonhuman primates
of innate immune activation in the grey matter (in part support further development of this ligand for human
because the effective resolution of cortical grey matter use. Another promising alternative is 18F-​JNJ-64413739,
with PET is lower than that with conventional MRI). a radioligand that targets the P2X7 receptor134, which is
An initial report described increased 11C-​PK11195 strongly expressed in microglia/macrophages, although
TSPO PET radioligand uptake in subcortical grey mat- it is also expressed in B cells. Radioligands that target
ter of the thalamus126. However, as the thalamus and other candidate targets, including cyclooxygenase 1,
brainstem have higher microglial densities than other cyclooxygenase 2 (ref.135) and the CB2 cannabinoid
regions, even in the healthy brain, interpretation of this receptor136, are being developed and evaluated. None
observation requires quantitative correlations between of these newer radioligands have yet been used for the
histopathology and TSPO radioligand binding in post- study of MS, but the emerging range of potential PET
mortem tissue. Subsequent work has shown that TSPO radioligand targets suggests that future studies could
radiotracer uptake in the cortical grey matter correlates employ different, molecularly specific radioligands seri-
with disability, as assessed by the Expanded Disability ally in the same person to better characterize chronic
Status Scale and the Multiple Sclerosis Impact Scale, and immune responses. However, extension of this rationale
with cognitive performance130,131, suggesting that TSPO with the development of radioligands that discriminate
PET identifies grey matter inflammatory activity that is between homeostatic, activated neuroprotective and
relevant to disability in MS. This study exploited the spa- pro-​inflammatory microglial phenotypes is a challenge
tial localization made possible by high-​resolution PET yet to be addressed.
and 7 T MRI to elegantly demonstrate localized increases
in 11C-​PBR28 uptake to the cortex131. PET of astrocytes. In addition to microglia, astrocytes
To date, increased TSPO radioligand uptake associ- also contribute to the innate immune response, and
ated with the cerebral cortex in MS has been interpreted evidence suggests that they are principle effectors of
as an indication of inflammation in the grey matter. neuronal and oligodendroglial cell death upon pro-​
However, distinguishing confidently between chronic inflammatory microglial activation137. Acetate is a poten-
meningeal and chronic cortical inflammation9,43 is prob- tial ligand for astrocyte PET as it is relatively selectively
lematic given the relatively poor spatial resolution of metabolized by astrocytes in the CNS. In a small pilot
PET and the uncertainties inherent in coregistration study, 11C-​acetate uptake in the bilateral thalami and dif-
of PET and MRI scans10,43. The strongest general argu- fusely in the white matter was shown to be greater in the
ment that the peripheral grey matter signal arises from brains of people with MS than in the brains of healthy

www.nature.com/nrneurol
Reviews

controls138. The numbers of T2 lesions and T1 black neurodegeneration and disability that are characteristic
holes correlated with the 11C-​acetate uptake, suggesting of the disease20,43,114,120,127,131,151.
that this measure is a relevant index of inflammatory New MRI and PET tools are becoming available that
pathology. enable researchers to visualize chronic brain inflam-
Other PET radioligand target proteins are believed to mation in vivo. A general hypothesis that is gaining
be relatively selectively expressed in astrocytes, although acceptance is that the balance between chronic inflam-
these radioligands have yet to be applied to the study of mation and acute inflammatory episodes changes with
MS. 11C-​DED binds to mitochondrial monoamine oxi- ageing and disease progression70. The availability of the
dase B, which is expressed predominantly in astrocytes. kinds of advanced imaging techniques reviewed above
The PET signal with this ligand is increased in regions means that this hypothesis can be tested with clinical
of dense astrocytosis in Creutzfeldt–Jakob disease139 studies. Important questions can then be addressed.
and in Alzheimer disease140, suggesting potential for For example, which aspects of the neuroinflammatory
its use in MS. An alternative PET radioligand that has responses contribute to homeostatic resilience to neural
been charac­terized in humans is 11C-​BU99008, which or glial injury and which accelerate neurodegene­
targets the imidazoline-2 binding site that is thought to ration? What explains the variation in chronic immune
be found predominantly in astrocytes and is implicated responses between individuals? In conjunction with
in regulating the expression of glial fibrillary acidic pro- additional imaging tools for assessment of demyelina-
tein141. The usefulness of this radioligand for MS has not tion and remyelination, neurodegeneration and synaptic
yet been explored, although one potential disadvantage loss152–155, the impact of chronic neuroinflammation and
now is that the specific target protein (or proteins) has its modulation in the CNS can now be studied directly.
not been identified. The fact that most current disease-​m odifying
A more widely explored index of astrocytosis is MRS treatments for MS seem to have modest or no clinical
measures of brain myo-​inositol, a metabolic marker efficacy in progressive forms of the disease has been
associated with glial cell activation and proliferation142. disappointing. These therapies might not optimally
Concentrations of myo-​inositol are greater in astrocytes modulate the complex balance of adaptive and innate
than in neurons and can be expected to increase with or pro-​inflammatory and protective immune responses
neuroinflammation142. Myo-​inositol levels are increased seen in progressive disease156. Conventional clinical trial
in brains of people with MS relative to the brains of designs might be underpowered to enable detection of
healthy controls143. MRS has advantages over PET in that the progression of disability independent of relapses,
it does not involve radiation, it is available with clinical and conventional secondary endpoints, such as white
MRI scanners, and the cost is relatively low. Direct evi- matter lesion activity, are poorly suited to progressive
dence to support the use of myo-​inositol levels measured disease156,157. The latest clinical trials with new molecules
by MRS as an index of astrogliosis largely comes from a and improved designs have produced more encourag-
biopsy study of brains from three patients with unusual, ing results158, but we have not achieved what we need
acute inflammatory lesions144. A study in which MRS — a way of reliably slowing, stopping or preventing the
was used to determine relative brain myo-​inositol con- chronic inflammation that leads to the progression of dis-
centrations in a small group of people with MS who were ability. New approaches are needed. Advanced imaging
also studied with TSPO PET suggested that the two tech- methods that are sensitive to chronic inflammation are
niques measure at least partially independent pheno­m­ among the new tools that could enable more informative
ena (despite their general correlation with higher levels clinical trials of potential therapies.
of inflammation)145. This evidence is consistent with This Review highlights the importance of being
the notion that they report on different aspects of neuro­ able to assess and monitor inflammatory mechanisms
inflammation in MS. However, the specificity of myo-​ throughout the course of MS. The challenge now is to
inositol as a biomarker for astrocytes is unclear because validate advanced methods for imaging neuroinflamma-
anabolic and catabolic pathways for myo-​inositol are also tion sufficiently for the confident use of them in clini-
expressed in other cell types146. cal trials and then to make some of the methods more
practical for routine use in the clinic. We also still need
Conclusions to discover even better ways of visualizing the chronic
Current clinical management of MS relies on con- inflammatory activity associated with progressive
ventional MRI measures of lesion count and distribu- MS in vivo. With success, we should be better able to
tion, and of BBB breakdown detected by gadolinium select patients who will benefit from newer treatments
contrast 4,147–150. However, these methods are rela- and accelerate development of therapeutics to address
tively insensitive to the chronic inflammation that has remaining, major unmet medical needs of people with
been identified neuropathologically in the CNS in progressive MS.
MS. A growing body of data suggests that this chronic
inflammation is a major factor in the progressive Published online xx xx xxxx

1. Baecher-​Allan, C., Kaskow, B. J. & Weiner, H. L. 3. Miller, D. H., Grossman, R. I., Reingold, S. C. & for early diagnosis, disease monitoring and
Multiple sclerosis: mechanisms and immunotherapy. McFarland, H. F. The role of magnetic resonance assessment of treatment related complications.
Neuron 97, 742–768 (2018). techniques in understanding and managing multiple Br. J. Radiol. 90, 20160721 (2017).
2. Young, I. R. et al. Nuclear magnetic resonance imaging sclerosis. Brain 121 (Pt 1), 3–24 (1998). 5. Rio, J., Comabella, M. & Montalban, X. Predicting
of the brain in multiple sclerosis. Lancet 2, 4. Igra, M. S., Paling, D., Wattjes, M. P., Connolly, D. J. A. responders to therapies for multiple sclerosis.
1063–1066 (1981). & Hoggard, N. Multiple sclerosis update: use of MRI Nat. Rev. Neurol. 5, 553–560 (2009).

Nature Reviews | Neurology


Reviews

6. Bermel, R. A., Fisher, E. & Cohen, J. A. The use of MR 31. Alvarez, J. I., Cayrol, R. & Prat, A. Disruption of 57. Savarin, C. et al. Astrocyte response to IFN-​gamma
imaging as an outcome measure in multiple sclerosis central nervous system barriers in multiple sclerosis. limits IL-6-mediated microglia activation and progressive
clinical trials. Neuroimaging Clin. N. Am. 18, Biochim. Biophys. Acta 1812, 252–264 (2011). autoimmune encephalomyelitis. J. Neuroinflammation
687–701.xi (2008). 32. Leech, S., Kirk, J., Plumb, J. & McQuaid, S. Persistent 12, 79 (2015).
7. Mayo, L., Quintana, F. J. & Weiner, H. L. The innate endothelial abnormalities and blood-​brain barrier 58. Metz, I. et al. Pathologic heterogeneity persists in
immune system in demyelinating disease. Immunol. leak in primary and secondary progressive multiple early active multiple sclerosis lesions. Ann. Neurol.
Rev. 248, 170–187 (2012). sclerosis. Neuropathol. Appl. Neurobiol. 33, 86–98 75, 728–738 (2014).
8. Serafini, B., Rosicarelli, B., Magliozzi, R., Stigliano, E. (2007). 59. Frischer, J. M. et al. Clinical and pathological
& Aloisi, F. Detection of ectopic B-​cell follicles with 33. Kwon, E. E. & Prineas, J. W. Blood-​brain barrier insights into the dynamic nature of the white matter
germinal centers in the meninges of patients with abnormalities in longstanding multiple sclerosis lesions. multiple sclerosis plaque. Ann. Neurol. 78, 710–721
secondary progressive multiple sclerosis. Brain Pathol. An immunohistochemical study. J. Neuropathol. (2015).
14, 164–174 (2004). Exp. Neurol. 53, 625–636 (1994). 60. Luchetti, S. et al. Progressive multiple sclerosis patients
9. Howell, O. W. et al. Meningeal inflammation is 34. Lee, N. J. et al. Spatiotemporal distribution of show substantial lesion activity that correlates with
widespread and linked to cortical pathology in fibrinogen in marmoset and human inflammatory clinical disease severity and sex: a retrospective
multiple sclerosis. Brain 134, 2755–2771 (2011). demyelination. Brain 141, 1637–1649 (2018). autopsy cohort analysis. Acta Neuropathol. 135,
10. Machado-​Santos, J. et al. The compartmentalized 35. Louveau, A. et al. Structural and functional features 511–528 (2018).
inflammatory response in the multiple sclerosis brain of central nervous system lymphatic vessels. Nature 61. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W.
is composed of tissue-​resident CD8+ T lymphocytes 523, 337–341 (2015). & Rossi, F. M. Local self-​renewal can sustain CNS
and B cells. Brain 141, 2066–2082 (2018). 36. Louveau, A. et al. CNS lymphatic drainage and microglia maintenance and function throughout
11. Campbell, G. R., Worrall, J. T. & Mahad, D. J. neuroinflammation are regulated by meningeal adult life. Nat. Neurosci. 10, 1538–1543 (2007).
The central role of mitochondria in axonal lymphatic vasculature. Nat. Neurosci. 21, 62. van der Valk, P. & Amor, S. Preactive lesions in
degeneration in multiple sclerosis. Mult. Scler. 20, 1380–1391 (2018). multiple sclerosis. Curr. Opin. Neurol. 22, 207–213
1806–1813 (2014). 37. Rasmussen, M. K., Mestre, H. & Nedergaard, M. (2009).
12. Mahad, D. H., Trapp, B. D. & Lassmann, H. The glymphatic pathway in neurological disorders. 63. Ingram, G. et al. Complement activation in multiple
Pathological mechanisms in progressive multiple Lancet Neurol. 17, 1016–1024 (2018). sclerosis plaques: an immunohistochemical analysis.
sclerosis. Lancet Neurol. 14, 183–193 (2015). 38. Simon, M. J. & Iliff, J. J. Regulation of cerebrospinal Acta Neuropathol. Commun. 2, 53 (2014).
13. Bruck, W. Inflammatory demyelination is not central fluid (CSF) flow in neurodegenerative, neurovascular 64. Singh, S. et al. Microglial nodules in early multiple
to the pathogenesis of multiple sclerosis. J. Neurol. and neuroinflammatory disease. Biochim. Biophys. Acta sclerosis white matter are associated with
252 (Suppl. 5), v10–v15 (2005). 1862, 442–451 (2016). degenerating axons. Acta Neuropathol. 125,
14. Lassmann, H., van Horssen, J. & Mahad, D. 39. Wicken, C., Nguyen, J., Karna, R. & Bhargava, P. 595–608 (2013).
Progressive multiple sclerosis: pathology and Leptomeningeal inflammation in multiple sclerosis: 65. Prineas, J. W. et al. Immunopathology of secondary-​
pathogenesis. Nat. Rev. Neurol. 8, 647–656 (2012). insights from animal and human studies. Mult. Scler. progressive multiple sclerosis. Ann. Neurol. 50,
15. Matthews, P. M. et al. A practical review of the Relat. Disord. 26, 173–182 (2018). 646–657 (2001).
neuropathology and neuroimaging of multiple 40. Magliozzi, R., Columba-​Cabezas, S., Serafini, B. & 66. Costantini, E., D'Angelo, C. & Reale, M. The role of
sclerosis. Pract. Neurol. 16, 279–287 (2016). Aloisi, F. Intracerebral expression of CXCL13 and immunosenescence in neurodegenerative diseases.
16. Kutzelnigg, A. & Lassmann, H. Pathology of multiple BAFF is accompanied by formation of lymphoid follicle-​ Mediators Inflamm. 2018, 6039171 (2018).
sclerosis and related inflammatory demyelinating like structures in the meninges of mice with relapsing 67. Khademi, M. et al. Intense inflammation and nerve
diseases. Handb. Clin. Neurol. 122, 15–58 (2014). experimental autoimmune encephalomyelitis. damage in early multiple sclerosis subsides at older
17. Stadelmann, C. Multiple sclerosis as a J. Neuroimmunol. 148, 11–23 (2004). age: a reflection by cerebrospinal fluid biomarkers.
neurodegenerative disease: pathology, mechanisms 41. Sellebjerg, F. et al. Increased cerebrospinal fluid PLOS ONE 8, e63172 (2013).
and therapeutic implications. Curr. Opin. Neurol. 24, concentrations of the chemokine CXCL13 in active 68. Djikic, J. et al. Age-​associated changes in rat immune
224–229 (2011). MS. Neurology 73, 2003–2010 (2009). system: lessons learned from experimental
18. Peterson, J. W., Bo, L., Mork, S., Chang, A. 42. Festa, E. D. et al. Serum levels of CXCL13 are elevated autoimmune encephalomyelitis. Exp. Gerontol. 58,
& Trapp, B. D. Transected neurites, apoptotic neurons, in active multiple sclerosis. Mult. Scler. 15, 1271–1279 179–197 (2014).
and reduced inflammation in cortical multiple sclerosis (2009). 69. Zrzavy, T. et al. Dominant role of microglial and
lesions. Ann. Neurol. 50, 389–400 (2001). 43. Magliozzi, R. et al. Meningeal B-​cell follicles in macrophage innate immune responses in human
19. Lucchinetti, C. F. et al. Inflammatory cortical secondary progressive multiple sclerosis associate ischemic infarcts. Brain Pathol. 28, 791–805 (2018).
demyelination in early multiple sclerosis. N. Engl. with early onset of disease and severe cortical 70. Bar-​Or, A. & Antel, J. P. Central nervous system
J. Med. 365, 2188–2197 (2011). pathology. Brain 130, 1089–1104 (2007). inflammation across the age span. Curr. Opin. Neurol.
20. Kutzelnigg, A. & Lassmann, H. Cortical lesions and 44. Carassiti, D. et al. Neuronal loss, demyelination and 29, 381–387 (2016).
brain atrophy in MS. J. Neurol. Sci. 233, 55–59 volume change in the multiple sclerosis neocortex. 71. Mundt, S. et al. Conventional DCs sample and
(2005). Neuropathol. Appl. Neurobiol. 44, 377–390 (2018). present myelin antigens in the healthy CNS and allow
21. Trapp, B. D. et al. Axonal transection in the lesions 45. Wegner, C. & Matthews, P. M. A new view of the parenchymal T cell entry to initiate neuroinflammation.
of multiple sclerosis. N. Engl. J. Med. 338, 278–285 cortex, new insights into multiple sclerosis. Brain 126, Sci. Immunol. 4, eaau8380 (2019).
(1998). 1719–1721 (2003). 72. Duszczyszyn, D. A. et al. Thymic involution and
22. Cifelli, A. et al. Thalamic neurodegeneration in 46. Wegner, C., Esiri, M. M., Chance, S. A., Palace, J. & proliferative T-​cell responses in multiple sclerosis.
multiple sclerosis. Ann. Neurol. 52, 650–653 Matthews, P. M. Neocortical neuronal, synaptic, and J. Neuroimmunol. 221, 73–80 (2010).
(2002). glial loss in multiple sclerosis. Neurology 67, 960–967 73. Raz, L., Knoefel, J. & Bhaskar, K. The neuropathology
23. Evangelou, N. et al. Regional axonal loss in the corpus (2006). and cerebrovascular mechanisms of dementia.
callosum correlates with cerebral white matter lesion 47. Nylander, A. & Hafler, D. A. Multiple sclerosis. J. Clin. J. Cereb. Blood Flow Metab. 36, 172–186 (2016).
volume and distribution in multiple sclerosis. Brain Invest. 122, 1180–1188 (2012). 74. Geraldes, R., Esiri, M. M., DeLuca, G. C. & Palace, J.
123 (Pt 9), 1845–1849 (2000). 48. Correale, J., Gaitan, M. I., Ysrraelit, M. C. & Fiol, M. P. Age-​related small vessel disease: a potential
24. Tietz, S. & Engelhardt, B. Brain barriers: crosstalk Progressive multiple sclerosis: from pathogenic contributor to neurodegeneration in multiple sclerosis.
between complex tight junctions and adherens mechanisms to treatment. Brain 140, 527–546 Brain Pathol. 27, 707–722 (2017).
junctions. J. Cell Biol. 209, 493–506 (2015). (2017). 75. Perry, V. H., Cunningham, C. & Holmes, C.
25. Hawley, R. J. et al. Neurochemical correlates 49. Smyth, L. J., Kirby, J. A. & Cunningham, A. C. Role Systemic infections and inflammation affect chronic
of sympathetic activation during severe alcohol of the mucosal integrin alpha(E)(CD103)beta(7) in neurodegeneration. Nat. Rev. Immunol. 7, 161–167
withdrawal. Alcohol Clin. Exp. Res. 18, 1312–1316 tissue-​restricted cytotoxicity. Clin. Exp. Immunol. 149, (2007).
(1994). 162–170 (2007). 76. Conde, J. R. & Streit, W. J. Microglia in the aging
26. Meyer, C., Martin-​Blondel, G. & Liblau, R. S. 50. Kuhlmann, T. et al. An updated histological brain. J. Neuropathol. Exp. Neurol. 65, 199–203
Endothelial cells and lymphatics at the interface classification system for multiple sclerosis lesions. (2006).
between the immune and central nervous systems: Acta Neuropathol. 133, 13–24 (2017). 77. Ohrfelt, A. et al. Soluble TREM-2 in cerebrospinal
implications for multiple sclerosis. Curr. Opin. Neurol. 51. Stangel, M., Kuhlmann, T., Matthews, P. M. & fluid from patients with multiple sclerosis treated
30, 222–230 (2017). Kilpatrick, T. J. Achievements and obstacles of with natalizumab or mitoxantrone. Mult. Scler. 22,
27. Man, S., Ubogu, E. E. & Ransohoff, R. M. remyelinating therapies in multiple sclerosis. Nat. Rev. 1587–1595 (2016).
Inflammatory cell migration into the central nervous Neurol. 13, 742–754 (2017). 78. Zrzavy, T. et al. Pro-​inflammatory activation of
system: a few new twists on an old tale. Brain Pathol. 52. Frischer, J. M. et al. The relation between microglia in the brain of patients with sepsis.
17, 243–250 (2007). inflammation and neurodegeneration in multiple Neuropathol. Appl. Neurobiol. 45, 278–290 (2019).
28. Vercellino, M. et al. Involvement of the choroid plexus sclerosis brains. Brain 132, 1175–1189 (2009). 79. Wendeln, A. C. et al. Innate immune memory in the
in multiple sclerosis autoimmune inflammation: 53. Reynolds, R. et al. The neuropathological basis of clinical brain shapes neurological disease hallmarks. Nature
a neuropathological study. J. Neuroimmunol. 199, progression in multiple sclerosis. Acta Neuropathol. 556, 332–338 (2018).
133–141 (2008). 122, 155–170 (2011). 80. Rao, V. T. et al. MicroRNA expression patterns in
29. McDonnell, G. V., McMillan, S. A., Douglas, J. P., 54. Hametner, S. et al. Iron and neurodegeneration in the human astrocytes in relation to anatomical location
Droogan, A. G. & Hawkins, S. A. Serum soluble multiple sclerosis brain. Ann. Neurol. 74, 848–861 and age. J. Neuropathol. Exp. Neurol. 75, 156–166
adhesion molecules in multiple sclerosis: raised (2013). (2016).
sVCAM-1, sICAM-1 and sE-​selectin in primary 55. Miron, V. E. et al. M2 microglia and macrophages 81. Thompson, A. J. et al. Major differences in the dynamics
progressive disease. J. Neurol. 246, 87–92 (1999). drive oligodendrocyte differentiation during CNS of primary and secondary progressive multiple sclerosis.
30. Kirk, J., Plumb, J., Mirakhur, M. & McQuaid, S. remyelination. Nat. Neurosci. 16, 1211–1218 Ann. Neurol. 29, 53–62 (1991).
Tight junctional abnormality in multiple sclerosis white (2013). A highly important, pioneering study that still
matter affects all calibres of vessel and is associated 56. Zrzavy, T. et al. Loss of 'homeostatic' microglia and deserves reading for its clear demonstration that
with blood-​brain barrier leakage and active patterns of their activation in active multiple sclerosis. substantial inflammatory activity can be found in
demyelination. J. Pathol. 201, 319–327 (2003). Brain 140, 1900–1913 (2017). patients with chronic MS.

www.nature.com/nrneurol
Reviews

82. Sormani, M. P. et al. Clinical trials of multiple sclerosis 99. Vellinga, M. M. et al. Use of ultrasmall 120. Zivadinov, R. et al. Leptomeningeal contrast
monitored with enhanced MRI: new sample size superparamagnetic particles of iron oxide (USPIO)- enhancement is associated with progression of cortical
calculations based on large data sets. J. Neurol. enhanced MRI to demonstrate diffuse inflammation atrophy in MS: a retrospective, pilot, observational
Neurosurg. Psychiatry 70, 494–499 (2001). in the normal-​appearing white matter (NAWM) of longitudinal study. Mult. Scler. 23, 1336–1345
83. Geurts, J. J. et al. Intracortical lesions in multiple multiple sclerosis (MS) patients: an exploratory study. (2017).
sclerosis: improved detection with 3D double J. Magn. Reson. Imaging 29, 774–779 (2009). 121. Zivadinov, R. et al. Evaluation of leptomeningeal
inversion-​recovery MR imaging. Radiology 236, 100. Neuwelt, A. et al. Iron-​based superparamagnetic contrast enhancement using pre- and postcontrast
254–260 (2005). nanoparticle contrast agents for MRI of infection subtraction 3D-​FLAIR imaging in multiple sclerosis.
84. Kober, T. et al. MP2RAGE multiple sclerosis magnetic and inflammation. AJR Am. J. Roentgenol. 204, AJNR Am. J. Neuroradiol. 39, 642–647 (2018).
resonance imaging at 3 T. Invest. Radiol. 47, W302–W313 (2015). 122. Magliozzi, R., Reynolds, R. & Calabrese, M. MRI of
346–352 (2012). 101. Lu, M., Cohen, M. H., Rieves, D. & Pazdur, R. FDA cortical lesions and its use in studying their role in MS
85. Harel, A. et al. Phase-​sensitive inversion-​recovery MRI report: ferumoxytol for intravenous iron therapy pathogenesis and disease course. Brain Pathol. 28,
improves longitudinal cortical lesion detection in in adult patients with chronic kidney disease. 735–742 (2018).
progressive MS. PLOS ONE 11, e0152180 (2016). Am. J. Hematol. 85, 315–319 (2010). 123. Owen, D. R. et al. Pro-​inflammatory activation of
86. Tofts, P. S. & Kermode, A. G. Measurement of the 102. Bailie, G. R. Comparison of rates of reported adverse primary microglia and macrophages increases 18 kDa
blood-​brain barrier permeability and leakage space events associated with i.v. iron products in the United translocator protein expression in rodents but not
using dynamic MR imaging. 1. Fundamental concepts. States. Am. J. Health Syst. Pharm. 69, 310–320 humans. J. Cereb. Blood Flow Metab. 37, 2679–2690
Magn. Reson. Med. 17, 357–367 (1991). (2012). (2017).
An elegant framework for the quantitative 103. Daldrup-​Link, H. E. Ten things you might not know 124. Cosenza-​Nashat, M. et al. Expression of the translocator
interpretation of dynamic gadolinium contrast MRI about iron oxide nanoparticles. Radiology 284, protein of 18 kDa by microglia, macrophages and
that has informed all efforts to assess chronic 616–629 (2017). astrocytes based on immunohistochemical localization in
blood–brain barrier breakdown with MRI that have 104. Zarghami, N. et al. Optimization of molecularly abnormal human brain. Neuropathol. Appl. Neurobiol.
come since. targeted MRI in the brain: empirical comparison 35, 306–328 (2009).
87. Soon, D., Tozer, D., Altmann, D., Tofts, P. & Miller, D. of sequences and particles. Int. J. Nanomedicine 13, 125. Vowinckel, E. et al. PK11195 binding to the peripheral
Quantification of subtle blood-​brain barrier disruption 4345–4359 (2018). benzodiazepine receptor as a marker of microglia
in non-​enhancing lesions in multiple sclerosis: a study 105. Geurts, J. J. et al. Cortical lesions in multiple sclerosis: activation in multiple sclerosis and experimental
of disease and lesion subtypes. Mult. Scler. 13, combined postmortem MR imaging and autoimmune encephalomyelitis. J. Neurosci. Res. 50,
884–894 (2007). histopathology. AJNR Am. J. Neuroradiol. 26, 345–353 (1997).
88. Cramer, S. P., Simonsen, H., Frederiksen, J. L., 572–577 (2005). 126. Banati, R. B. et al. The peripheral benzodiazepine
Rostrup, E. & Larsson, H. B. Abnormal blood-​brain An important study validating the MRI detection binding site in the brain in multiple sclerosis:
barrier permeability in normal appearing white matter of cortical lesions using postmortem brains. quantitative in vivo imaging of microglia as a measure
in multiple sclerosis investigated by MRI. Neuroimage Conceptually even more important, however, was of disease activity. Brain 123 (Pt 11), 2321–2337
Clin. 4, 182–189 (2014). the demonstration of limited sensitivity to these (2000).
89. Pasquini, L. et al. Gadolinium-​based contrast agent-​ lesions, suggesting that in vivo imaging may While not the first report, this is the most
related toxicities. CNS Drugs 32, 229–240 (2018). capture only the ‘tip of the iceberg’ of cortical influential early TSPO PET report. It includes
90. Marti-​Bonmati, L. & Marti-​Bonmati, E. Retention of demyelination with MS. a first effort to validate the signal using
gadolinium compounds used in magnetic resonance 106. Geurts, J. J. et al. Does high-​field MR imaging improve postmortem autoradiography and highlights both
imaging: a critical review and the recommendations of cortical lesion detection in multiple sclerosis? the heterogeneity in tracer uptake across lesions
regulatory agencies. Radiologia 59, 469–477 (2017). J. Neurol. 255, 183–191 (2008). and the involvement of grey matter. While the
91. Elliott, C. et al. Slowly expanding/evolving lesions as a 107. Wiggermann, V., Hernandez-​Torres, E., Traboulsee, A., significance of all of the observations was not fully
magnetic resonance imaging marker of chronic active Li, D. K. & Rauscher, A. FLAIR2: a combination of FLAIR appreciated at the time, it describes the full range
multiple sclerosis lesions. Mult. Scler. https://doi.org/ and T2 for improved MS lesion detection. AJNR Am. of phenomena that have been explored since.
10.1177/1352458518814117 (2018). J. Neuroradiol. 37, 259–265 (2016). 127. Datta, G. et al. Neuroinflammation and its relationship
This article describes convincing evidence for the 108. Kilsdonk, I. D. et al. Multicontrast MR imaging at 7T to changes in brain volume and white matter lesions in
slow expansion of some chronic T2 hyperintense in multiple sclerosis: highest lesion detection in cortical multiple sclerosis. Brain 140, 2927–2938 (2017).
lesions and makes the powerful inference that this gray matter with 3D-​FLAIR. AJNR Am. J. Neuroradiol. 128. Datta, G. et al. 11C-​PBR28 and 18F-​PBR111 detect
relates to neuropathologically chronic active 34, 791–796 (2013). white matter inflammatory heterogeneity in multiple
lesions. 109. Calabrese, M. et al. Regional distribution and sclerosis. J. Nucl. Med. 58, 1477–1482 (2017).
92. Truyen, L. et al. Accumulation of hypointense lesions evolution of gray matter damage in different This paper represents the first effort to consolidate
("black holes") on T1 spin-​echo MRI correlates with populations of multiple sclerosis patients. PLOS ONE observations in the TSPO PET ‘subfield’ by showing
disease progression in multiple sclerosis. Neurology 10, e0135428 (2015). the quantitative correspondence between
47, 1469–1476 (1996). 110. Calabrese, M. et al. Imaging distribution and observations conducted over similar patient
93. Harrison, D. M. et al. Lesion heterogeneity on high-​ frequency of cortical lesions in patients with multiple populations using the same analytical methodology.
field susceptibility MRI is associated with multiple sclerosis. Neurology 75, 1234–1240 (2010). It also describes lesion heterogeneity in detail for
sclerosis severity. AJNR Am. J. Neuroradiol. 37, 111. Puthenparampil, M. et al. Cortical relapses in multiple the first time, proposing correspondences with
1447–1453 (2016). sclerosis. Mult. Scler. 22, 1184–1191 (2016). neuropathological classifications of potential
94. Dal-​Bianco, A. et al. Slow expansion of multiple 112. Calabrese, M. et al. Detection of cortical inflammatory clinical significance.
sclerosis iron rim lesions: pathology and 7 T magnetic lesions by double inversion recovery magnetic 129. Rissanen, E. et al. In vivo detection of diffuse
resonance imaging. Acta Neuropathol. 133, 25–42 resonance imaging in patients with multiple sclerosis. inflammation in secondary progressive multiple
(2017). Arch. Neurol. 64, 1416–1422 (2007). sclerosis using PET imaging and the radioligand
Extending the work of Elliott et al. (2018), 113. Scalfari, A. et al. The cortical damage, early relapses, 11
C-​PK11195. J. Nucl. Med. 55, 939–944 (2014).
this report provides additional evidence that and onset of the progressive phase in multiple 130. Colasanti, A. et al. In vivo assessment of brain white
slowly expanding MRI lesions represent the sclerosis. Neurology 90, e2107–e2118 (2018). matter inflammation in multiple sclerosis with
neuropathologically chronic active plaques by 114. Papadopoulou, A. et al. Contribution of cortical and 18
F-​PBR111 PET. J. Nucl. Med. 55, 1112–1118
demonstrating that the rims of these MRI lesions white matter lesions to cognitive impairment in (2014).
show evidence for pathological iron accumulation, multiple sclerosis. Mult. Scler. 19, 1290–1296 (2013). 131. Herranz, E. et al. Neuroinflammatory component of
corresponding to iron-​rich macrophages/microglia 115. Calabrese, M. et al. Morphology and evolution of gray matter pathology in multiple sclerosis. Ann. Neurol.
in these plaques. cortical lesions in multiple sclerosis. A longitudinal 80, 776–790 (2016).
95. Absinta, M. et al. Identification of chronic active multiple MRI study. Neuroimage 42, 1324–1328 (2008). 132. Betlazar, C., Harrison-​Brown, M., Middleton, R. J.,
sclerosis lesions on 3T MRI. AJNR Am. J. Neuroradiol. An important characterization of cortical lesions Banati, R. & Liu, G. J. Cellular sources and regional
39, 1233–1238 (2018). in vivo with demonstration of the frequency of their variations in the expression of the neuroinflammatory
96. Hametner, S. et al. The influence of brain iron and appearance, emphasizing that cortical lesions are marker translocator protein (TSPO) in the normal
myelin on magnetic susceptibility and effective associated with all stages of MS. brain. Int. J. Mol. Sci. 19, E2707 (2018).
transverse relaxation — a biochemical and histological 116. Calabrese, M. et al. Cortical lesions in primary 133. Horti, A. G. et al. PET imaging of microglia by
validation study. Neuroimage 179, 117–133 (2018). progressive multiple sclerosis: a 2-year longitudinal targeting macrophage colony-​stimulating factor 1
97. Corot, C. et al. Macrophage imaging in central nervous MR study. Neurology 72, 1330–1336 (2009). receptor (CSF1R). Proc. Natl Acad. Sci. USA 116,
system and in carotid atherosclerotic plaque using 117. Zurawski, J., Lassmann, H. & Bakshi, R. Use of 1686–1691 (2019).
ultrasmall superparamagnetic iron oxide in magnetic magnetic resonance imaging to visualize 134. Kolb, H. et al. Preclinical evaluation and non-human
resonance imaging. Invest. Radiol. 39, 619–625 leptomeningeal inflammation in patients with multiple primate receptor occupancy study of 18F-JNJ-
(2004). sclerosis: a review. JAMA Neurol. 74, 100–109 (2017). 64413739, a novel PET radioligand for P2X7
98. Tourdias, T. et al. Assessment of disease activity 118. Bergsland, N. et al. Leptomeningeal contrast receptors. J. Nucl. Med. https://doi.org/10.2967/
in multiple sclerosis phenotypes with combined enhancement is related to focal cortical thinning in jnumed.118.212696 (2019).
gadolinium- and superparamagnetic iron oxide-​ relapsing-​remitting multiple sclerosis: a cross-​sectional 135. Kim, M. J. et al. Evaluation of two potent and selective
enhanced MR imaging. Radiology 264, 225–233 MRI study. AJNR Am. J. Neuroradiol. 40, 620–625 PET radioligands to image COX-1 and COX-2 in rhesus
(2012). (2019). monkeys. J. Nucl. Med. 59, 1907–1912 (2018).
A pioneering demonstration of the potential 119. Absinta, M. et al. Gadolinium-​based MRI 136. Spinelli, F., Mu, L. & Ametamey, S. M. Radioligands for
utility of superparamagnetic iron oxide particle characterization of leptomeningeal inflammation in positron emission tomography imaging of cannabinoid
contrast for the assessment of cell trafficking into multiple sclerosis. Neurology 85, 18–28 (2015). type 2 receptor. J. Labelled Comp. Radiopharm. 61,
inflammatory lesions in MS. This remains the This important paper both introduces the concept 299–308 (2018).
single robust approach for direct assessment of leptomeningeal enhancement for assessment of 137. Liddelow, S. A. et al. Neurotoxic reactive astrocytes
of inflammatory cell trafficking into the brains of chronic inflammation in MS and validates it with an are induced by activated microglia. Nature 541,
humans. autopsy case study. 481–487 (2017).

Nature Reviews | Neurology


Reviews

138. Takata, K. et al. 11C-​acetate PET imaging in patients 149. Suthiphosuwan, S., Kim, D., Bharatha, A. & Oh, J. 163. Feeney, C. et al. Kinetic analysis of the translocator
with multiple sclerosis. PLOS ONE 9, e111598 Imaging markers for monitoring disease activity in protein positron emission tomography ligand [18F]
(2014). multiple sclerosis. Curr. Treat. Options Neurol. 19, 18 GE-180 in the human brain. Eur. J. Nucl. Med.
An underappreciated paper that provides an (2017). Mol. Imaging 43, 2201–2210 (2016).
interesting proof-​of-principle for imaging astrocyte 150. Oreja-​Guevara, C. & Paradig, M. S. G. Overview of 164. Vomacka, L. et al. TSPO imaging using the novel PET
activation in neurodegenerative disease. magnetic resonance imaging for management of ligand [18F]GE-180: quantification approaches in
139. Engler, H. et al. Imaging astrocytosis with PET in relapsing-​remitting multiple sclerosis in everyday patients with multiple sclerosis. EJNMMI Res. 7, 89
Creutzfeldt-​Jakob disease: case report with practice. Eur. J. Neurol. 22 (Suppl. 2), 22–27 (2015). (2017).
histopathological findings. Int. J. Clin. Exp. Med. 5, 151. Magliozzi, R. et al. A gradient of neuronal loss and 165. Unterrainer, M. et al. TSPO PET with [18F]GE-180
201–207 (2012). meningeal inflammation in multiple sclerosis. sensitively detects focal neuroinflammation in patients
140. Carter, S. F. et al. Evidence for astrocytosis in Ann. Neurol. 68, 477–493 (2010). with relapsing-​remitting multiple sclerosis. Eur. J.
prodromal Alzheimer disease provided by 152. Sormani, M. P. & Pardini, M. Assessing repair in Nucl. Med. Mol. Imaging 45, 1423–1431 (2018).
11C-​deuterium-L-​deprenyl: a multitracer PET multiple sclerosis: outcomes for phase II clinical trials. 166. Hagens, M. H. J. et al. In vivo assessment of
paradigm combining 11C-​Pittsburgh compound B Neurotherapeutics 14, 924–933 (2017). neuroinflammation in progressive multiple sclerosis:
and 18F-​FDG. J. Nucl. Med. 53, 37–46 (2012). 153. Ontaneda, D. & Fox, R. J. Imaging as an outcome a proof of concept study with [18F]DPA714 PET.
141. Tyacke, R. J. et al. Evaluation of 11C-​BU99008, a PET measure in multiple sclerosis. Neurotherapeutics 14, J. Neuroinflammation 15, 314 (2018).
ligand for the imidazoline2 binding site in human 24–34 (2017).
brain. J. Nucl. Med. 59, 1597–1602 (2018). 154. Bodini, B., Louapre, C. & Stankoff, B. Advanced Acknowledgements
142. Chang, L., Munsaka, S. M., Kraft-​Terry, S. & imaging tools to investigate multiple sclerosis P.M.M. acknowledges the early input of ideas from
Ernst, T. Magnetic resonance spectroscopy to pathology. Presse Med. 44, e159–e167 (2015). L. Steinman and H. Lassmann, who emphasized the potential
assess neuroinflammation and neuropathic pain. 155. Filippi, M. MRI measures of neurodegeneration in importance of long-​lived B cells in the brain parenchyma of
J. Neuroimmune Pharmacol. 8, 576–593 (2013). multiple sclerosis: implications for disability, disease people with MS. This and related work has been supported
143. Vrenken, H. et al. MR spectroscopic evidence for glial monitoring, and treatment. J. Neurol. 262, 1–6 by the Imperial College Healthcare Trust – National Institute
increase but not for neuro-​axonal damage in MS (2015). for Health Research (NIHR) Biomedical Research Centre.
normal-​appearing white matter. Magn. Reson. Med. 156. Tur, C. & Montalban, X. Progressive MS trials: lessons P.M.M. has also been in receipt of generous personal and
53, 256–266 (2005). learned. Mult. Scler. 23, 1583–1592 (2017). research support from the Edmond J. Safra Foundation
144. Bitsch, A. et al. Inflammatory CNS demyelination: 157. Ontaneda, D., Fox, R. J. & Chataway, J. Clinical trials and Lily Safra, an NIHR Senior Investigator’s Award, the
histopathologic correlation with in vivo quantitative in progressive multiple sclerosis: lessons learned and Medical Research Council and the UK Dementia Research
proton MR spectroscopy. AJNR Am. J. Neuroradiol. future perspectives. Lancet Neurol. 14, 208–223 Institute, which is supported by the Medical Research Council,
20, 1619–1627 (1999). (2015). The Alzheimer’s Society and Alzheimer’s Research UK.
145. Datta, G. et al. Translocator positron-​emission 158. Ciotti, J. R. & Cross, A. H. Disease-​modifying
tomography and magnetic resonance spectroscopic treatment in progressive multiple sclerosis. Curr. Treat. Competing interests
imaging of brain glial cell activation in multiple Options Neurol. 20, 12 (2018). P.M.M. acknowledges consultancy fees from Adelphi
sclerosis. Mult. Scler. 23, 1469–1478 (2017). 159. Alam, M. M., Lee, J. & Lee, S. Y. Recent progress in Communications, Biogen, Celgene and Roche. He has
146. Rae, C. D. A guide to the metabolic pathways and the development of TSPO PET ligands for received honoraria or speakers’ honoraria from Biogen,
function of metabolites observed in human brain 1H neuroinflammation imaging in neurological diseases. Novartis and Roche, and has received research or educational
magnetic resonance spectra. Neurochem. Res. 39, Nucl. Med. Mol. Imaging 51, 283–296 (2017). funds from Biogen, GlaxoSmithKline, Nodthera and Novartis.
1–36 (2014). 160. Owen, D. R. et al. An 18-kDa translocator protein He is a paid member of the Scientific Advisory Board for Ipsen
147. Wattjes, M. P., Steenwijk, M. D. & Stangel, M. MRI in (TSPO) polymorphism explains differences in binding Pharmaceuticals.
the diagnosis and monitoring of multiple sclerosis: affinity of the PET radioligand PBR28. J. Cereb. Blood
an update. Clin. Neuroradiol. 25 (Suppl. 2), 157–165 Flow Metab. 32, 1–5 (2012). Peer review information
(2015). 161. Kobayashi, M. et al. 11C-​DPA-713 has much greater Nature Reviews Neurology thanks J. Geurts and the other,
148. Vagberg, M. et al. Guidelines for the use of magnetic specific binding to translocator protein 18 kDa (TSPO) anonymous, reviewer(s) for their contribution to the peer
resonance imaging in diagnosing and monitoring the in human brain than 11C-(R)-PK11195. J. Cereb. Blood review of this work.
treatment of multiple sclerosis: recommendations of Flow Metab. 38, 393–403 (2018).
the Swedish Multiple Sclerosis Association and the 162. Owen, D. R. et al. Determination of [11C]PBR28 binding Publisher’s note
Swedish Neuroradiological Society. Acta Neurol. Scand. potential in vivo: a first human TSPO blocking study. Springer Nature remains neutral with regard to jurisdictional
135, 17–24 (2017). J. Cereb. Blood Flow Metab. 34, 989–994 (2014). claims in published maps and institutional affiliations.

www.nature.com/nrneurol

You might also like