You are on page 1of 10

312

Imaging in Encephalitis
Arun Venkatesan, MD, PhD1 Balaji Jagdish, BS1

1 Division of Neuroimmunology and Neuroinfectious Diseases, Address for correspondence Arun Venkatesan, MD, PhD, Division of
Department of Neurology, Johns Hopkins Encephalitis Center, Johns Neuroimmunology and Neuroinfectious Diseases, Department of
Hopkins University School of Medicine, Baltimore, Maryland Neurology, Johns Hopkins University School of Medicine, Baltimore,
MD 21287 (e-mail: avenkat2@jhmi.edu).
Semin Neurol 2019;39:312–321.

Abstract Despite recent advances in diagnostic and therapeutic modalities for infectious and
autoimmune encephalitis, the management of patients with suspected or confirmed
encephalitis poses a great challenge to physicians. Neuroimaging, including magnetic
resonance imaging (MRI) and positron emission tomography (PET) scanning, can play a
Keywords crucial role in substantiating the diagnosis of encephalitis and eliminating clinical
► autoimmune mimics of encephalitis from consideration. Moreover, characteristic neuroimaging
encephalitis patterns can aid in defining specific infectious and autoimmune etiologies. Volumetric

Downloaded by: Kent State University. Copyrighted material.


► infectious and functional MRI, in particular, are being increasingly used to characterize outcomes
encephalitis following encephalitis and can shed light on brain reorganization and function after the
► MRI acute phase of disease has resolved. Here, we discuss the uses of structural, functional,
► outcomes and PET neuroimaging in the clinical assessment of the acute and recovery phases of
► prognosis encephalitis.

Encephalitis refers to inflammation of the brain and can Neuroimaging in Diagnosis


present with an array of clinical symptoms including altered
level of consciousness, memory loss, psychiatric changes, sleep Neuroimaging plays a critical role in the acute evaluation of
disturbances, seizures, and weakness.1 While there are numer- patients with suspected encephalitis, as it may support the
ous causes, etiologies can be classified into two broad cate- diagnosis of encephalitis, point to specific etiologies, or identify
gories: autoimmune and infectious. Autoimmune encephalitis alternate conditions that mimic encephalitis. While computed
is a result of the generation of immune responses against tomography (CT) scanning employs X-rays to best define bony
components of the host’s central nervous system (CNS). In structures, conventional magnetic resonance imaging (MRI)
contrast, infectious encephalitis is often a result of viral or uses nuclear magnetic resonance to detect hydrogen nuclei
bacterial infections, and thus may additionally manifest with and provides excellent contrast between soft tissues. In all cases
fever and other systemic infectious symptoms and signs.1,2 of encephalitis, MRI is preferred over CT given the greater
As many clinical manifestations are shared between auto- sensitivity and specificity of MRI.3–5 In some situations, such
immune and infectious encephalitis, various diagnostic mod- as clinical instability or limited availability of MRI, a head CT
alities are employed to help discern the specific etiology. While must be relied upon and can be particularly useful in excluding
electroencephalography (EEG) and cerebrospinal fluid (CSF) acute intracranial hemorrhage and confirming brain shift. With
analysis are used in attempting to confirm the diagnosis of conventional MRI imaging, complementary sequences are
encephalitis and determining the specific cause, neuroimaging obtained, including T1-weighted imaging (T1W) which defines
plays an increasingly important role. This review will discuss structural abnormalities, T2-weighted imaging (T2W) and
neuroimaging modalities that can be used to help establish the fluid-attenuated inversion recovery (FLAIR) sequences which
diagnosis of encephalitis and suggest specific causes, which may identify areas of edema or inflammation, and susceptibil-
may correlate with outcomes. ity-weighted imaging for identification of areas of hemorrhage.
Postgadolinium T1W imaging can identify regions where the
blood–brain barrier has been disrupted, as can occur in the
Arun Venkatesan's ORCID is https://orcid.org/0000-0002-9335- setting of active inflammation. Particularly useful in the
7361.

Issue Theme Neuroinfectious Disease, Copyright © 2019 by Thieme Medical DOI https://doi.org/
Part 1; Guest Editor, Anna M. Cervantes- Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0039-1687838.
Arslanian, MD New York, NY 10001, USA. ISSN 0271-8235.
Tel: +1(212) 584-4662.
Imaging in Encephalitis Venkatesan, Jagdish 313

diagnosis of encephalitis is diffusion-weighted imaging (DWI), nium administration. In contrast with HSV encephalitis,
which identifies areas where diffusion of water is restricted. autoimmune causes tend to present with bilateral, sym-
This sequence may be more sensitive than other conventional metric temporal lobe involvement (►Fig. 2) and more often
sequences for detection of early abnormalities in encephalitis.6,7 demonstrate lesions that are outside the limbic region.12 The
These conventional MRI sequences can support the diag- character of extralimbic involvement may provide clues to
nosis of encephalitis by demonstrating new T2/FLAIR, DWI, specific autoimmune syndromes; for example, anti-Hu ence-
and/or gadolinium enhancing lesions,2,8 while importantly phalitis may present with additional lesions in the cerebel-
excluding other causes of acute neurologic dysfunction such lum and brainstem,15–17 anti-Ma encephalitis with T2-FLAIR
as ischemic or hemorrhagic stroke, posterior reversible abnormalities in the brainstem and thalamus,15,18 and anti-
encephalopathy syndrome, rapidly expanding brain tumors, GAD encephalitis may be accompanied by signs of cerebellar
and anoxic injury. In addition, the pattern of MRI abnorm- atrophy.19,20 Autoimmune encephalitis associated with anti-
alities may suggest specific etiologies as described below. bodies targeting neuronal cell surface proteins, including the
voltage-gated potassium complex antigens LGI1 and CASPR2,
Temporal Lobe Abnormalities NMDA-receptor (NMDAR), AMPA-receptor, and GABA B-
Conventional MRI imaging demonstrating T2-FLAIR hyper- receptors, may also exhibit T2-FLAIR hyperintensities in
intense lesions that localize to the medial temporal lobes can the medial temporal lobes.19,21
be indicative of many pathologies. The major infectious
etiologic consideration is herpes simplex virus (HSV), most Deep Gray Matter Abnormalities
commonly HSV-1. Lesions may be present in one or both When MRI imaging shows T2-FLAIR hyperintensities involving
lobes, and the involvement is typically asymmetric (►Fig. 1). the deep gray matter of the brain (basal ganglia, thalamus),

Downloaded by: Kent State University. Copyrighted material.


Additional lesions may be seen in associated limbic areas arboviruses including Japanese encephalitis virus, West Nile
such as the insula, cingulate gyri, and inferolateral frontal virus (WNV), and tick-borne encephalitis virus are prominent
cortices with characteristic sparing of the basal ganglia early considerations.22–25 These viruses, along with enteroviruses
in the disease process.9–12 While hemorrhage in the medial and rabies, may also affect the anterior horn cells in the spinal
temporal lobes has previously been noted to be typical of cord, resulting in acute flaccid paralysis.26–28 In Japanese
HSV encephalitis, it is now typically only seen as a late-stage encephalitis (JE), T2-FLAIR hyperintensities can be present
manifestation since patients are likely being diagnosed ear- throughout the subcortical gray matter, including the substan-
lier.12 Of note, in immunocompromised individuals the tia nigra, basal ganglia, cerebellum, thalamus, and in some
radiologic pattern may differ markedly, often with more patients the hippocampus, where the body and tail tend to be
widespread brain and/or meningeal involvement.13 Other involved.10,29,30 In children, an additional consideration is the
infectious etiologies that can present with temporal lobe respiratory virus group (i.e., influenza, parainfluenza, adeno-
involvement include varicella zoster virus (VZV), syphilis, virus, etc.), which has been found to be associated with sym-
and Mycobacterium tuberculosis.12,14 metric, bithalamic T2-FLAIR hyperintensities and accompanied
In up to two-thirds of cases, brain MRI is abnormal in in some cases by hemorrhage.25 Rarely, autoimmune encepha-
acute autoimmune limbic encephalitis and typically demon- litis presents with prominent deep gray matter involvement.
strates medial temporal lobe T2-FLAIR abnormalities with Basal ganglia abnormalities have been reported in anti-CRMP5
variable enhancement on T1W imaging following gadoli- encephalitis, while lesions in the caudate, putamen, globus

Fig. 1 HSV-1 encephalitis. Right temporal lobe abnormalities are clearly seen on FLAIR (A), while DWI (B) also shows areas of restricted diffusion
in the right insula and cingulate gyri as well as left temporal lobe (arrows). DWI, diffusion-weighted imaging; FLAIR, fluid-attenuated inversion
recovery.

Seminars in Neurology Vol. 39 No. 3/2019


314 Imaging in Encephalitis Venkatesan, Jagdish

Fig. 2 Autoimmune limbic encephalitis. Bilateral mesial temporal lobe abnormalities seen on FLAIR (A) and more clearly on DWI (B). Note the
symmetric nature of the lesions. (C) FDG-PET imaging demonstrates hyperavidity in the left mesial temporal lobe. DWI, diffusion-weighted
imaging; FDG-PET, fluorodeoxyglucose-positron emission tomography; FLAIR, fluid-attenuated inversion recovery.

Downloaded by: Kent State University. Copyrighted material.


pallidus, and substantia nigra have been described in antido- also present with T2-FLAIR hyperintensities in the brainstem,
pamine D2 receptor encephalitis, most notably in children.19,31 though the imaging manifestations are protean and in some
cases MRI is normal.37,38
Brainstem Abnormalities Autoimmune causes of brainstem encephalitis predomi-
A broad range of infectious and autoimmune etiologies can nate over infectious ones.33 Acute demyelinating disorders,
result in brainstem encephalitis.32,33 Listeria monocytogenes is including acute disseminated encephalomyelitis (ADEM) and
infamous for causing encephalitis of the pons and medulla neuromyelitis optica (NMO), often present with T2-FLAIR
(termed rhombencephalitis) predominantly in immunocom- hyperintensities in the brainstem and may additionally involve
petent individuals, and is often accompanied by multiple rim- the spinal cord or supratentorial areas. In one study of demye-
enhancing lesions within the lower brainstem (►Fig. 3).34,35 linating brainstem lesions, patients with ADEM more fre-
Other infectious considerations include neurobrucellosis, quently demonstrated midbrain and ventral lesions and
arboviruses, and enteroviruses. Of the latter, enterovirus-71 those with NMO more often had pontine or medullary and
is associated with posteriorly located, often bilateral, brain- dorsal lesions, while both groups had lesions with less well-
stem abnormalities that are hyperintense on T2W imaging and defined margins than individuals with multiple sclerosis.39
hypointense on T1W imaging.36 CNS Whipple’s disease can Behcet’s disease is a frequent cause of brainstem encephalitis,
particularly in the eastern rim of the Mediterranean and the
Middle East.9,35 Other autoimmune considerations include
Bickerstaff’s brainstem encephalitis, anti-Ri and antiglycine
receptor encephalitis, and CLIPPERS (chronic lymphocytic
inflammation with pontine perivascular enhancement
responsive to steroids). The latter is a syndrome with multiple
etiologies whose MRI features include small, homogenous
gadolinium enhancing nodules in the posterior fossa without
rim enhancement or mass effect (►Fig. 4).40

White Matter Abnormalities


Abnormalities of the white matter may arise in several
settings, including as a result of a small-vessel vasculopathy
with resultant hypoperfusion or infarction, or in the setting
of demyelination. VZV can cause a vasculopathy in both
immunocompetent and immunocompromised individuals,
though small-vessel vasculopathy with concomitant white
matter lesions is more common in the latter.41 T2-FLAIR and/
or DWI lesions may solely involve the white matter or may be
present at the gray–white junction. The punctate, innumer-
able deep white matter lesions that can occur in the acute
Fig. 3 Listeria rhombencephalitis. Coronal T1 postgadolinium scan phase of pediatric Rocky Mountain spotted fever encephalitis
shows multiple rim enhancing lesions in the brainstem. have been described as having a “starry sky” appearance on

Seminars in Neurology Vol. 39 No. 3/2019


Imaging in Encephalitis Venkatesan, Jagdish 315

Fig. 4 CLIPPERS. Sagittal FLAIR (A) and axial T1 postgadolinium (B) images demonstrate scattered punctate abnormalities in the dorsal pons.
CLIPPERS, chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids; FLAIR, fluid-attenuated inversion
recovery.

T2-FLAIR or DWI42 and also likely represent a small-vessel

Downloaded by: Kent State University. Copyrighted material.


vasculopathy. In neuroborreliosis, multiple deep and peri-
ventricular white matter lesions can develop that are
thought to represent demyelination.43,44 Those who develop
progressive multifocal leukoencephalopathy as a result of
John Cunningham virus infection of myelin-producing oli-
godendrocytes, on the other hand, typically develop multi-
focal subcortical white matter lesions with involvement of
the subcortical U-fibers, and little or no mass effect or
gadolinium enhancement on MRI.45,46 Individuals with
acquired immunodeficiency syndrome or other severe
immunocompromised conditions are at risk for cytomega-
lovirus encephalitis, which can manifest with periventricu-
lar T2-FLAIR hyperintensities, termed an “owl’s eye”
appearance.9,10 Finally, lesions in the splenium of the corpus
callosum may occur in the setting of encephalitis (►Fig. 5).
Often found in association with influenza infection, this has
also been noted with Epstein–Barr virus, rotavirus, and
Mycoplasma pneumoniae.47 The reversible nature of these
lesions suggests the possibility of transient metabolic com-
promise, perhaps in association with excitotoxic injury.48
Notably, such lesions are not specific for infection but can
be seen in a wide variety of conditions including hypogly-
cemia, drug toxicity, and seizure.

Multifocal Abnormalities Fig. 5 Influenza-associated encephalitis. DWI demonstrates


restricted diffusion within the splenium of the corpus callosum; this
As described above and in ►Table 1, several infectious and
resolved on follow-up imaging 2 weeks later. DWI, diffusion-weighted
autoimmune etiologies can present with multifocal CNS imaging.
involvement. In particular, ADEM, a syndrome characterized
by encephalopathy and focal neurologic signs often following plete “C-shaped” patterns of enhancement, likely as a result
infection or vaccination, typically presents with multiple of perivenous inflammation.49 Lesions often demonstrate
areas of T2-FLAIR hyperintensity in the cerebral cortex, restriction of diffusion in a peripheral pattern on DWI
brainstem, optic nerve, and spinal cord. The lesions may sequences, perhaps as a result of trapping of water within
involve white or gray matter, and enhancement is common; layers of injured myelin or the presence of hypercellular
when enhancement is observed, all of the lesions enhance to infiltrates at the leading edge of expanding lesions.50 Lesions
a similar degree since they tend to arise synchronously. in anti-GABAa receptor encephalitis may be mistaken for
While many infectious agents will result in complete rim ADEM, as multifocal cortical and subcortical lesions are
enhancement following gadolinium administration, in typically seen. One point of distinction, however, is that
ADEM the enhancement is typically characterized by incom- the lesions in anti-GABAa receptor encephalitis are often

Seminars in Neurology Vol. 39 No. 3/2019


316 Imaging in Encephalitis Venkatesan, Jagdish

Table 1 Selected causes of multifocal encephalitis

Pathogen Characteristic brain MRI findings


VZV Ischemic or hemorrhagic lesions in white matter or gray–white matter junction
Enteroviruses Abnormalities may involve deep gray matter (i.e., thalamus, basal ganglia),
brainstem, and spinal cord
Arboviruses Abnormalities may involve deep gray matter (i.e. thalamus, basal ganglia),
brainstem, and spinal cord
JC virus Multifocal subcortical white matter lesions
Measles Cerebral edema, multifocal lesions, can resemble ADEM in acute setting
Mumps Lesions in brainstem, hippocampus, splenium of corpus callosum
Nipah virus Widespread punctate subcortical and deep white matter lesions
Rabies virus Multifocal abnormalities throughout cortex, deep gray nuclei, brainstem, cerebral
white matter; gray matter is involved much more frequently than white matter
Borrelia spp. Multifocal lesions in subcortical and periventricular white matter, may mimic
multiple sclerosis
Rickettsia spp. Multiple punctate areas of restricted diffusion, “starry sky” pattern
Mycobacterium tuberculosis Multiple rim-enhancing and/or vascular lesions

Downloaded by: Kent State University. Copyrighted material.


Fungal infections Multiple rim-enhancing lesions
(Coccidioides, Histoplasma, Blastomyces)
Balamuthia mandrillaris Multifocal T2 hyperintensities with rim enhancement
Baylisascaris procyonis Multifocal or confluent white matter abnormalities with nodular enhancement
Autoimmune condition
ADEM Multifocal CNS involvement, white matter > gray matter, “C-shaped” rim of
enhancement, variable spinal cord involvement, lesions develop synchronously
MOG encephalomyelitis Multiple cortical or subcortical lesions, longitudinally extensive spinal cord lesions
Anti-GABAa receptor encephalitis Multiple asynchronous cortical and subcortical lesions

Abbreviations: ADEM, acute disseminated encephalomyelitis; CNS, central nervous system; JC, John Cunningham; MOG, myelin oligodendrocyte
glycoprotein; MRI, magnetic resonance imaging; VZV, varicella zoster virus.

asynchronous.51 Such lesions may be seen less frequently in with other patients with confirmed and probable autoim-
other autoimmune encephalitides (►Fig. 6). mune encephalitis. In anti-LGI1 encephalitis, where basal
ganglia T1 and T2 hyperintensities have been reported on
Additional Neuroimaging Modalities MRI, FDG-PET has shown evidence of basal ganglia hyper-
Particularly in cases of neuronal surface antibody-mediated metabolism,56–58 though prospective studies are needed.
encephalitis, MRI may be normal. Indeed, a majority of
patients with anti-NMDAR encephalitis (50–77%) do not
Neuroimaging Biomarkers and Outcomes
present with any abnormalities upon standard MRI, nor do
up to one-third of patients with autoimmune limbic ence- The mortality rate for acute encephalitis has been reported to
phalitis.15,52–54 As a result, there has been increased interest be between 5 and 15%. Half or more of survivors are left with
in the evaluation of other neuroimaging modalities that may substantial neurological and cognitive deficits, which may
contribute to the diagnosis of autoimmune encephalitis. In include lower intelligence quotients, behavioral impairments,
this regard, fluorodeoxyglucose-positron emission tomogra- attention-deficit hyperactivity disorder, and residual motor
phy (FDG-PET) may prove useful. Indeed, hyperavidity in the deficits.59 Mortality rates in autoimmune encephalitis tend to
mesial temporal lobes is frequently seen in autoimmune be lower than infectious cases, although prolonged recovery
limbic encephalitis, and current consensus includes this as and the potential for immune relapses may contribute to long-
fulfilling radiographic criteria.2 Specific FDG-PET signatures term morbidity. Notably, those patients with autoimmune
may be associated with subgroups of autoimmune encepha- encephalitis associated with antibodies targeting intracellular
litis. In a study of eight patients with anti-NMDAR encepha- proteins, such as anti-Hu, anti-Ma/Ta, anti-GAD, and anti-CV2,
litis who were evaluated within 12 weeks or less of symptom have a poorer prognosis than those with antibodies targeting
onset, FDG-PET/CT showed cortical hypometabolism in cell surface proteins.60 Several potentially modifiable factors
seven patients and significant medial occipital lobe hypo- are predictive of a poor outcome in cohorts of all-cause
metabolism in six of the eight patients.55 These patients also encephalitis; these include delays to treatment, raised intra-
exhibited greater occipital hypometabolism when compared cranial pressure and cerebral edema, lower body temperature,

Seminars in Neurology Vol. 39 No. 3/2019


Imaging in Encephalitis Venkatesan, Jagdish 317

associated with the severity of neuropsychological seque-


lae.72 More recently, restricted diffusion on MRI was found in
two-thirds of patients with herpes simplex encephalitis who
experienced poor outcome.73
WNV encephalitis carries a 10% mortality rate.59 Survivors
experience various neurocognitive deficits such as deficiencies
in language/social communication, memory impairment,
executive dysfunction, and defects in attention and concentra-
tion.74 A cross-sectional study of individuals who had suffered
from West Nile neuroinvasive disease showed significant
cortical thinning in both cerebral hemispheres on MRI, pri-
marily in the frontal and limbic cortices, as compared with age-
and gender-matched controls.75 Cortical thinning in the left
hemisphere correlated with neuropsychological impairment
as measured by the Repeatable Battery for the Assessment of
Neuropsychological Status (RBANS). Additionally, regional
atrophy was noted in the cerebellum, brain stem, thalamus,
putamen, and globus pallidus compared with controls. Thus,
despite the propensity for deep gray matter involvement in the
acute phase of WNV encephalitis,76 the resulting damage

Downloaded by: Kent State University. Copyrighted material.


appears to be more widespread and will need to be carefully
characterized in prospective studies.
JEV is responsible for numerous cases of encephalitis
worldwide, is associated with a high mortality rate (up to
30%), and the majority of surviving children suffer from
Fig. 6 Anti-CASPR2 encephalitis. FLAIR imaging demonstrates an functional impairment, especially in behavior, language,
atypical pattern of multiple T2 hyperintensities predominantly in the and limb use.77,78 In one series of over 100 children in
subcortical white matter of the right hemisphere. FLAIR, fluid-atte- Malaysia, those with poor outcome at hospital discharge
nuated inversion recovery. were more likely to have experienced seizures before or at
hospital admission, to have a faster heart rate, to have
uncontrolled seizures, thrombocytopenia, and hospital- required intubation, to have a reduced Glasgow coma score,
acquired infections.61–63 An EEG has been found to have and to have abnormal motor signs.79 Despite the prevalence
some value, since normal EEG early in admission is associated and severity of JE, however, neuroimaging biomarkers have
with positive outcomes.64 There has also been interest in the not been well characterized. Three patients with physical
development of blood and CSF-based biomarkers of prognosis, and cognitive disability 3 years following onset of JE showed
including elevated levels of neural proteins or inflammatory continued abnormal signal intensities on MRI in the thala-
cytokines which may correlate with poor outcome.65–67 Over- mus, basal ganglia, and brainstem, as might be expected
all, however, there exists a relative paucity of clinically useful given the prominent deep gray abnormalities seen in the
biomarkers for prognosis following encephalitis and thus there acute setting.80 Overall, loss of clinical follow-up and lack of
is considerable interest in developing neuroimaging biomar- ready access to MRI have likely contributed to the dearth of
kers. Below we will describe outcomes and neuroimaging neuroimaging data following JE.
correlates for selected infectious and autoimmune causes of While the mortality following anti-NMDAR encephalitis
encephalitis. While there are few established neuroimaging patients is low if the disease is identified early and immu-
biomarkers for prognosis, the discussion will focus on promis- notherapy is instituted, many survivors suffer from memory
ing approaches and challenges. deficits and executive dysfunction.81–83 FDG-PET may shed
The mortality rate for patients with HSV encephalitis is up light on prognosis, as the time to normalization of occipital
to 15% despite treatment with acyclovir, with the majority of hypometabolism may correlate with outcomes in some
survivors experiencing severe neurologic and cognitive patients with anti-NMDAR encephalitis.55,82 In addition, struc-
sequelae.9,68,69 Clinical factors associated with poor prog- tural and functional neuroimaging correlates to neurocogni-
nosis include delayed initiation of acyclovir, older age, a tive dysfunction following anti-NMDAR encephalitis are being
lower Glasgow coma scale on admission, seizures, and devel- vigorously explored.52 In a structural MRI study of 40 anti-
opment of hospital-acquired infections.68–70 From a neuroi- NMDAR encephalitis patients who were clinically considered
maging perspective, the extent and location of injury to the to be past the acute phase of the disease, hippocampal
temporal lobes have been shown to correlate with dysfunc- volumetry demonstrated atrophy of the entire left and right
tion. A study of long-term sequelae in 11 HSV encephalitic hippocampus along with CA4/DG, subiculum, and presubicu-
individuals demonstrated that MRI findings of left-sided lum.81 Atrophy of the CA1 and CA2/3 subfields of the right
amygdala damage and reduction in hippocampal and amyg- hippocampus and reduction in fimbria volume of the left
dala volume71 and bilateral temporal lobe swelling were hippocampus was also present. Additionally, hippocampal

Seminars in Neurology Vol. 39 No. 3/2019


318 Imaging in Encephalitis Venkatesan, Jagdish

microstructure integrity, as measured by mean diffusivity, was up in larger studies. fMRI performed on 27 postacute stage
abnormal. These data highlight the broad impact of anti- anti-LGl1 encephalitis individuals also demonstrated that
NMDAR encephalitis on hippocampal input, output, and the disease affects a wider range of brain regions than the
microstructural integrity. Moreover, there were strong corre- limbic system.87 The principal findings were of increased
lations of hippocampal volume and mean diffusivity with connectivity in the ventral and dorsal default mode network,
verbal and visuospatial memory performance, and thus these and decreased connectivity within the salience network, all
measures have potential as radiological biomarkers of cogni- associated with better memory performance. The authors
tive function following anti-NMDAR encephalitis.81 hypothesized that these changes in functional connectivity
Functional MRI (fMRI) measures have also been found to may arise as secondary or compensatory changes to focal
correlate with neurocognitive dysfunction following anti- damage to the hippocampus or alternatively as a direct result
NMDAR encephalitis. In a study of 24 individuals with post- of more global effects of the LGI1 antibody. The latter is
acute phase anti-NMDAR encephalitis, fMRI analysis showed supported by the finding that while LGI1 is predominantly
significantly reduced connectivity between the anterior expressed in the hippocampus, it is also found to a lesser
default mode network (comprised of the medial prefrontal extent in the inner layers of the cortex.94 Taken together,
cortex, anterior cingulate cortex, parts of the posterior cingu- these structural and functional studies support the notion
late cortex, and parts of the medial temporal lobes) and the that LGI encephalitis causes widespread brain dysfunction
anterior hippocampus bilaterally.52 This reduction of func- and reorganization and point the way toward radiological
tional connectivity strongly correlated with patients’ memory measures that may serve as useful correlates for outcomes.
performance.52 Within the same study, diffusion tensor ima-
ging showed alterations in white matter integrity especially in
Conclusions

Downloaded by: Kent State University. Copyrighted material.


the cingulum bundle, which has been associated with impair-
ments of working memory and executive function.52 More Despite recent advancements in diagnosis and treatment,
recently, an observational study of 43 individuals past the encephalitis continues to pose a substantial challenge for
acute phase of anti-NMDAR encephalitis again demonstrated clinicians. Neuroimaging can play an important role in con-
altered functional connectivity. fMRI showed decoupling of firming that a patient indeed has encephalitis and in defining
the medial temporal and the default-mode networks, impaired the specific etiology. While our knowledge of outcomes follow-
frontotemporal connectivity, and reduced hippocampal func- ing encephalitis is still quite rudimentary, correlations with
tional connectivity. Functional connectivity was also impaired structural and functional neuroimaging parameters have
within distributed large-scale networks throughout the shown some promise. While conventional MRI has proven
brain.84 Memory impairment was shown to correlate with useful to our understanding of causes and outcomes in ence-
hippocampal and medial temporal lobe network connectivity, phalitis, additional neuroimaging modalities such as PET ima-
while schizophrenia-like symptoms (i.e., hallucinations, delu- ging and fMRI may prove complementary and are likely to
sions, mutism, catatonia) correlated with impairments in contribute to a mechanistic understanding of postencephalitis
functional connectivity in the frontoparietal networks.84 brain reorganization and function. Larger cross-sectional stu-
Longitudinal studies will be necessary to determine how these dies, as well as longitudinal studies that correlate imaging with
network changes evolve over time. long-term function, are needed to better define and improve
The 2-year case fatality rate in those with LGI1 encepha- patient outcomes for this often devastating condition.
litis is up to 19%, and immune-mediated relapses are com-
mon, occurring in up to one-third of cases.85,86 Survivors Conflict of Interest
report amnesia, spatial disorientation, and insomnia, and None.
have been found to have deficiencies in working memory and
visuospatial abilities.85–87 Clinical predictors of poor out-
come include absence of response to initial immunotherapy
and the development of clinical relapses.88 During the acute References
stage of the disease, hippocampal T2/FLAIR hyperintense 1 Vallabhaneni D, Naveed MA, Mangla R, Zidan A, Mehta RI. Perfu-
signal abnormalities are observed in most patients and often sion imaging in autoimmune encephalitis. Case Rep Radiol 2018;
evolve into hippocampal atrophy with persistent cognitive 2018:3538645
dysfunction.89–91 Global atrophy has also been noted follow- 2 Graus F, Titulaer MJ, Balu R, et al. A clinical approach to diagnosis of
autoimmune encephalitis. Lancet Neurol 2016;15(04):391–404
ing the acute stage.92,93 In a study of nine patients in whom
3 Steiner I, Budka H, Chaudhuri A, et al. Viral encephalitis: a review of
multimodal MRI was performed, diffusion tensor imaging diagnostic methods and guidelines for management. Eur J Neurol
demonstrated widespread increases in fractional anisotropy 2005;12(05):331–343
throughout the brain, reflecting decreased white matter 4 Schroth G, Kretzschmar K, Gawehn J, Voigt K. Advantage of
integrity and corroborating the notion that LGI1 affects the magnetic resonance imaging in the diagnosis of cerebral infec-
brain in a global fashion.93 Interestingly, better outcomes on tions. Neuroradiology 1987;29(02):120–126
5 Marchbank ND, Howlett DC, Sallomi DF, Hughes DV. Magnetic
modified Rankin Scale were associated with higher cerebel-
resonance imaging is preferred in diagnosing suspected cerebral
lar gray matter volume, while increased cognitive capacity infections. BMJ 2000;320(7228):187–188
was associated with higher putaminal volumes; the signifi- 6 Kiroğlu Y, Calli C, Yunten N, et al. Diffusion-weighted MR imaging
cance of these findings is unclear and will need to be followed of viral encephalitis. Neuroradiology 2006;48(12):875–880

Seminars in Neurology Vol. 39 No. 3/2019


Imaging in Encephalitis Venkatesan, Jagdish 319

7 Tsuchiya K, Katase S, Yoshino A, Hachiya J. Diffusion-weighted MR 27 Ooi MH, Wong SC, Lewthwaite P, Cardosa MJ, Solomon T. Clinical
imaging of encephalitis. AJR Am J Roentgenol 1999;173(04): features, diagnosis, and management of enterovirus 71. Lancet
1097–1099 Neurol 2010;9(11):1097–1105
8 Venkatesan A, Tunkel AR, Bloch KC, et al; International Encepha- 28 Jain H, Deshpande A, Favaz AM, Rajagopal KV. MRI in rabies
litis Consortium Case definitions, diagnostic algorithms, and encephalitis. BMJ Case Rep 2013;2013. doi: 10.1136/bcr-2013-
priorities in encephalitis: consensus statement of the interna- 201825
tional encephalitis consortium. Clin Infect Dis 2013;57(08): 29 Handique SK, Das RR, Barman K, et al. Temporal lobe involvement
1114–1128 in Japanese encephalitis: problems in differential diagnosis. AJNR
9 Bertrand A, Leclercq D, Martinez-Almoyna L, Girard N, Stahl JP, Am J Neuroradiol 2006;27(05):1027–1031
De-Broucker T. MR imaging of adult acute infectious encephalitis. 30 Basumatary LJ, Raja D, Bhuyan D, Das M, Goswami M, Kayal AK.
Med Mal Infect 2017;47(03):195–205 Clinical and radiological spectrum of Japanese encephalitis. J Neurol
10 Rath TJ, Hughes M, Arabi M, Shah GV. Imaging of cerebritis, Sci 2013;325(1–2):15–21
encephalitis, and brain abscess. Neuroimaging Clin N Am 2012; 31 Dale RC, Merheb V, Pillai S, et al. Antibodies to surface dopamine-2
22(04):585–607 receptor in autoimmune movement and psychiatric disorders.
11 Defres S, Keller SS, Das K, et al; ENCEPH UK study group A Brain 2012;135(Pt 11):3453–3468
feasibility study of quantifying longitudinal brain changes in 32 Jubelt B, Mihai C, Li TM, Veerapaneni P. Rhombencephalitis/
herpes simplex virus (HSV) encephalitis using magnetic reso- brainstem encephalitis. Curr Neurol Neurosci Rep 2011;11(06):
nance imaging (MRI) and stereology. PLoS One 2017;12(01): 543–552
e0170215 33 Tan IL, Mowry EM, Steele SU, et al. Brainstem encephalitis:
12 Chow FC, Glaser CA, Sheriff H, et al. Use of clinical and neuroima- etiologies, treatment, and predictors of outcome. J Neurol 2013;
ging characteristics to distinguish temporal lobe herpes simplex 260(09):2312–2319
encephalitis from its mimics. Clin Infect Dis 2015;60(09): 34 Charlier C, Poirée S, Delavaud C, et al; MONALISA Study Group
1377–1383 Imaging of human neurolisteriosis: a prospective study of 71

Downloaded by: Kent State University. Copyrighted material.


13 Tan IL, McArthur JC, Venkatesan A, Nath A. Atypical manifesta- cases. Clin Infect Dis 2018;67(09):1419–1426
tions and poor outcome of herpes simplex encephalitis in the 35 Moragas M, Martínez-Yélamos S, Majós C, Fernández-Viladrich P,
immunocompromised. Neurology 2012;79(21):2125–2132 Rubio F, Arbizu T. Rhombencephalitis: a series of 97 patients.
14 Whitley RJ, Cobbs CG, Alford CA Jr, et al; NIAD Collaborative Medicine (Baltimore) 2011;90(04):256–261
Antiviral Study Group Diseases that mimic herpes simplex ence- 36 Chen F, Liu T, Li J, Xing Z, Huang S, Wen G. MRI characteristics and
phalitis. Diagnosis, presentation, and outcome. JAMA 1989;262 follow-up findings in patients with neurological complications of
(02):234–239 enterovirus 71-related hand, foot, and mouth disease. Int J Clin
15 Kelley BP, Patel SC, Marin HL, Corrigan JJ, Mitsias PD, Griffith B. Exp Med 2014;7(09):2696–2704
Autoimmune encephalitis: pathophysiology and imaging review 37 Compain C, Sacre K, Puéchal X, et al. Central nervous system
of an overlooked diagnosis. AJNR Am J Neuroradiol 2017;38(06): involvement in Whipple disease: clinical study of 18 patients and
1070–1078 long-term follow-up. Medicine (Baltimore) 2013;92(06):324–330
16 da Rocha AJ, Nunes RH, Maia AC Jr, do Amaral LL. Recognizing 38 Black DF, Aksamit AJ, Morris JM. MR imaging of central nervous
autoimmune-mediated encephalitis in the differential diagnosis system Whipple disease: a 15-year review. AJNR Am J Neuroradiol
of limbic disorders. AJNR Am J Neuroradiol 2015;36(12): 2010;31(08):1493–1497
2196–2205 39 Lu Z, Zhang B, Qiu W, et al. Comparative brain stem lesions on MRI
17 Gultekin SH, Rosenfeld MR, Voltz R, Eichen J, Posner JB, Dalmau J. of acute disseminated encephalomyelitis, neuromyelitis optica,
Paraneoplastic limbic encephalitis: neurological symptoms, and multiple sclerosis. PLoS One 2011;6(08):e22766
immunological findings and tumour association in 50 patients. 40 Tobin WO, Guo Y, Krecke KN, et al. Diagnostic criteria for chronic
Brain 2000;123(Pt 7):1481–1494 lymphocytic inflammation with pontine perivascular enhance-
18 Dalmau J, Graus F, Villarejo A, et al. Clinical analysis of anti-Ma2- ment responsive to steroids (CLIPPERS). Brain 2017;140(09):
associated encephalitis. Brain 2004;127(Pt 8):1831–1844 2415–2425
19 Heine J, Prüss H, Bartsch T, Ploner CJ, Paul F, Finke C. Imaging of 41 Gilden D, Cohrs RJ, Mahalingam R, Nagel MA. Varicella zoster virus
autoimmune encephalitis–relevance for clinical practice and vasculopathies: diverse clinical manifestations, laboratory features,
hippocampal function. Neuroscience 2015;309:68–83 pathogenesis, and treatment. Lancet Neurol 2009;8(08):731–740
20 Saiz A, Blanco Y, Sabater L, et al. Spectrum of neurological 42 Bradshaw MJ, Lalor KB, Vu N, Pruthi S, Bloch KC. Child neurology:
syndromes associated with glutamic acid decarboxylase antibo- Rocky Mountain spotted fever encephalitis. Neurology 2017;88
dies: diagnostic clues for this association. Brain 2008;131(Pt (11):e92–e95
10):2553–2563 43 Agarwal R, Sze G. Neuro-lyme disease: MR imaging findings.
21 Höftberger R, Titulaer MJ, Sabater L, et al. Encephalitis and GABAB Radiology 2009;253(01):167–173
receptor antibodies: novel findings in a new case series of 20 44 Agosta F, Rocca MA, Benedetti B, Capra R, Cordioli C, Filippi M. MR
patients. Neurology 2013;81(17):1500–1506 imaging assessment of brain and cervical cord damage in patients
22 Knox J, Cowan RU, Doyle JS, et al. Murray Valley encephalitis: a with neuroborreliosis. AJNR Am J Neuroradiol 2006;27(04):892–894
review of clinical features, diagnosis and treatment. Med J Aust 45 Sahraian MA, Radue EW, Eshaghi A, Besliu S, Minagar A. Progres-
2012;196(05):322–326 sive multifocal leukoencephalopathy: a review of the neuroima-
23 Wong SH, Smith DW, Fallon MJ, Kermode AG. Murray valley ging features and differential diagnosis. Eur J Neurol 2012;19(08):
encephalitis mimicking herpes simplex encephalitis. J Clin Neu- 1060–1069
rosci 2005;12(07):822–824 46 Berger JR, Aksamit AJ, Clifford DB, et al. PML diagnostic criteria:
24 Deresiewicz RL, Thaler SJ, Hsu L, Zamani AA. Clinical and neuror- consensus statement from the AAN Neuroinfectious Disease
adiographic manifestations of eastern equine encephalitis. N Engl Section. Neurology 2013;80(15):1430–1438
J Med 1997;336(26):1867–1874 47 Bulakbasi N, Kocaoglu M, Tayfun C, Ucoz T. Transient splenial
25 Beattie GC, Glaser CA, Sheriff H, et al. Encephalitis with thalamic and lesion of the corpus callosum in clinically mild influenza-asso-
basal ganglia abnormalities: etiologies, neuroimaging, and poten- ciated encephalitis/encephalopathy. AJNR Am J Neuroradiol
tial role of respiratory viruses. Clin Infect Dis 2013;56(06):825–832 2006;27(09):1983–1986
26 Solomon T, Kneen R, Dung NM, et al. Poliomyelitis-like illness due to 48 Gallucci M, Limbucci N, Paonessa A, Caranci F. Reversible focal
Japanese encephalitis virus. Lancet 1998;351(9109):1094–1097 splenial lesions. Neuroradiology 2007;49(07):541–544

Seminars in Neurology Vol. 39 No. 3/2019


320 Imaging in Encephalitis Venkatesan, Jagdish

49 Young NP, Weinshenker BG, Parisi JE, et al. Perivenous demyelina- treatment at the onset of herpetic meningoencephalitis. Anti-
tion: association with clinically defined acute disseminated microb Agents Chemother 2015;59(06):3084–3089
encephalomyelitis and comparison with pathologically con- 71 Caparros-Lefebvre D, Girard-Buttaz I, Reboul S, et al. Cognitive
firmed multiple sclerosis. Brain 2010;133(Pt 2):333–348 and psychiatric impairment in herpes simplex virus encephalitis
50 Abou Zeid N, Pirko I, Erickson B, et al. Diffusion-weighted imaging suggest involvement of the amygdalo-frontal pathways. J Neurol
characteristics of biopsy-proven demyelinating brain lesions. 1996;243(03):248–256
Neurology 2012;78(21):1655–1662 72 Reed LJ, Lasserson D, Marsden P, Bright P, Stanhope N, Kopelman
51 Spatola M, Petit-Pedrol M, Simabukuro MM, et al. Investigations in MD. Correlations of regional cerebral metabolism with memory
GABAA receptor antibody-associated encephalitis. Neurology 2017; performance and executive function in patients with herpes
88(11):1012–1020 encephalitis or frontal lobe lesions. Neuropsychology 2005;19
52 Finke C, Kopp UA, Scheel M, et al. Functional and structural brain (05):555–565
changes in anti-N-methyl-D-aspartate receptor encephalitis. Ann 73 Singh TD, Fugate JE, Hocker S, Wijdicks EFM, Aksamit AJ Jr,
Neurol 2013;74(02):284–296 Rabinstein AA. Predictors of outcome in HSV encephalitis.
53 Wang R, Lai XH, Liu X, et al. Brain magnetic resonance-imaging J Neurol 2016;263(02):277–289
findings of anti-N-methyl-D-aspartate receptor encephalitis: a 74 Davis LE, DeBiasi R, Goade DE, et al. West Nile virus neuroinvasive
cohort follow-up study in Chinese patients. J Neurol 2018;265(02): disease. Ann Neurol 2006;60(03):286–300
362–369 75 Murray KO, Nolan MS, Ronca SE, et al. The neurocognitive and MRI
54 Zhang T, Duan Y, Ye J, et al. Brain MRI characteristics of patients outcomes of West Nile virus infection: preliminary analysis using
with anti-N-methyl-D-aspartate receptor encephalitis and their an external control group. Front Neurol 2018;9:111
associations with 2-year clinical outcome. AJNR Am J Neuroradiol 76 Sejvar JJ. Clinical manifestations and outcomes of West Nile virus
2018;39(05):824–829 infection. Viruses 2014;6(02):606–623
55 Probasco JC, Solnes L, Nalluri A, et al. Decreased occipital lobe 77 Griffiths MJ, Lemon JV, Rayamajhi A, et al. The functional, social and
metabolism by FDG-PET/CT: an anti-NMDA receptor encephalitis economic impact of acute encephalitis syndrome in Nepal–a long-

Downloaded by: Kent State University. Copyrighted material.


biomarker. Neurol Neuroimmunol Neuroinflamm 2017;5(01):e413 itudinal follow-up study. PLoS Negl Trop Dis 2013;7(09):e2383
56 Irani SR, Michell AW, Lang B, et al. Faciobrachial dystonic seizures 78 Cao L, Fu S, Gao X, et al. Low protective efficacy of the current
precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011;69 Japanese encephalitis vaccine against the emerging genotype 5
(05):892–900 Japanese encephalitis virus. PLoS Negl Trop Dis 2016;10(05):
57 Flanagan EP, Kotsenas AL, Britton JW, et al. Basal ganglia T1 e0004686
hyperintensity in LGI1-autoantibody faciobrachial dystonic sei- 79 Ooi MH, Lewthwaite P, Lai BF, et al. The epidemiology, clinical
zures. Neurol Neuroimmunol Neuroinflamm 2015;2(06):e161 features, and long-term prognosis of Japanese encephalitis in
58 Dodich A, Cerami C, Iannaccone S, et al. Neuropsychological and central sarawak, malaysia, 1997-2005. Clin Infect Dis 2008;47
FDG-PET profiles in VGKC autoimmune limbic encephalitis. Brain (04):458–468
Cogn 2016;108:81–87 80 Shoji H, Murakami T, Murai I, et al. A follow-up study by CT and
59 Venkatesan A. Epidemiology and outcomes of acute encephalitis. MRI in 3 cases of Japanese encephalitis. Neuroradiology 1990;32
Curr Opin Neurol 2015;28(03):277–282 (03):215–219
60 Lancaster E. The diagnosis and treatment of autoimmune ence- 81 Finke C, Kopp UA, Pajkert A, et al. Structural hippocampal damage
phalitis. J Clin Neurol 2016;12(01):1–13 following anti-N-methyl-D-aspartate receptor encephalitis. Biol
61 Sonneville R, Gault N, de Montmollin E, et al. Clinical spectrum Psychiatry 2016;79(09):727–734
and outcomes of patients with encephalitis requiring intensive 82 Finke C, Kopp UA, Prüss H, Dalmau J, Wandinger KP, Ploner CJ.
care. Eur J Neurol 2015;22(01):6–16, e1 Cognitive deficits following anti-NMDA receptor encephalitis.
62 Thakur KT, Motta M, Asemota AO, et al. Predictors of outcome in J Neurol Neurosurg Psychiatry 2012;83(02):195–198
acute encephalitis. Neurology 2013;81(09):793–800 83 Yeshokumar AK, Gordon-Lipkin E, Arenivas A, et al. Neurobeha-
63 Singh TD, Fugate JE, Rabinstein AA. The spectrum of acute vioral outcomes in autoimmune encephalitis. J Neuroimmunol
encephalitis: causes, management, and predictors of outcome. 2017;312:8–14
Neurology 2015;84(04):359–366 84 Peer M, Prüss H, Ben-Dayan I, Paul F, Arzy S, Finke C. Functional
64 Sutter R, Kaplan PW, Cervenka MC, et al. Electroencephalography connectivity of large-scale brain networks in patients with anti-
for diagnosis and prognosis of acute encephalitis. Clin Neurophy- NMDA receptor encephalitis: an observational study. Lancet
siol 2015;126(08):1524–1531 Psychiatry 2017;4(10):768–774
65 Tsukahara H, Fujii Y, Matsubara K, et al. Prognostic value of brain 85 Li W, Wu S, Meng Q, et al. Clinical characteristics and short-term
injury biomarkers in acute encephalitis/encephalopathy. Pediatr prognosis of LGI1 antibody encephalitis: a retrospective case
Int 2013;55(04):461–464 study. BMC Neurol 2018;18(01):96
66 Sellner J, Davies NW, Howard RS, Petzold A. Neurofilament heavy 86 van Sonderen A, Thijs RD, Coenders EC, et al. Anti-LGI1 encepha-
chain as a marker of neuroaxonal pathology and prognosis in litis: clinical syndrome and long-term follow-up. Neurology
acute encephalitis. Eur J Neurol 2014;21(06):845–850 2016;87(14):1449–1456
67 Michael BD, Griffiths MJ, Granerod J, et al. The interleukin-1 87 Heine J, Prüss H, Kopp UA, et al. Beyond the limbic system:
balance during encephalitis is associated with clinical severity, disruption and functional compensation of large-scale brain
blood-brain barrier permeability, neuroimaging changes, and networks in patients with anti-LGI1 encephalitis. J Neurol Neu-
disease outcome. J Infect Dis 2016;213(10):1651–1660 rosurg Psychiatry 2018;89(11):1191–1199
68 Sili U, Kaya A, Mert A; HSV Encephalitis Study Group Herpes 88 Ariño H, Armangué T, Petit-Pedrol M, et al. Anti-LGI1-associated
simplex virus encephalitis: clinical manifestations, diagnosis cognitive impairment: presentation and long-term outcome.
and outcome in 106 adult patients. J Clin Virol 2014;60(02): Neurology 2016;87(08):759–765
112–118 89 Finke C, Prüss H, Heine J, et al. Evaluation of cognitive deficits and
69 Raschilas F, Wolff M, Delatour F, et al. Outcome of and prognostic structural hippocampal damage in encephalitis with leucine-rich,
factors for herpes simplex encephalitis in adult patients: results of glioma-inactivated 1 antibodies. JAMA Neurol 2017;74(01):50–59
a multicenter study. Clin Infect Dis 2002;35(03):254–260 90 Navarro V, Kas A, Apartis E, et al; collaborators Motor cortex and
70 Erdem H, Cag Y, Ozturk-Engin D, et al. Results of a multinational hippocampus are the two main cortical targets in LGI1-antibody
study suggest the need for rapid diagnosis and early antiviral encephalitis. Brain 2016;139(Pt 4):1079–1093

Seminars in Neurology Vol. 39 No. 3/2019


Imaging in Encephalitis Venkatesan, Jagdish 321

91 Gadoth A, Pittock SJ, Dubey D, et al. Expanded phenotypes and 93 Szots M, Blaabjerg M, Orsi G, et al. Global brain atrophy and
outcomes among 256 LGI1/CASPR2-IgG-positive patients. Ann metabolic dysfunction in LGI1 encephalitis: a prospective multi-
Neurol 2017;82(01):79–92 modal MRI study. J Neurol Sci 2017;376:159–165
92 Irani SR, Stagg CJ, Schott JM, et al. Faciobrachial dystonic seizures: 94 Herranz-Pérez V, Olucha-Bordonau FE, Morante-Redolat JM,
the influence of immunotherapy on seizure control and preven- Pérez-Tur J. Regional distribution of the leucine-rich glioma
tion of cognitive impairment in a broadening phenotype. Brain inactivated (LGI) gene family transcripts in the adult mouse brain.
2013;136(Pt 10):3151–3162 Brain Res 2010;1307:177–194

Downloaded by: Kent State University. Copyrighted material.

Seminars in Neurology Vol. 39 No. 3/2019

You might also like