You are on page 1of 27

Handbook of Clinical Neurology, Vol.

133 (3rd series)


Autoimmune Neurology
S.J. Pittock and A. Vincent, Editors
© 2016 Elsevier B.V. All rights reserved

Chapter 13

Autoimmune epilepsy
JEFFREY BRITTON*
Department of Neurology, Mayo Clinic, Rochester, MN, USA

Abstract
Seizures are a common manifestation of autoimmune limbic encephalitis and multifocal paraneoplastic
disorders. Accumulating evidence supports an autoimmune basis for seizures in the absence of syndromic
manifestations of encephalitis. The autoimmune epilepsies are immunologically mediated disorders in
which recurrent seizures are a primary and persistent clinical feature. When other etiologies have been
excluded, an autoimmune etiology is suggested in a patient with epilepsy upon detection of neural auto-
antibodies and/or the presence of inflammatory changes on cerebrospinal fluid (CSF) or magnetic reso-
nance imaging. In such patients, immunotherapy may be highly effective, depending on the particular
autoimmune epilepsy syndrome present. In this chapter, several autoimmune epilepsy syndromes are dis-
cussed. First, epilepsies secondary to other primary autoimmune disorders will be discussed, and then
those associated with antibodies that are likely to be pathogenic, such as voltage-gated potassium chan-
nel–complex and N-methyl-D-aspartate receptor, gamma-aminobutyric acid A and B receptor antibodies.
For each syndrome, the typical clinical, imaging, electroencephaloram, CSF, and serologic features, and
pathophysiology and treatment are described. Finally, suggested guidelines for the recognition, evalua-
tion, and treatment of autoimmune epilepsy syndromes are provided.

2014), and the fact that certain forms of epilepsy, such as


INTRODUCTION
infantile spasms and Landau–Kleffner syndrome, may
Epilepsy affects 65 million people worldwide, of whom respond dramatically to immunotherapy (Sinclair and
30% remain inadequately controlled despite best available Snyder, 2005; Arts et al., 2009; Go et al., 2012). Further
therapy (England et al., 2012). Identification of a treatable evidence for the potential role of immune mechanisms
etiology, such as a surgically remediable lesion, may be the in the pathogenesis of seizures was established by the
only chance to achieve a significant seizure reduction in discovery of paraneoplastic limbic encephalitis in the
such patients. Similarly, the diagnosis of an autoimmune 1960s (Brierley et al., 1960; Corsellis et al., 1968). The sub-
etiology in a patient with medically intractable epilepsy sequent identification of circulating neural antigen-
opens up the possibility of immunotherapy, which would targeting antibodies in these patients provided further
otherwise not usually be considered. insight as to the potential pathogenesis of these disorders
The role of autoimmunity in certain subgroups of (Graus et al., 1986; Dalmau et al., 1992). And most
epilepsy has been recognized for decades, from the dis- recently, the favorable response to therapy in certain non-
covery of the inflammatory pathogenesis of Rasmussen’s paraneoplastic encephalitides with detectable neuronal
encephalitis (Rasmussen et al., 1958), to the observation surface antibodies has led to an increasing interest in iden-
of the occurrence of seizures as a relatively common tifying such disorders (Graus and Saiz, 2008).
central nervous system (CNS) manifestation of systemic Antibodies to an expanding number of neural anti-
autoimmune diseases such as lupus (Mikdashi et al., 2005; gens are now recognized, and significant improvements
González-Duarte et al., 2008; Hanly et al., 2012; Tsai et al., have been achieved in the commercialization of tests

*Correspondence to: Jeffrey Britton, MD, Associate Professor, Neurology, Mayo Clinic, 200 1st St SW, Rochester MN 55905, USA.
Tel: +1-507-538-1036, Fax: +1-407-266-4419, E-mail: britton.jeffrey@mayo.edu
220 J. BRITTON
and availability of testing. As a result, increasing numbers Epidemiology
of patients are being diagnosed with autoimmune epilepsy
Rasmussen’s encephalitis is uncommon, with an incidence
syndromes (Peltola et al., 2000; Tüzün and Dalmau, 2007;
of 2.4 per 107 persons under age 18 years per year (Bien
Graus and Saiz, 2008; Graus et al., 2008; Liimatainen et al.,
et al., 2013). Rasmussen’s encephalitis typically affects
2010; Quek et al., 2012). In addition, correlations between
children, although late-onset cases have been described.
epilepsy and several other autoimmune diseases are being
identified, suggesting that autoimmune mechanisms may
play a broader role in the pathogenesis of epilepsy (Ong Clinical presentation, course, and prognosis
et al., 2014). Rasmussen’s encephalitis presents with focal seizures
In this chapter, the clinical features of several autoim- involving one side of the body. The seizures are typically
mune epilepsy syndromes are presented. Current ther- focal motor clonic in nature, and can involve the face,
apy will also be discussed. A list of the autoimmune the extremities, trunk, or abdomen. A low-grade fever
epilepsy syndromes is shown in Table 13.1. A more may be present at its inception. The seizures typically pro-
detailed discussion of these syndromes follows. gress over time, often evolving to focal status epilepticus
(epilepsy partialis continua). A spastic hemiparesis typi-
RASMUSSEN’S ENCEPHALITIS cally develops, and this can become severe. Neurologic
Rasmussen’s encephalitis is an asymmetric encephalitis decline typically develops as seizures continue. Antiepi-
that presents with intractable focal seizures, progressive leptic drug therapy is usually modestly effective, and sei-
hemiparesis, and unilateral hemispheric atrophy. It was zures typically continue despite treatment. Progression
first reported in the literature in 1958 (Rasmussen et al., of the disease usually occurs over months but can
1958). The cause of Rasmussen’s encephalitis is unknown, “burn itself out” after a period of time, leaving the
but an autoimmune etiology is suspected. The condition is patient with a static hemiplegia and residual focal motor
poorly responsive to anticonvulsant therapy. seizures. Such patients may live for many years after ces-
sation of the “active” phase of the disease. However, if
Table 13.1 seizures continue and neurologic deterioration con-
tinues, the disease can prove fatal. Figure 13.1 illustrates
Autoimmune epilepsy syndromes the typical clinical course of Rasmussen’s encephalitis.
Primary autoimmune epilepsies
Rasmussen’s encephalitis Diagnostic evaluation
Immunotherapy-responsive epilepsies
Epileptic spasms (West syndrome) SEROLOGY
Landau–Kleffner syndrome
There is no serologic testing specific for the diagnosis of
Systemic autoimmune disorders associated with epilepsy
Systemic lupus erythematosus
Rasmussen’s encephalitis.
Celiac disease
Sj€
ogren’s syndrome CEREBROSPINAL FLUID
Linear scleroderma (Perry–Romberg)
Although the inflammatory nature of Rasmussen’s is
Other
Antibody-mediated epilepsy syndromes clear, cerebrospinal fluid (CSF) examination is normal
Antibodies targeting cell surface antigens in over 50% of cases at time of evaluation.
Voltage-gated potassium channel (VGKC)
N-methyl-D-aspartate (NMDA) NEUROIMAGING
Glutamic acid decarboxylase 65 kD isoform (GAD-65; this is an
intracellular target but can be associated with other cell Magnetic resonance imaging (MRI) and computed
surface antibodies) tomography (CT) imaging of the brain typically shows
Gamma-aminobutyric acid-B (GABA-B) unilateral hemispheric atrophy (Fig. 13.2). Serial imaging
Gamma-aminobutyric acid-A (GABA-A) may show progression of atrophy over time. In addition
a-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid to hemispheric atrophy, MRI may show increased T2
(AMPA) signal and swelling of the cortical gray matter over the
Acetylcholine receptor (AChR) ganglionic affected hemisphere (Bien et al., 2002b). Despite
Antibodies targeting intracellular antigens
the presence of inflammation, contrast enhancement is
Type 1 antineuronal nuclear antibody (ANNA-1) (Hu)
Ma1/Ma2
typically absent (Bien et al., 2005). Fluorodeoxyglucose
Collapsin response mediator protein-5 (CRMP-5) positron emission tomography (FDG-PET) may show
Seronegative autoimmune limbic encephalitis hypermetabolic abnormalities if seizures occur during
FDG administration (Bien et al., 2005).
AUTOIMMUNE EPILEPSY 221
Immunotherapy
Hemisphere volume EPC

Motor function
Inflammation

Seizures

Prodromal stage Acute stage (8-12 months) Residual stage

Fig. 13.1. Natural clinical course and expected effect of immunotherapy. The natural clinical course of Rasmussen’s encephalitis
was characterized in the past century. The disease might have a preceding prodromal stage with infrequent seizures, and presents
with an acute stage of drug-resistant epilepsy. The epilepsy is characterized by very frequent seizures of different semiologies in the
same patient, often epilepsia partialis continua, with the emergence of a fluctuating, then permanent, hemiplegia (motor function)
and concurrent progressive hemispheric volume loss on neuroimaging. With the advent of immunotherapy, the natural clinical
course seems to be changing. The rate of motor function and hemispheric volume loss is slowed, and seizures decrease in frequency
and plateau. Cognitive deterioration is not shown because it is more variable, although usually becomes manifest during the acute
phase. EPC, epilepsia partialis continua. (Reproduced from Varadkar et al., 2014, with permission.)

EEG brain, cerebral infarctions, and porencephalic cysts, all


of which may cause focal seizures in childhood. Acutely,
Electroencephalogram (EEG) usually shows interictal
infectious causes of meningoencephalitis need to be
epileptiform abnormalities and intermittent or continu-
considered. Mitochondrial disorders may also present
ous focal seizure activity over the affected hemisphere
with focal seizures and status epilepticus. Neuroimaging
(Longaretti et al., 2012). Asymmetric delta slowing
is an important step in the evaluation of these patients to
may also be present. Although the EEG may show focal
help exclude many of the aforementioned diagnoses.
or asymmetric abnormalities commensurate with the
unilateral nature of the underlying encephalitis, EEG
Pathophysiology
abnormalities may also be present over the unaffected
hemisphere due to secondary bilateral synchrony. The inflammatory nature of Rasmussen’s is indisput-
It should be noted that EEG will not always show a able. Pathology from autopsy and surgical specimens
confirmatory discharge during clinical seizures if the shows microglial nodules and perivascular mononuclear
seizure focus in question involves a small cortical region cell infiltration. Although the pathologic changes are sim-
localized remote from the scalp surface (Fattouch ilar to those seen in viral encephalitis, a specific infectious
et al., 2008). agent has never been implicated, and Rasmussen’s
encephalitis is currently considered an autoimmune dis-
order of unknown etiology and mechanisms (Varadkar
CANCER EVALUATION
et al., 2014). Elevations of interleukin-6 (IL-6) have been
Rasmussen’s encephalitis does not occur as paraneoplas- identified in CSF in Rasmussen’s encephalitis patients
tic syndrome and cancer evaluation is not warranted in (Tekgul et al., 2005). Inflammatory cytokine abnormali-
typical cases. ties have been described in the cortex of Rasmussen’s
encephalitis cases as well, with IL-1beta, IL-8, IL-12p70,
and macrophage inflammatory protein (MIP)-1beta
DIFFERENTIAL DIAGNOSIS
elevations identified in surgical specimens (Choi
Rasmussen’s encephalitis is very difficult to diagnose et al., 2009).
at onset but, over time, the clinical presentation is char- At one time, antibodies to GluR3 receptors were
acteristic. Early in the illness, other causes of focal thought to play a causative or pathogenic role in Rasmus-
seizures and status epilepticus need to be considered, sen’s encephalitis, when induction of antibodies targeting
including focal or lateralized malformations of cortical GluR3 antigens were shown to reproduce a Rasmussen’s
development, neoplasms, vascular malformations of the encephalitis illness in rabbits, and GluR3 antibodies were
222 J. BRITTON

Fig. 13.2. (A, B) Neuroimaging in Rasmussen’s encephalitis. Magnetic resonance imaging brain scans of children with Rasmus-
sen’s encephalitis, showing contrasting cases of radiologic progression. (A) Progressive right-hemisphere atrophy, high signal, and
basal ganglia loss over 1 year (from left to right) in a child with Rasmussen’s encephalitis. The disease was mostly centered near the
right Sylvian fissure (arrow). (B) Slowly progressive disease with more subtle right-hemisphere atrophy in a child on immuno-
suppressant treatment at 6 months (left), 18 months (center), and 30 months (right) of disease course. (C) Computed tomography of
the head in a 58-year-old patient with a history of Rasmussen’s encephalitis as a child, showing severe left-hemispheric atrophy. He
continues to have daily, fleeting focal motor seizures on his right, one to two major hemiconvulsive seizures per year, and a spastic
right hemiplegia. Used with permission of Mayo Foundation for Medical Education and Research. All rights reserved.

reported in serum of affected patients (Rogers et al., 2001; Mantegazza et al., 2002). Also, GluR3 antibodies
1994). However, subsequent evaluation, mainly using pep- were not observed in a large cohort using a range of assays
tide enzyme-linked immunosorbent assays, showed (Watson et al., 2004). Furthermore, analysis of the epi-
GluR3 antibodies to be present in other chronic epilepsies, topes of antibodies recognizing GluR episolon2 subunit
and absent in some or all cases of Rasmussen’s, eliminat- (another name for the N-methyl-D-aspartate receptor
ing them from being considered causative (Wiendl et al., (NMDAR) NR2B subunit) on Western blots was found
AUTOIMMUNE EPILEPSY 223
to be broad and with expansion as disease progressed, sug- having nondisabling seizures (Vining et al., 1997). Most
gesting that these autoantibodies may occur secondary to patients remain ambulatory despite variable severities
cytotoxic T-cell-mediated neuronal damage, rather than of hemiparesis.
being a primary causative factor (Takahashi et al., 2005).
The inflammatory response in Rasmussen’s enceph- SEIZURES OCCURRING IN SYSTEMIC
alitis is currently considered to be T-cell-mediated. AUTOIMMUNE DISEASES
Inspection of the inflammatory infiltrates in surgical
specimens shows a mixed infiltrate of dendritic cells Seizures are not uncommon in the setting of systemic
and cytotoxic CD3+/CD8 + T lymphocytes (Bauer autoimmune diseases (Vincent and Crino, 2011).
et al., 2002; Bien et al., 2002a; Rhodes et al., 2007). Li Although the mechanisms leading to seizures in these dis-
et al. (1997) reported that PCR analysis of T-cell receptor orders are likely multifactorial, antibodies targeting neu-
BV gene transcripts in surgical specimens suggested that ral antigens may play a role (Vincent et al., 2010). Seizures
the local immune responses in Rasmussen’s syndrome are more characteristic in some autoimmune disorders
includes restricted T-cell populations responding to than others; however, a study utilizing insurance claims
discrete antigenic epitopes. data from over 2.5 million beneficiaries showed the risk
of epilepsy to be 3.8 times higher among patients with a
variety of autoimmune diseases (Ong et al., 2014). These
Treatment associations were not only found in autoimmune diseases
Despite the clear involvement of inflammation in its in which a link with seizures was already established (e.g.,
pathogenesis, the response to immunosuppressive ther- systemic lupus erythematosus (SLE)), but also in diseases
apy is often disappointing. Several reports in the litera- in which a correlation had not previously been appreci-
ture attest to individuals or small groups of patients ated, such as myasthenia gravis, psoriasis, and Graves
responding to a variety of immunotherapy regimens, disease.
including corticosteroids and cyclophosphamide (Hart
et al., 1994; Krauss et al., 1996; Kashihara et al., 2010), Systemic lupus erythematosus (SLE)
intravenous immunoglobulin (IVIG) (Hart et al., 1994;
Neurologic symptoms, including seizures, are common in
Leach et al., 1999; Villani et al., 2001; Buenz and
SLE. The seizures associated with SLE can be disabling.
Howe, 2007), adrenocorticotropic hormone (Granata
et al., 2003), selective immunoglobulin G (IgG) immu-
noadsorption by protein A (Antozzi et al., 1998), and EPIDEMIOLOGY
plasmapheresis (Andrews et al., 2001). A randomized Seizures are relatively common in SLE. In a study using
trial of tacrolimus and IVIG showed lengthening of sur- insurance claims data, the adjusted hazard ratio for epi-
vival with either treatment, but disappointingly, no consis- lepsy in the SLE versus non-SLE population was 2.33
tent improvement in seizures (Bien et al., 2013). Finally, (95% confidence interval (CI) ¼ 1.89–2.88) (Tsai et al.,
there remains uncertainty as to whether Rasmussen’s 2014). The incidence was much higher in younger SLE
encephalitis is a primary autoimmune disease. In one patients: the hazard ratio for those aged 20 years
report, significant improvement was seen following the was 8.05 (95% CI ¼ 4.30–15.0).
administration of the antiviral ganciclovir in 2 of 3
patients treated within 3 months of disease onset. Five
CLINICAL PRESENTATION
additional patients treated 6 months or longer after dis-
ease onset did not respond. This raises the question as Seizures are one of 19 clinical features comprising
to whether Rasmussen’s encephalitis is triggered by a viral what is referred to as neuropsychiatric SLE (NPSLE)
etiology, which transforms into a chronic progressive pre- (ACR, 1999). Other CNS manifestations of NPSLE
dominantly immune-mediated disease following resolu- include headache, neurocognitive deficits, depression,
tion of the active infection (McLachlan et al., 2011). and psychosis. Seizures may be the presenting symptom
In most cases, however, immunotherapy is temporiz- of SLE. The most common seizure types seen in NPSLE
ing. In refractory cases, hemispherectomy or hemispher- are generalized tonic-clonic and complex partial sei-
otomy remains the most effective option for attaining zures. Affected patients may also present in status
seizure control and in arresting the neurologic deteriora- epilepticus. In one cohort, 14% of patients with SLE
tion that otherwise typically occurs in this disease (Vining had a history of seizures, which became chronic and
et al., 1997; Marras et al., 2010). In the largest published recurrent in 43% (Mikdashi et al., 2005). As in epilepsy
series of hemispherectomies in children, 89% of patients from other causes, other neuropsychiatric symptoms are
with Rasmussen’s encephalitis experienced an excellent more prevalent in SLE patients with seizures than
seizure outcome, being either seizure-free or reduced to without.
224 J. BRITTON
DIAGNOSTIC EVALUATION (Devinsky et al., 1988). Vasculitis is not a typical patho-
logic finding in NPSLE, and should not be assumed to be
Clinical
present in SLE patients presenting with seizures. Other
There is no characteristic seizure type or presentation potential causes of seizures in SLE include medication
specific to NPSLE. Patients with seizures who have an side effects, electrolyte disturbances related to renal fail-
existing diagnosis of SLE should undergo evaluation ure, and opportunistic infections.
to exclude non-SLE causes. However, seizures may be Primary autoimmune mechanisms are also thought to
the presenting symptom in SLE. play a role in seizures associated with SLE. Moderate to
high titers of anticardiolipin and anti-Smith antibodies,
Serology and high titers of antiribosomal P protein antibodies in
CSF have been correlated with an increased risk of sei-
Antinuclear antibody (ANA) testing is highly sensitive zures in SLE (Liou et al., 1996; Mikdashi et al., 2005;
for the diagnosis of SLE, but not specific. In a study Yoshio et al., 2005). The prevalence of antineuronal anti-
of normal subjects, the prevalence of ANA positivity bodies has also been identified in SLE patients, and
ranged from 31.7% of individuals when a titer criterion found more commonly in those with NPSLE than in
of 1:40 was used and 3.3% at a titer of 1:320 (Tan et al., those without CNS manifestations (Kang et al., 2008).
1997). Anti-ds-DNA antibody testing, by contrast, is Antibodies binding to peptides of the anti-NMDA recep-
highly specific for the diagnosis of SLE, but has a limited tor subunit NR2- were identified in CSF and serum
sensitivity of 70%. Anticardiolipin and anti-Smith anti- (DeGiorgio et al., 2001) of patients with SLE, which cor-
bodies are characteristically present in NPSLE. related with the presence of NPSLE symptoms (Arinuma
et al., 2008). These antibodies are reported to interact
Cerebrospinal fluid in vitro with NMDA receptors, leading to calcium
influx, suggesting antibody-mediated neuroexcitation
The CSF may show an inflammatory profile, with mildly
as a possible seizure mechanism in NPSLE (Gono
elevated mononuclear cell concentration and CSF
et al., 2011). In addition, anti-intermediate neurofilament
protein. However it may be normal.
alpha-internexin antibodies have been identified in 50%
of patients with NPSLE symptoms in the CSF and serum.
Differential diagnosis Administration of this antibody to murine models pro-
The differential diagnosis of seizures presenting in a duced an encephalopathy, suggesting they could play a
patient with SLE is broad. For patients treated with pathogenic role in NPSLE (Lu et al., 2010). These data
chronic immunosuppressive therapy, opportunistic infec- should be interpreted with caution, as the pathophysio-
tious and neoplastic diseases of the CNS need to be con- logic relevance of such antibodies remains unproven.
sidered. For patients with SLE nephritis, electrolyte Furthermore, discovery of clinically pertinent antibodies
disturbances should be excluded. Opiates, tramadol, in sera of lupus patients is confounded by the presence
selective serotonin reuptake inhibitor and serotonin and of “sticky” immunoglobulins/immune complexes that
norepinephrine reuptake inhibitor medications, com- result in a high frequency of false positivity in immuno-
monly used for pain management and mood disorders assays, if adequate controls for patient-specific sticki-
in SLE and other chronic illnesses, all lower the seizure ness are not included.
threshold. Thromboembolism and cerebral venous throm-
bosis secondary to SLE-associated valvular abnormalities
and presence of lupus anticoagulant need to be consid- THERAPEUTIC APPROACH
ered as well. Other secondary causes of seizures unrelated
to SLE also need to be considered, such as neoplasms, vas- The management of NPSLE varies depending on sever-
cular malformations, other structural abnormalities, ity. Hydroxychloroquine may be tried in milder cases.
trauma, and genetic etiologies. Corticosteroids are commonly used in more severe
cases (Muscal and Brey, 2010). High-dose (2 g/kg)
IVIG therapy has been proposed as an option in treat-
PATHOPHYSIOLOGY
ing several manifestations of SLE, including NPSLE
The causes of seizures in patients with SLE are many. (Zandman-Goddard et al., 2009). Finally, nonmyeloa-
Seizures may result from cerebral infarction due to blative hematopoietic stem cell transplant following
Libman–Sacks endocarditis and thromboembolism due administration of high-dose cyclophosphamide has
to presence of lupus anticoagulant. In fatal cases of been used in severe SLE cases, including those with
NPSLE with seizures, autopsy has been reported to show NPSLE, with a reported 50% disease-free survival rate
evidence of thrombotic thrombocytopenic purpura (Burt et al., 2006).
AUTOIMMUNE EPILEPSY 225
Other autoimmune connective tissue diseases headache, depression, entrapment syndromes, periph-
eral neuropathy, and epilepsy (Briani et al., 2008). Ataxia
While Sj€ogren’s may present with predominantly sali-
is a characteristic nonepileptic neurologic manifestation.
vary and lacrimal gland dysfunction, it may also present
Seizures have been reported in 2–6% of patients with
with more systemic manifestations, including those
celiac disease (Tengah et al., 2004; Bürk et al., 2009).
involving the CNS in which seizures may result. Seizures
A history of exacerbation of neurologic symptoms
are a prominent feature in an unusual form of sclero-
concurrent with dietary exposure to gluten-containing
derma referred to as linear scleroderma (also referred
foods can sometimes be elicited. A unique syndrome
to as Perry–Romberg syndrome, morphea, and “coup
referred to as CEC (celiac disease, epilepsy, and cerebral
de sabre”) (Amaral et al., 2012). This disorder is clinically
calcifications), has been recognized for several years
manifested by a localized sclerodermal lesion affecting
(Bouquet et al., 1992; Fois et al., 1994). In this syndrome,
the face as well as the brain, localized to the region
CT imaging shows calcifications in the parietal-occipital
underlying the facial lesion. The seizures in linear sclero-
cortical regions (Bouquet et al., 1992; Cury and Hobi
derma are typically refractory to antiepileptic medica-
Moreira, 2014). Such patients may have partial seizures
tion, and unfortunately usually do not improve with
manifested by transient episodes of blindness correlating
systemic immunotherapy.
with the occipital calcifications. This syndrome accounts
Seizures do not typically complicate rheumatoid
for only a proportion of celiac patients with seizures.
arthritis or the inflammatory myopathies. Primary
There is little information as to the long-term progno-
CNS vasculitis can present with seizures in 16% of cases
sis of seizures occurring in the setting of celiac disease.
(Salvarani et al., 2007). Opportunistic infections or neo-
In general, treatment success hinges on adherence to the
plastic conditions are also a concern in patients with any
gluten-free diet.
autoimmune disorder requiring chronic immunosup-
pressive therapy and need to be considered in the event
DIAGNOSTIC EVALUATION
of a seizure.
Serologic testing
Celiac disease Testing for celiac disease typically begins with testing for
Celiac disease is an autoimmune disorder of the gastro- antitissue tranasglutaminase (anti-TTG) IgA antibodies,
intestinal tract due to hypersensitivity to gliadin, a gluten which is 94% sensitive and 97% specific for celiac disease
protein in wheat, rye, and barley. Celiac disease has (Fasano and Catassi, 2012). IgG anti-TTG testing is advis-
received increasing public awareness in recent years. able in IgA-deficient patients. Antiendomysial IgA anti-
A number of neurologic presentations have been associ- bodies are highly specific (>98%), but are expensive
ated with celiac disease, including seizures (Chapman and operator-dependent. IgA antigliadin antibody testing
et al., 1978), although the nature of any causal relation- has fallen out of favor due to a 15–20% false-positive
ship between these diseases and celiac disease is still rate. However, testing for IgG antibodies targeting
not clear. deaminated gliadin peptides has >90% sensitivity and
specificity and is useful in IgA-deficient patients. The
sensitivity of antibody testing in celiac disease is signifi-
EPIDEMIOLOGY AND GENETICS cantly reduced in patients on the gluten-free diet. In such
In a population-based study, patients with celiac disease cases, testing for the presence of HLA-DQ2 and HLA-
were determined to be at 1.42 higher risk of epilepsy, sug- DQ8 alleles is advised, as their absence makes the diag-
gesting a modest association (Ludvigsson et al., 2012). nosis highly unlikely.
Celiac disease characteristically occurs in patients carry-
ing alleles for HLA-DQ2 and/or HLA-DQ8. Absence Small-bowel biopsy
of these alleles confers a protective effect against the Endoscopy with small intestinal villous biopsy is the
development of celiac disease. definitive diagnostic procedure in celiac disease. The char-
acteristic pathologic findings in celiac disease include
CLINICAL PRESENTATION, COURSE, AND PROGNOSIS lymphocytic infiltration of the endothelium, crypt hyper-
plasia, and villous atrophy.
The nonneurologic symptoms characteristic of celiac dis-
ease include diarrhea, weight loss, bloating, abdominal
Neuroimaging
distension, iron deficiency, fatigue, aphthous stomatitis,
dermatitis herpetiformis, and osteopenia. In a clinical In celiac patients with seizures, CT or MRI should be con-
series of 71 patients with celiac disease, a variety of neu- sidered. The presence of parietal-occipital calcifications
rologic symptoms occurred in 22.5%, including in such cases would suggest the CEC syndrome.
226 J. BRITTON
Similarly, celiac disease testing is recommended in et al., 2011). Patients characteristically have high titers of
patients with seizures and occipital calcifications antithyroperoxidase (anti-TPO) antibodies and typically
(Bouquet et al., 1992). respond to corticosteroid therapy. These features have
led to use of the acronym SREAT (steroid-responsive
Cancer investigation encephalopathy associated with autoimmune thyroiditis)
in place of Hashimoto’s encephalopathy (Mahad et al.,
Celiac disease does not usually occur as a paraneoplastic 2005; Castillo et al., 2006). Another term used for this
syndrome. An evaluation for cancer is not warranted in syndrome is the less-specific acronym NAIM (nonvascu-
typical cases. litic autoimmune inflammatory meningoencephalitis),
which was coined based on the absence of frank vascu-
PATHOPHYSIOLOGY litis seen on brain biopsy in affected cases (Caselli
The neurologic manifestations of celiac disease are et al., 1999).
thought most likely to be immune in origin. Antigliadin
antibodies, which are ubiquitously present in celiac dis- EPIDEMIOLOGY
ease, have been shown to cross-react with a neural anti-
Awareness of Hashimoto’s encephalopathy has
gen, synapsin 1 (Alaedini et al., 2007). This suggests a
increased over the last several years but it is still rela-
potential pathogenic link between celiac disease and its
tively uncommon, with an estimated prevalence of
neurologic manifestations. However, other investigators
2.1/100 000 (Ferracci et al., 2003). It is significantly more
have found no clear association between the presence of
common in women. The wide availability of antiTPO
antineural antibodies and neurologic symptoms in celiac
antibody testing has made identification of potential
patients (Briani et al., 2008). The etiology of the cerebral
cases readily accessible, likely leading to an increase in
calcifications in the CEC syndrome is unknown, but
recognition of this entity.
thought to be related to folate or vitamin D deficiency
resulting from malabsorption (Bouquet et al., 1992).
CLINICAL PRESENTATION, COURSE, AND PROGNOSIS
THERAPEUTIC APPROACH Seizures are present in up to 66% of cases (Chong et al.,
Treatment of the CNS manifestations of celiac disease 2003; Castillo et al., 2006). Other clinical presentations
primarily involves institution of a gluten-free diet. If include chronic cognitive decline, dementia, depression,
the diet is adhered to, anticonvulsant therapy alone is irritability, psychosis, ataxia, myoclonus, tremors, and
usually ineffective. A role for immunosuppressant ther- chorea. Hashimoto’s encephalopathy may also present
apy has not been established. Folate replacement therapy as an acute disseminated encephalomyelitis-like illness,
should be considered to help prevent neurologic compli- transverse myelitis, a syndrome mimicking Creutzfeldt–
cations in celiac disease. Iron and vitamin deficiency Jakob disease, and coma. Epilepsy presentations include
may occur as well in celiac disease and replacement ther- progressive myoclonic epilepsy of childhood, a syndrome
apy should be considered. identical to limbic encephalitis, and new-onset status epi-
lepticus (Arya et al., 2013; Chaigne et al., 2013). Despite
SEIZURES DUE TO ANTIBODY- the presence of antithyroid antibodies, patients with
MEDIATED NEUROLOGIC AUTOIMMUNE Hashimoto’s encephalopathy are typically euthyroid.
DISORDERS The encephalopathy and seizures in Hashimoto’s
encephalopathy are amenable to immunotherapy. The
Hashimoto’s encephalopathy, better termed prognosis is generally good, but depends in part on the
autoimmune encephalopathy associated with severity at presentation. Recurrences can occur, and in
thyroid antibodies some cases maintenance immunotherapy proves neces-
Hashimoto’s encephalopathy, first described by Lord sary. The prognosis in patients presenting in status
Brain in 1966, is an inflammatory CNS disorder charac- epilepticus and limbic encephalitis is more guarded,
terized by waxing and waning mental status changes, but good responses are still possible.
seizures, and stroke-like episodes, that is associated with
thyroid autoantibodies (Brain et al., 1966). It has been DIAGNOSTIC EVALUATION
increasingly recognized over the last several years as
Serology
an important treatable cause of encephalopathy,
although the lack of a clear definition of the syndrome, The serologic hallmark of Hashimoto’s encephalopathy
and overlap with the newer neuronal antibody-associated is the presence of anti-TPO antibodies. Antithyroglobu-
syndromes, raises questions about its frequency (Tüzün lin and antithyroid microsomal antibodies are also
AUTOIMMUNE EPILEPSY 227
elevated. Patients with Hashimoto’s encephalopathy may
also have a positive ANA, potentially causing confusion
with NPSLE. CSF may show an inflammatory profile but
may not, and the lack of inflammatory CSF abnormali-
ties does not exclude the diagnosis. In one large series, an
inflammatory profile was seen in 25% (Castillo et al.,
2006). However, none of these antibodies, which are rel-
atively common in the most susceptible age groups,
should exclude the diagnosis of a more specific autoim-
mune form of encephalitis, such as those associated with
NMDA receptor or voltage-gated potassium channel
(VGKC)–complex antibodies (Tüzün et al., 2011).

Neuroimaging
MRI may be normal. In one series, MRI was normal in
74% of cases (Castillo et al., 2006). Abnormal T2 signal
hyperintensity in the hippocampi may be seen in some
cases (Song et al., 2004; Tang et al., 2012). FDG-PET
may show multifocal hypometabolism during active
encephalopathy, with resolution to normal following
treatment (Seo et al., 2003).

EEG Fig. 13.3. Electroencephalograms (EEGs) from patient with


autoimmune encephalopathy associated with thyroid anti-
The EEG usually shows moderately severe abnormalities bodies. (A) EEG prior to treatment shows generalized triphasic
during the encephalopathy, which may improve signifi- waves and anterior predominant delta-frequency slowing. (B)
cantly in parallel with clinical response following treat- EEG performed 2 years posttreatment is normal. (Reproduced
ment (Fig. 13.3). The EEG abnormalities seen in from Schauble et al., 2003, with permission.)
Hashimoto’s encephalopathy include interictal epilepti-
form discharges, seizures, and generalized triphasic reductase-I antigens have also been identified, and are
waves. None of these findings is specific to Hashimoto’s located in endothelial and neuronal tissue, potentially
and can be seen in encephalopathies resulting from other accounting for the neurologic features of this condition
causes. (Beatrice et al., 2008). The pathologic relevance of the
immune response to these targets remains to be
Differential diagnosis determined.
The differential diagnosis of Hashimoto’s encephalopa- Few autopsies of Hashimoto’s encephalopathy have
thy is broad due to its protean manifestations, and been reported given most respond to therapy and are
includes encephalopathies due to diffuse toxic- not acutely fatal. Perivenular inflammatory changes
metabolic, infectious, vascular, and other inflammatory have been described in the few autopsy and biopsy cases
etiologies. Hashimoto’s encephalopathy should also be in the literature, but frank vasculitis is typically not pre-
on the differential diagnosis of patients presenting with sent (Caselli et al., 1999; Striano et al., 2006).
unexplained subacute dementia, de novo status epilepti-
cus, and limbic encephalitis. THERAPEUTIC APPROACH
One of the striking features of this presumed autoim-
PATHOPHYSIOLOGY
mune encephalopathy is the marked and relatively
Anti-TPO antibodies are considered markers of autoim- quick response to steroids (Olmez et al., 2013). Cortico-
munity rather than playing a direct role in the pathophys- steroid therapy dosage strategies have not been formal-
iology of Hashimoto’s encephalopathy. Antibodies to ized, but typically consist of 1 gram methylprednisolone
antigens expressed in neural tissue have been identified IV daily for 3–5 days, followed by weekly administra-
in Hashimoto’s encephalopathy, including antibodies tion. Alternatively, 60 mg oral prednisone daily has
targeting the amino-terminal of alpha-enolase (Fujii been advocated, with a gradual reduction afterwards
et al., 2005; Matsunaga et al., 2013). Serum and CSF if allowed by the clinical course. IVIG has been
antibodies targeting dimethylargininase-I and aldehyde used for those who do not tolerate corticosteroid
228 J. BRITTON
treatment. Plasmapheresis has also been reported to be CLINICAL PRESENTATION, COURSE, AND PROGNOSIS
efficacious in refractory cases (Bektas et al., 2012; Pari
The seizures seen in association with GAD-65 antibody
et al., 2014). Hashimoto’s encephalopathy may relapse
consist of simple partial, complex partial, and secondary
over time, prompting some authorities to recommend
generalized tonic-clonic seizures. Affected patients may
chronic maintenance therapy in some cases. No supe-
also present with the limbic encephalitis syndrome and
rior maintenance treatment has been identified. Ritux-
nonconvulsive status epilepticus. It should be noted that,
imab, IVIG, azathioprine, and mycophenolate have
while seizures clearly occur in the setting of GAD-65
all been reportedly used for this purpose (Olmez
positivity, they are present in the minority of cases. In
et al., 2013).
one study evaluating the neurologic findings in patients
with high GAD-65 antibody titers, seizures or limbic
encephalopathy were collectively present in only 10%
Glutamic acid decarboxylase 65 kD isoform
(6 of 61) (Saiz et al., 2008).
(GAD-65) antibody-associated encephalitis
The clinical course of epilepsy and seizures associ-
and seizures
ated with GAD-65 antibodies is variable, and the progno-
GAD-65 antibody is classically associated with stiff-man sis is guarded. While some patients will respond to
syndrome (McKeon et al., 2012). However, over time it immunotherapy, many do not, in which case seizures
has become apparent that GAD-65 antibodies are associ- remain disabling (Toledano et al., 2014).
ated with a variety of neurologic features, including
ataxia, vertigo, brainstem dysfunction, limbic encepha- DIAGNOSTIC EVALUATION
lopathy, extrapyramidal signs, myelopathy, and seizures Clinical
(Pittock et al., 2006; Saiz et al., 2008). Seizures and epi-
lepsy may be a prominent neurologic feature in affected Patients with seizures associated with GAD-65 antibodies
patients. typically present with complex partial and secondary
generalized tonic-clonic seizures. The semiology of the
seizures occurring in the context of this antibody has
EPIDEMIOLOGY no distinguishing feature. Certain other clinical features,
however, suggest the possibility of GAD-65 antibody
GAD-65 antibodies are prevalent in diabetes mellitus positivity, such as the presence of diabetes type 1 and con-
type 1, occurring in over 70% of patients (Petersen current autoimmune thyroid disease. Diabetes type 1, for
et al., 1994). It is important to note that GAD-65 anti- example, is present in over 50% of patients with GAD-65-
bodies are also present in the normal population. In associated neurologic syndromes, and concurrent thy-
one study comparing the prevalence of GAD-65 in the roid disease and/or antithyroid antibody positivity are
type 1 diabetes and nondiabetes populations, GAD-65 present in 30–40% (Pittock et al., 2006; McKeon et al.,
antibody was found in 4% of normal subjects. 2012). Finally, vitiligo is present in 15% of patients with
While GAD-65 antibodies can occur in association GAD-65-associated neurologic syndromes, and if pre-
with an acute or subacute limbic encephalitis syndrome sent should suggest the diagnosis (Pittock et al., 2006).
(Malter et al., 2010), they may also be present in patients
with chronic epilepsy. In a case-control study comparing
Serology and CSF
253 patients from an epilepsy clinic with 250 nonepilepsy
controls, GAD-65 positivity was found in 15 (5.9%) of As indicated previously, antibodies targeting GAD-65 are
epilepsy patients and 3 (1.5%) controls ( p ¼ 0.026) present in approximately 4% of the normal population
(Liimatainen et al., 2010). High titers (>1000 relative (>0.02 nmol/L or >0.5 U/mL), and in most patients with
units/mL) were found in 7 patients with epilepsy and type 1 diabetes, so cannot be considered specific for the
none of the controls. In another study in which serum presence of an autoimmune etiology in a patient with epi-
from two large epilepsy cohorts was screened for neuro- lepsy. However, the antibody titers seen in autoimmune-
pil antibodies, a lower prevalence of GAD-65 antibodies mediated neurologic disorders, including seizures, are
was observed (1.7%) (Brenner et al., 2013). The majority typically much higher, often 100-fold higher, than those
of GAD-65-positive epilepsy patients have temporal- seen in the general and type 1 diabetes populations. Other
lobe seizures. In a study comparing 51 patients with antibodies are commonly encountered. For example, in
partial epilepsy, 49 patients with generalized epilepsy, one study, 50% of GAD-65 patients had antithyroid anti-
38 patients with other neurologic conditions, and 48 nor- bodies, and many other antibodies were present, including
mal controls, GAD-65 positivity was found in 8 (16%) calcium channel, VGKC–complex, striated muscle,
partial epilepsy patients and in none of the other groups amphiphysin, parietal cell, and islet cell antibodies in a
(Peltola et al., 2000). smaller proportion (Pittock et al., 2006).
AUTOIMMUNE EPILEPSY 229
Neuroimaging etiologies of temporal-lobe seizures should still be
considered, such as structural lesions of the temporal
MRI may show mesial temporal T2 signal hyperintensi- lobe and others. For patients presenting with limbic
ties with or without gadolinium enhancement (Fig. 13.4). encephalitis and nonconvulsive status epilepticus, other
These findings are not unique to GAD-65 antibody, and etiologies of these syndromes need to be considered.
may be seen in limbic encephalitis from other causes. Hashimoto’s encephalopathy presents a particular prob-
Indeed, there is a GAD antibody-associated form of lim- lem in assigning a diagnosis, as antithyroid antibodies,
bic encephalitis (Malter et al., 2010). Over time, atrophy which are a key diagnostic feature in Hashimoto’s
of the hippocampal structures may develop. Imaging in encephalopathy, may also be present in GAD-65
patients lacking the full features of limbic encephalopa- patients. In such cases, high titers of GAD-65 would typ-
thy may be normal. ically suggest a GAD-65-associated syndrome. This dis-
tinction may be relevant for counseling purposes, as the
EEG
response to immunotherapy is generally regarded as
The EEG in GAD-65 patients with epilepsy may show being more satisfactory in Hashimoto’s encephalopathy
epileptiform abnormalities involving the frontotemporal than in GAD-65 antibody-associated encephalitis.
regions (Fig. 13.5). There are no unique EEG features
seen in association with this antibody.
PATHOPHYSIOLOGY
Cancer investigation
It has not been established whether GAD-65 antibodies
GAD-65 antibodies are not generally considered a sign play a pathogenic role or are mainly present as a marker
of paraneoplastic disease. In one series, underlying neo- of autoimmune-mediated neuronal injury. GAD plays a
plasms were found in 7% (4 of 61) of patients, three of critical role in the glutamate-gamma-aminobutyric acid
which were neuroendocrine tumors (Saiz et al., 2008). A (GABA) cycle, catalyzing the conversion of glutamic
acid to GABA, the major inhibitory neurotransmitter in
Differential diagnosis
the brain. GAD exists as two major isoforms, 67 kD
For patients presenting chiefly with temporal-lobe (GAD-67) and 65 kD (GAD-65). The GAD-65 isoform
seizures and a low-titer anti-GAD-65 antibody, other is transiently activated in response to demands for

Fig. 13.4. Imaging from a 62-year-old woman with limbic encephalitis and frequent subclinical frontotemporal seizures associ-
ated with glutamic acid decarboxylase 65 kD isoform (GAD-65) antibody. (A) Coronal fluid-attenuated inversion recovery
(FLAIR)-weighted imaging shows hyperintensity involving the bilateral orbitofrontal (bracket), right hypothalamic (arrow),
and bilateral hippocampal regions (star). (B) 18-Fluorodeoxyglucose positron emission tomography (FDG-PET) showing hyper-
metabolic abnormalities in the bilateral hippocampal regions (arrows) and striatum (star). Used with permission of Mayo Foun-
dation for Medical Education and Research. All rights reserved.
230 J. BRITTON

Fig. 13.5. Electroencephalogram in patient with glutamic acid decarboxylase 65 kD isoform (GAD-65)-associated limbic enceph-
alitis, showing a right frontotemporal seizure manifested as a rhythmic discharge arising first in the F8 derivation (arrow). Used
with permission of Mayo Foundation for Medical Education and Research. All rights reserved.

temporary boosts in GABAergic neurotransmission diabetes due to the effects of the former on serum glucose
(Fenalti et al., 2007). It is therefore conceivable that an concentrations. In such cases, IVIG may be a consideration.
antibody directed against GAD-65 could disrupt the The responder rates to corticosteroid and IVIG ther-
glutamate-GABA balance to one favoring an accumula- apy are generally higher for seizures related to other neu-
tion of glutamate and reduction in GABA, leading to ral antibody-mediated syndromes, as illustrated by the
increased neuronal excitation and seizures. In the poor responses of patients with GAD antibodies and lim-
stiff-person syndrome this is thought to take place bic encephalitis (Malter et al., 2010). The reasons why sei-
mainly in the spinal cord and brainstem (see zures in association with this antibody do not respond are
Chapter 17). There is evidence that GAD-65 antibodies not completely clear. One theory holds that GAD-65 anti-
may affect GABA activity. In a study of 4 patients with bodies may arise after irreparable neuronal injury or
epilepsy and high serum GAD antibody levels, cortical apoptosis has already occurred, which exposes intracel-
GABA levels as measured on MR spectroscopy were lular enzymes like GAD-65 to circulating self-
reduced compared to 10 controls (Stagg et al., 2010). antibodies, in which case immunotherapy is too late. In
In another study, application of GAD-65 sera to a net- contrast, antibodies targeting membrane surface anti-
work of hippocampal neurons in culture led to suppres- gens may be more prone to causing functional changes
sion of recorded inhibitory postsynaptic potentials in neurons due to interaction at receptor targets, as
(Vianello et al., 2008). These findings suggest that opposed to causing cellular destruction, thereby present-
GAD-65 antibodies may lead to a reduction in ing at a stage where neuronal salvage is still possible.
GABAergic-mediated inhibition, which could hypotheti- In patients unresponsive to corticosteroid or IVIG
cally lead to increased seizure propensity, but does not therapy, there is little guidance from the literature. Plas-
address the question of how antibodies to an intracellular mapheresis, rituximab, mycophenolate, and azathio-
protein cause the disease. prine are options, but the efficacy of these therapies in
this setting has not been established.
THERAPEUTIC APPROACH
A trial of immunotherapy is justified in GAD-65-positive
Voltage-gated potassium channel–complex
patients with medically unresponsive seizures; however
antibody-associated encephalitis
the results are often disappointing, unfortunately. In our
autoimmune epilepsy cohort, only 2 of 6 GAD-65 VGKC–complex antibodies are an important cause of
antibody-positive patients improved with immunotherapy limbic encephalitis and temporal-lobe seizures
(Fig. 13.6) (Toledano et al., 2014). Corticosteroid therapy (Buckley et al., 2001; Pozo-Rosich et al., 2003; Thieben
needs to be used with caution in GAD-65 patients with et al., 2004; Irani et al., 2010). These antibodies are
AUTOIMMUNE EPILEPSY 231
Immunotherapy Response: Flow Chart Response to Immunotherapy According to Antibody Type
Patients with suspected autoimmune epilepsy
n = 29
PMA Ab positive PMA Ab negative
n = 16 n = 13

Abs to plasma
Onco-neural Abs
membranes gAChR Ab CC P/Q; CC N GAD 65 Abs No neuronal Abs
n=1
n = 12 n=3 n=1 n=6 n=6
Ma1/Ma2
VGKC-complex

Responders Responders Nonresponders Nonresponders Responders Nonresponders Responders Nonresponders


Nonresponders
n = 12 n=2 n=1 n=1 n=2 n=4 n=2 n=4

100
Responders

Responders (%)
n = 13 80
IVMP Responders
60
n = 23 n=2
Nonresponders IVIg
40
n = 10 n=5
Nonresponders 20
n=3
0
VGKC gAChr GAD65 Ma1/Ma2 No neural
abs
C PMA Abs Ab to intracellular
proteins

60
Responders Responders (%)
50
n=2
IVIg Responders 40
n=6 n=1 30
Nonresponders IVMP
n=4 n=2 20
Nonresponders
10
B
n=1
0
IVMP as IVIg as IVMP as IVIg as
D 1st agent 1st agent 2nd agent 2nd agent

Fig. 13.6. Immunotherapy trial response in autoimmune epilepsy. (A) Flow chart of response to immunotherapy trial according to
antibody type. (B) Flow chart of response to immunotherapy trial according to immunotherapeutic agent tried. (C) Percent of
responders by antibody type. (D) Percent of responders to first and second immunotherapeutic agent tried. Abs, antibodies;
CC N and CC P/Q, voltage-gated calcium channel N-type and P/Q-type; gAChR, neuronal ganglionic nicotinic acetylcholine
receptor antibody; GAD65, glutamic acid decarboxylase 65; IVIg, IV immune globulin; IVMP, IV methylprednisolone; PMA
Ab, plasma membrane antigen antibodies; VGKC, voltage-gated potassium channel. (Reproduced from Toledano et al., 2014,
with permission.)

associated with a number of neurologic presentations neuronal surface antigen antibodies in 45 patients with
other than seizures, including Morvan’s syndrome, neu- limbic encephalitis, VGKC–complex antibodies were
romytonia, gastrointestinal dysmotility, and a chronic found in 13 (29%) (Graus et al., 2008).
pain syndrome thought to be mediated by hyperexcitabil-
ity of nociceptive pathways (Klein et al., 2012, 2013).
CLINICAL PRESENTATION, COURSE, AND PROGNOSIS
Patients with seizures related to VGKC–complex anti-
EPIDEMIOLOGY
bodies may experience a number of seizure semiologies.
The prevalence of VGKC–complex antibodies in the epi- Early descriptions described hypersalivation as a charac-
lepsy and limbic encephalitis population is not definitively teristic feature of seizures in these patients (Buckley
known, as discovery of this antibody and its association et al., 2001). Since these earliest case reports, other seizure
with seizures is relatively recent. In a study of serum from semiologies have been associated with VGKC–complex
106 women with epilepsy, VGKC–complex antibodies antibody.
were found in 6 (7%) (Majoie et al., 2006). In another study The most characteristic seizure type seen in VGKC–
screening for neuropil antibodies in serum from two large complex antibody encephalitis are called faciobrachial
epilepsy center cohorts, VGKC–complex antibodies were dystonic seizures, which are manifested by the sudden
found in 5% (Brenner et al., 2013). The prevalence of onset of flexion contraction of one upper extremity or
VGKC–complex antibodies in the limbic encephalitis pop- the other, accompanied by contraction of the face and
ulation is higher. In a study evaluating the prevalence of head and neck ipsilateral to the affected upper extremity
232 J. BRITTON
(Irani et al., 2011). These seizures typically last a second recovery of memory deficits occurring in association
or so, and occur multiple times per day. The trunk, lower with VGKC–complex encephalitis is unfortunately less
extremity, and abdomen may be involved, in addition to dependable compared to the prognosis for seizures.
the face and upper extremity, and they are sometimes
accompanied by a brief vocalization. This seizure type DIAGNOSTIC EVALUATION
is present in approximately a fourth of VGKC–complex
antibody epilepsy patients (Toledano et al., 2014). Clinical
Patients with VGKC–complex antibodies may also VGKC–complex antibody-associated epilepsy should
experience simple partial autonomic seizures manifested be considered in patients with a history of new onset
by unilateral piloerection or palpitations. Others describe of seizures, confusion, and memory deficits. The sei-
their seizures as being manifested by a brief ascending zures are often brief and occur at high frequency.
or descending “wave” passing through their body. The The presence of faciobrachial dystonic seizures and
above-described seizures typically last a second or so other focal seizures occurring at a frequency of several
and occur multiple times a day in affected patients, similar per day is also highly suggestive.
to faciobrachial dystonic seizures. These may be accompa-
nied by brief periods of amnesia. Patients with VGKC–
Serology and CSF
complex antibodies may also present with complex partial
seizures and in complex partial status epilepticus. VGKC–complex antibodies were originally thought to tar-
The other neurologic presentations of VGKC– get antigens residing directly on the voltage-gated potas-
complex antibodies are further reviewed in Chapter 10. sium channel subunits. However, it was subsequently
Hyponatremia is a relatively common laboratory finding discovered that these antibodies actually target proteins
and can provide a helpful clue to the antibody. that remain complexed with potassium channels in the rab-
The prognosis of VGKC–complex antibody-mediated bit brain extracts used in the assay, as opposed to the potas-
seizures is relatively favorable. Unlike NMDA encephali- sium channel itself (Irani et al., 2010). Three other protein
tis, patients with VGKC–complex-associated seizure dis- targets were identified as the actual targets: (1) leucine-rich
orders are usually manageable in the outpatient setting. glioma inactivated 1 protein (LGI1), which is extensively
The seizures in the setting of VGKC–complex encephali- represented in the hippocampus comprising the majority
tis are typically highly responsive to immunotherapy. of the samples; (2) contactin-associated protein-2
Some patients demonstrate hippocampal edema at onset. (CASPR-2), which is localized primarily in the juxtanodal
Atrophy may develop subsequently, in which case deficits region of myelinated axons; and (3) Tag1/contactin-2, a
in short-term memory may become evident (Fig. 13.7). The protein associated with CASPR-2 (Irani et al., 2010). In

Fig. 13.7. Serial head magnetic resonance imaging scans in a patient with voltage-gated potassium channel–complex antibody
encephalitis. Comparison of the hippocampi over an 8-month period (arrows) shows the development of significant atrophy. Used
with permission of Mayo Foundation for Medical Education and Research. All rights reserved.
AUTOIMMUNE EPILEPSY 233
18 of 96 cases, the precise antigenic target within the Cancer investigation
VGKC–complex was undefined.
CSF examination in VGKC–complex antibody The identification of VGKC–complex antibodies alone
patients showed mild elevations in mononuclear cell does not strongly suggest the presence of an underlying
count in 41% and mild elevations in CSF protein in neoplasm. In a large series of 96 VGKC–complex-
47% (Jarius et al., 2008). In this series, there was no evi- positive patients comprising epilepsy and nonepilepsy
dence of intrathecal immunoglobulin production. clinical presentations, 6 were diagnosed with an underly-
ing tumor (5 thymoma, 1 endometrial carcinoma) (Irani
Neuroimaging et al., 2010). This rate is far lower, for example, than the
rate of cancer seen in association with paraneoplastic
MRI may show swelling and hyperintensity in the hippo- antibodies such as anti-Hu (ANNA-1) or anti-Ma1 or
campi on fluid-attenuated inversion recovery (FLAIR) Ma2. Indeed, when antibodies targeting intraneuronal
and T2-weighted sequences. These findings were present targets are present in combination with VGKC–complex
in 33/42 (78.6%) of cases in one series (Kotsenas et al., antibodies, the potential for an underlying cancer is
2014). Gadolinium enhancement and restricted diffusion higher (Graus and Saiz, 2008). In the latter situation, a
may be present in addition in the early phases. Serial cancer evaluation is clearly warranted. However, the
imaging often shows resolution of hippocampal edema, value of pursuing an extensive cancer evaluation in
and may show progression to hippocampal atrophy over the setting of isolated VGKC–complex antibody positiv-
time (Fig. 13.7). ity is arguable.
A unique MRI finding described in the setting of
VGKC–complex antibody positivity is FLAIR hyperin- Differential diagnosis
tensity abnormalities involving the striatum (Hiraga
et al., 2006; Plantone et al., 2013; Kotsenas et al., For patients presenting with the syndrome of limbic
2014). FDG-PET hypermetabolism of the striatum has encephalitis, the differential diagnosis is extensive and
also been described in association with VGKC–complex includes consideration of other autoimmune causes dis-
antibody (Rey et al., 2012). cussed in this chapter. In the acute setting, infectious lim-
FDG-PET may show hypermetabolic abnormalities in bic encephalitis due to human herpes viruses 1 and 6 needs
the hippocampal regions in VGKC–complex antibody to be considered. Other diseases affecting the limbic sys-
encephalitis (Fauser et al., 2005; Kotsenas et al., 2014). tem also need to be excluded, including vascular diseases,
This may reflect the high seizure frequency sometimes neoplasms, or other structural lesions. In patients pre-
seen in this population. However, it may also reflect senting with an acute amnestic syndrome in the absence
excess glucose utilization secondary to inflammation of seizures, Wernicke’s encephalopathy would be
in these regions. These FDG-PET findings are not unique included in the differential diagnosis. Affective symp-
to VGKC–complex and can be seen in limbic encephalitis toms and anxiety are common in these patients, and this
from other etiologies (Fig. 13.4). can cause misdiagnosis of a primary psychiatric disorder.

EEG PATHOPHYSIOLOGY
The EEG typically shows interictal abnormalities local- LGI1 is highly represented in the hippocampi, and is the
ized in the temporal regions. In patients with a high sei- predominant VGKC–complex antibody type in patients
zure frequency, electrographic temporal-lobe seizure presenting with limbic encephalitis and seizures. LGI1
discharges may also be present. Such findings are not is weakly expressed in the peripheral nervous system
specific for VGKC–complex and are common to limbic and can be present in patients with peripheral nervous
encephalitis of all causes (Fig. 13.3). Importantly, the system disease (Irani et al., 2010; Klein et al., 2013).
faciobrachial seizures in VGKC–complex antibody CASPR-2 is primarily localized to the juxtanodal region
encephalitis are often brief and not associated with loss in myelinated nerve fibers and most often presents with
of awareness, suggesting that a small cortical regional neuromyotonia and other peripheral nervous system
area may be involved during the seizure, which may ren- manifestations, although it is also expressed widely in
der the seizure undetectable on scalp EEG. This can lead the brain, and CASPR-2 antibodies have been identified
to a misdiagnosis that the recorded events are functional in patients with CNS presentations. The mechanism by
if the EEG reader does not take this into account when which these antibodies lead to seizures is not definitively
interpreting the recording (Tao et al., 2007). The absence established. However, as potassium channels allow
of confirmatory ictal EEG findings is the rule rather than potassium efflux resulting in neural inhibition, any
the exception for faciobrachial dystonic seizures, and impairment of their function could favor neural excita-
for simple partial seizures and auras without impaired tion, which could theoretically result in seizures and
reactivity, which are not uncommon in this population. peripheral hyperexcitability phenomena such as
234 J. BRITTON
neuromyotonia. IgG from one patient with LGI1 anti- recent years, a syndrome associated with antibodies tar-
bodies caused epileptogenic effects on rodent hippocam- geting the NMDA receptor has been identified (Dalmau
pal slices (Lalic et al., 2011). et al., 2007). NMDA receptor antibody encephalopathy
Data on the neuropathology of VGKC–complex anti- has many characteristic clinical features, and is poten-
body encephalitis is limited. In one report of an autopsy in tially fatal. However, it is also highly amenable to immu-
a VGKC–complex antibody patient, the hippocampi con- nosuppressant therapy, and the chances for a good
tained activated microglia and occasional B lymphocytes, neurologic recovery are realistic in many cases.
with sparing of other limbic structures, including the cin-
gulate gyrus and insular cortex (Khan et al., 2009).
Immunoglobulin staining showed a nonspecific pattern, EPIDEMIOLOGY
and did not show specific deposits near neurons or blood
The prevalence and incidence of this disorder are not
vessels. These findings generally support a predominant
precisely known. In a study evaluating the prevalence
B-cell-mediated inflammatory mechanism in this disor-
of neural antibody positivity in a large epilepsy cohort,
der, with some complement-mediated damage (see
NMDA receptor antibodies were identified in 1.7%.
Chapter 7 for further details).
Therefore, it is an unlikely cause of epilepsy outside
of its occurrence in association with the NMDA receptor
Therapeutic approach
antibody encephalopathy syndrome.
Most patients respond favorably to corticosteroid ther-
apy. If corticosteroids are contraindicated or inadvis-
able, IVIG can be considered. There have been no CLINICAL PRESENTATION, COURSE, AND PROGNOSIS
randomized clinical trials establishing the efficacy supe-
Seizures are often present at the onset of NMDA recep-
riority of one over the other in this condition. If the
tor antibody encephalopathy, and consist of complex
patient does not respond to one first-line treatment,
partial and secondary generalized tonic-clonic seizures.
the other is generally tried next.
These can occur at a high frequency at onset, and
The responder rate to either first-line immunotherapy
patients may present in status epilepticus. However,
treatment is generally excellent in terms of seizure
nonepileptic symptoms are also very prominent at onset
response. In our series of patients treated with IV meth-
as well, and in some cases may overshadow the seizures.
ylprednisolone and/or IVIG, 100% of 10 patients with
Affected patients often have a prominent psychosis at
seizures associated with VGKC–complex antibodies
presentation, for example, with delusions and auditory
improved substantially in terms of seizure rate (Irani
and visual hallucinations (Titulaer et al., 2013a, b).
et al., 2010; Quek et al., 2012; Toledano et al., 2014).
Affected patients are also prone to respiratory failure
Within days of initiation of treatment with IV methyl-
and the need for ventilatory support is not uncommon.
prednisolone, the seizure frequency may decrease from
Dyskinesias are another characteristic feature of this
multiple daily simple partial seizures to zero or a few per
disorder, and affect the limbs, digits, head and neck,
day. In the event of failure of first-line therapy, plasma-
and face (Stamelou et al., 2012). The dyskinesias may
pheresis can be considered. Finally, rituximab was found
occur continuously, and can last for days and weeks.
to be efficacious in a series of 5 patients (Irani et al.,
Affected patients may develop catatonia in addition.
2014). Despite the good seizure response, the recovery
Patients with NMDA receptor antibody-associated
of memory functions is seldom complete, and hippo-
encephalopathy can be critically ill and often require
campal atrophy is common with LGI1 antibodies
ICU care.
(Butler et al., 2014; Malter et al., 2014).
This disorder can affect all age groups. Of impor-
In responders, a decision must then be made as to
tance, this syndrome is highly correlated with the pres-
whether to initiate chronic maintenance treatment. For
ence of ovarian teratoma. In young women, adequate
this important question, there are unfortunately no avail-
evaluation for an underlying teratoma is essential.
able data. More natural history studies need to be done in
Although patients with NMDA receptor encephalop-
order to inform treatment decisions like this in these
athy may be gravely ill, the potential for recovery follow-
disorders.
ing immunotherapy with corticosteroids, IVIG, and
rituximab is relatively high (Titulaer et al., 2013a).
NMDA receptor antibody-associated
Recovery may take weeks or even months, but the pos-
encephalopathy
sibility of a good outcome is realistic. Interestingly,
NMDA receptors are excitatory receptors that mediate spontaneous remission without immunotherapy may
several critical neurologic functions, such as long-term also occur, although the reported recovery in these cases
potentiation and synaptic plasticity. Excessive activation took several months (Iizuka et al., 2008). This topic is
of NMDA receptors, however, may lead to seizures. In covered in much more detail in Chapter 12.
AUTOIMMUNE EPILEPSY 235
DIAGNOSTIC EVALUATION Cancer investigation
Clinical NMDA receptor antibody encephalopathy is associated
The prominent psychotic symptoms, dyskinesias, and with the presence of teratomas. Given this association,
propensity for respiratory failure are relatively unique evaluation for teratoma is indicated in women with this
to this disorder, in comparison to the other autoimmune disorder. Teratomas are typically localized to the ovaries;
encephalopathies, although psychosis can complicate however, teratomas have also been identified in the medi-
limbic encephalitis of other causes. The presence of a ter- astinum in this population. It may rarely be associated with
atoma in this setting would be a surprising feature in a other tumor types (breast, lung, testicular, pancreatic, and
patient with a seizure syndrome, and would strongly sug- thymic have been reported) (Titulaer et al., 2013a).
gest an NMDA receptor antibody etiology.
Differential diagnosis
Other causes of autoimmune and paraneoplastic encepha-
Serology and CSF litis should be considered, as well as herpes simplex viral
NMDA receptor antibodies can be found in serum, but encephalitis and other infectious meningoencephalitides
are sometimes only present in the CSF. NMDA receptor during the acute setting. The presence of the antibodies
antibodies have been reported in schizophrenia and nar- in herpes simplex virus encephalitis cases suggests that,
colepsy populations as well, raising some questions as to in some cases, NMDA receptor antibodies may form as
their specificity (Tsutsui et al., 2012). It is important to a parainfectious response in this disease (Pruss et al.,
note that these antibodies may continue to be present 2012a; Armangue et al., 2014), although there is no evidence
long after resolution of the acute illness; therefore their for molecular mimicry. Other causes of encephalopathy,
mere presence does not always correlate with active dis- such as those stemming from vascular, neoplastic, and
ease (Hansen et al., 2013). A recent interesting observa- toxic-metabolic etiologies, should also be considered.
tion is the presence of NMDA receptor antibodies
arising during or after herpes simplex encephalitis, often PATHOPHYSIOLOGY
with a relapse of encephalopathy symptoms following
NMDA receptor antibodies in vitro target the GluN1 sub-
initial response to antiviral therapy (Armangue et al.,
unit, leading to a selective decrease in surface density
2014). In children the association of NMDAR antibodies
and synaptic localization in neuronal cultures secondary
with postherpes simplex virus encephalitis is particularly
to antibody-mediated capping and internalization of
striking (see Chapter 26).
NMDA receptors (Hughes et al., 2010; Moscato et al.,
2014). This results in decrease in the synaptic localization
and function of NMDA receptors, and decreased
Imaging NMDR-mediated currents, but this is reversible and
Standard structural MRI of the brain is often normal in NMDAR recovery takes place when the antibody is
NMDA receptor antibody encephalopathy. However, removed from the cell culture. In mice infused with
nonstandard MRI modalities may be more revealing. CSF positive for the antibody, there is also a loss of
In one study, functional MRI showed reduced func- NMDA receptor with prominent changes in NMDA
tional connectivity of the left and right hippocampus receptor-mediated currents (Pruss et al., 2012b). In addi-
with the anterior default-mode network. In this same tion, the application of CSF from patients with NMDA
study, diffusion tensor imaging showed evidence of receptor antibody encephalopathy to murine hippocam-
extensive white-matter changes most prominent in the pal slices leads to suppression in long-term potentiation
cingulum (Finke et al., 2013). (Zhang et al., 2012). Importantly, autoimmune neuronal
destruction does not typically occur, and this is likely the
reason underlying the potential for recovery in this ill-
EEG ness and why neuroimaging is often unremarkable in
these patients, even in the acute phase (for further
The EEG characteristically shows diffuse moderate ampli- details, see Chapters 5 and 12).
tude rhythmic delta-frequency activity (Kirkpatrick et al.,
2011). This diffuse delta pattern is sometimes associated
THERAPEUTIC APPROACH
with superimposed intermittent bursts of low-amplitude,
high-frequency activity. Given the resemblance of these In female patients, it is important to evaluate for a ter-
waveforms to delta brushes seen in premature infant atoma. A large retrospective series demonstrated that
EEG recordings, this has been referred to as the “excess immunosuppressive therapy was efficacious in NMDA
delta brush” pattern (Schmitt et al., 2012) (Fig. 13.8). receptor encephalopathy (Titulaer et al., 2013a). Of 472
236 J. BRITTON

Fig. 13.8. Electroencephalogram patterns in N-methyl-D-aspartate receptor antibody encephalopathy. (A) Synchronous, rhythmic
generalized delta in a 48-year-old comatose woman on ventilatory support. She walked out of the hospital 3 weeks later. (B) Rhyth-
mic generalized delta with delta brush-like waveforms in a 2-year-old patient with severe encephalopathy, continuous dyskinesias
of face and limbs, and respiratory depression on ventilatory support. Used with permission of Mayo Foundation for Medical Edu-
cation and Research. All rights reserved.

patients receiving first-line therapy, 251 (53%) were Other neural antibodies associated with
improved within 4 weeks, although some patients autoimmune epilepsy
required 18 months to improve. First-line immunother-
GABA-B RECEPTOR ANTIBODY
apy usually consists of steroids, IVIG, or plasma
exchange in these patients. Of 221 nonresponders, those Antibodies directed at GABA-B receptor antigens may
who received second-line therapy had a better outcome cause limbic encephalitis (Lancaster et al., 2010;
than those who did not. Second-line immunotherapy Boronat et al., 2013). GABA-B receptor antibodies may
included rituximab or cyclophosphamide. At 2 years, also present with ataxia, brainstem encephalitis, and
81% had a good outcome, but 30 died. Predictors of good opsoclonus-myoclonus syndrome (Hoftberger et al.,
outcome were early treatment and lack of need for ICU- 2013; Mundiyanapurath et al., 2013). These antibodies
level care. There was a 12% risk of relapse within 2 years accounted for 14% of cases of limbic encephalitis in
in the series, with relapses typically being milder than the one series (Boronat et al., 2013) and 10% of limbic enceph-
original attack. alitis cases previously deemed seronegative prior to
AUTOIMMUNE EPILEPSY 237
discovery of this antibody in another (Jeffery et al., 2013). hypermetabolic abnormalities were described in FDG-
EEG and MRI usually show features typical of limbic PET in 1 patient, and improved corresponding with clin-
encephalitis. There is a significant association of ical improvement (Spatola et al., 2014).
GABA-B receptor antibodies and small-cell lung carci-
noma (33–70% of cases in published series) (Lancaster GLYCINE RECEPTOR ANTIBODY
et al., 2010; Boronat et al., 2011; Hoftberger et al.,
2013), so a proper evaluation for small-cell lung cancer Glycine receptor antibodies typically present with a clinical
is indicated in these patients. Despite their association syndrome referred to as the progressive encephalomyeli-
with small-cell lung cancer, GABA-B antibodies were tis, rigidity, and myoclonus (PERM) syndrome In the larg-
only found in 1.7% of sera from patients positive for other est published series of glycine receptor antibodies, 73%
paraneoplastic antibodies predictive of small-cell lung presented with PERM (Carvajal-Gonzalez et al., 2014),
cancer in one study (Jeffery et al., 2013). Improvement but seizures were present in around 10% and status epilep-
was seen in response to immunosuppressant treatment ticus has been reported in 2 patients (Zuliani et al., 2014).
with corticosteroids or IVIG in these patients in Most treated patients responded to immunotherapy.
75–90% (Lancaster et al., 2010; Hoftberger et al., 2013).
AUTOIMMUNE EPILEPSY: DIAGNOSTIC
GABA-A RECEPTOR ANTIBODY AND THERAPEUTIC APPROACH
GABA-A receptor antibodies have been recently reported. One-third of patients with epilepsy will not respond to
In this series, high titers of antibodies were found in CSF antiepileptic drug therapy. Of these, some may be candi-
from 6 patients with status epilepticus or epilepsy partialis dates for surgery, and some may benefit from other
continua: 4 of them required pharmacologic coma (Petit- treatments, such as ketogenic diet or stimulation
Pedrol et al., 2014). Seizures were also common in a UK devices. For those who are not candidates for such treat-
study (Pettingill et al., 2015), but in both studies low titers ments, however, options are limited and the outlook can
of GABA-A receptor antibodies were found in 3% of be bleak. A primary goal in the evaluation of patients
patients presenting with a variety of neurologic syn- with intractable epilepsy is to clarify the underlying eti-
dromes, some associated with GAD-65 antibodies. ology and to treat that etiology if feasible in order to gain
control of the patient’s seizures. Identification of an
AMPA RECEPTOR ANTIBODY autoimmune etiology opens another avenue of therapy
for such patients, specifically immunotherapy, which
a-Amino-3-hydroxy-5-methyl-4-isoxazole-propionic in some cases may be highly effective.
acid (AMPA) receptor antibodies have been reported.
They usually present with limbic encephalitis. However,
When to suspect an autoimmune epilepsy
2 patients in one series presented with psychosis, similar
syndrome
to patients with NMDA receptor antibody positivity
(Graus et al., 2010). Seven of 10 reported in one series Clinical features that should suggest the possibility of an
occurred in association with a tumor; tumor types autoimmune epilepsy syndrome are summarized in
included lung, breast, and thymus (Lai et al., 2009). In Figure 13.9 and Table 13.2. Autoimmune epilepsy syn-
this report, 90% responded to immunotherapy. Similar dromes often begin in an acute or subacute manner,
to NMDA receptor antibodies, AMPA receptor anti- as opposed to gradual. Fever may be present at the onset
bodies have been determined in vitro to cause a reduc- in some cases. A history of other autoimmune conditions
tion in the number of AMPA receptor clusters on the may also be present. Patients with autoimmune epilepsy
cell surface (Lai et al., 2009). Mesial temporal may also concurrently show signs of cognitive

Fig. 13.9. Clinical factors suggesting the diagnosis of an autoimmune epilepsy syndrome. Used with permission of Mayo Foun-
dation for Medical Education and Research. All rights reserved.
238 J. BRITTON
Table 13.2 presence of diabetes and vitiligo may suggest GAD-65
Clinical features suggestive of autoimmune epilepsy antibody positivity; hyponatremia may suggest VGKC–
complex antibodies; gastrointestinal dysmotility may
Acute to subacute onset (maximal seizure frequency suggest ganglionic acetylcholine receptor antibodies;
3 months) occipital calcifications may suggest celiac disease, and
Multiple seizure types in same patient diffuse rhythmic delta on EEG should suggest NMDA
Faciobrachial dystonic seizures
receptor antibodies. Similarly, the presence of bilateral
Antiepileptic drug resistance
hippocampal swelling and increased FLAIR or T2 signal
Personal or family history (first-degree relative) of
autoimmunity hyperintensity in a patient with seizures suggests limbic
History of recent or past neoplasia encephalitis, which should prompt evaluation for auto-
Viral prodrome immune and, in some cases, paraneoplastic causes. Sim-
Evidence of central nervous system inflammation ilar signal changes in the basal ganglia may prompt
● CSF (elevated protein, pleocytosis, oligloclonal bands, + CSF consideration of VGKC–complex encephalitis more spe-
index) cifically (Kotsenas et al., 2014). Also, medial temporal
● MRI (mesial temporal or parenchymal T2 hyperintensity) FDG-PET hypermetabolic abnormalities are a finding
● Hypermetabolism on functional imaging (PET) that is relatively unique to limbic encephalitis and, if pre-
Detection of neural autoantibody sent in a patient with seizures, should raise suspicion of
an antibody-mediated etiology.
CSF, cerebrospinal fluid; MRI, magnetic resonance imaging; PET,
positron emission tomography.
Evaluation of patients with suspected
autoimmune epilepsy
impairment, encephalopathy, significant psychiatric and
behavioral changes, movement disorder symptoms EEG is useful in the evaluation of patients suspected of
(myoclonus, tremor, dyskinesia), and new uncharacteris- having autoimmune epilepsy syndromes. The EEG is
tic headaches since onset of seizures. Seizure rates tend abnormal in over 90% of patients (Quek et al., 2012).
to be significantly higher in autoimmune epilepsy syn- These include focal or diffuse slowing of the back-
dromes than in epilepsy due to other etiologies, often ground, encephalopathic abnormalities such as triphasic
occurring several times per day. In our series, 81% of waves, interictal epileptiform abnormalities, and sei-
patients were experiencing daily seizures (Quek et al., zures (Rudzinski et al., 2011). Also, subclinical seizures
2012). The seizures usually have features suggesting contributing to cognitive deficits may be present, and
involvement of the limbic regions, although extralimbic can be detected on EEG. Continuous EEG monitoring
seizures may also be present (Rudzinski et al., 2011). can help more clearly determine the seizure frequency
The seizures in some patients may be brief and not in patients with autoimmune epilepsy syndromes. The
associated with loss of awareness. In such seizures, the EEG may also serve a role in helping to monitor the
symptoms may consist primarily of aura-like phenomena effects of treatment (Fig. 13.3).
with rising and descending sensations, accompanied by Neuroimaging is important in order to help exclude
features suggesting autonomic involvement. Manifesta- other causes of seizures, such as mass lesions, vascular
tions consistent with faciobrachial dystonic seizures disorders, and other disease processes. Despite the char-
should prompt testing for VGKC–complex antibodies. acteristic findings that can be seen on MRI at least in lim-
The EEG may be notably unremarkable during these bic encephalitis cases, it is important to realize that
and other focal, brief seizures that may occur in associ- structural MRI may be normal in the autoimmune epi-
ation with autoimmune epilepsy syndromes. This is even lepsies. In our autoimmune epilepsy series, for example,
true in cases of epilepsy partialis continua. Clearly, MRI was nondiagnostic in 47% of cases (Quek et al.,
however, more typical complex partial seizures, secondary 2012). Negative MRI is more the rule than the exception
generalized tonic-clonic seizures, and nonconvulsive in NMDA receptor encephalitis. Finally, FDG-PET may
complex partial status epilepticus also occur in these show hypermetabolic abnormalities in the limbic and
patients, in which case ictal EEG abnormalities will basal ganglia structures in autoimmune epilepsy syn-
usually be present. Autoimmune epilepsy syndromes dromes, which are not typical for epilepsy due to other
may present with accompanying neurologic and nonneur- etiologies. However, it should be noted that FDG-PET
ologic features that suggest a particular syndrome. For may be unremarkable in these patients, and may also
example, the presence of features such as psychosis, agi- show hypometabolic abnormalities, even in active auto-
tation, limb and oral dyskinesias, autonomic disturbances, immune epilepsy cases.
catatonia and respiratory failure, should suggest NMDA Serologic testing is critical in patients suspected
receptor antibody-associated encephalitis. Similarly, the of having an autoimmune epilepsy syndrome. The
AUTOIMMUNE EPILEPSY 239
identification of certain neural antibodies can help treat- The initial first-line therapy typically used in treat-
ment decisions, as clinical response to immunotherapy is ment of autoimmune epilepsy is corticosteroid therapy.
relatively well known and favorable for certain anti- Parenteral methylprednisolone is most commonly used;
bodies. Conversely, the presence of certain other anti- however, no trials have been performed in order to deter-
bodies (such as those targeting intraneuronal antigens) mine if IV therapy is superior to oral. A typical dosing
and the lack of an antibody in otherwise suspicious cases strategy is administration of 1 gram of methylpredniso-
have been shown to correlate with a less satisfactory lone daily for 3–5 days, then weekly for 6–12 weeks. The
response to treatment, which may prompt a pause prior potential side effects of corticosteroid therapy are well
to initiation of treatment (Toledano et al., 2014). Another known and include weight gain, hypertension, hypergly-
role of serologic testing is to help make a judgment as cemia, insomnia, mood changes, and susceptibility to
to the likelihood of the presence of an underlying opportunistic infections. For this reason, sulfamethoxa-
neoplasm. For example, the presence of ANNA-1 (anti- zole with trimethoprim is usually considered to prevent
Hu) and anti-GABA-B antibodies alone or in combination Pneumocystis pneumonia. With long-term corticoste-
with other antibodies should prompt evaluation for small- roid use, osteopenia can develop. To prevent this,
cell lung carcinoma or other underlying neoplasm. calcium and vitamin D supplementation is usually
CSF examination may be indicated in order to exclude advised. Another complication of chronic corticosteroid
other etiologies, such as herpes encephalitis and other use is aseptic necrosis of the hips, which should be con-
infectious meningoencephalitides. CSF may be tested sidered in the event of the development of hip or groin
for the presence and titer of neural antibodies as well, pain during treatment.
as neural antibodies may be absent in serum but present Another commonly used first-line therapy is IVIG
in CSF, as reported in a few cases with NMDA and (Gelfand, 2012). The cost and attendant approval
GABA-B receptor antibody encephalitis, although very requirements often involved make treatment with IVIG
rare with VGKC–complex antibodies. It should be real- challenging. IVIG is typically given at our institution at a
ized that the CSF may be unremarkable in other respects dose of 0.4 g/kg IV daily for 3–5 days, then weekly for
(cells, protein, oligoclonal bands) in up to 60% of cases, 6–12 weeks. IVIG side effects include headache, hives,
including those who respond to immunotherapy (Quek low-grade fever, aseptic meningitis, hemolytic anemia,
et al., 2012). venous thrombosis, and acute renal injury. IgA-depleted
IVIG therapy should be considered in IgA-deficient
patients to prevent the hypersensitivity reaction to IgA
Treatment of autoimmune epilepsy that may occur in association with IVIG administration.
syndromes Plasmapheresis is sometimes used in autoimmune
encephalitis. It tends to be used in critically ill patients
The diagnosis of an autoimmune etiology opens up the in order to reduce autoimmune antibody levels quickly,
potential for immunotherapy in affected patients. although its superiority to other treatments in this setting
Several publications referenced previously attest to the has not been demonstrated. Plasmapheresis usually neces-
dramatic response that can be observed in patients with sitates placement of a special intravenous cannula into the
certain antibody-mediated epilepsy syndromes. Unfor- subclavian or internal jugular vein, which runs risks of
tunately, there are no consensus guidelines for treatment embolism, pneumothorax, and infection. It is also very
of patients with autoimmune epilepsy (see Chapter 10). costly. There is no evidence establishing superiority of
Immunotherapy options include corticosteroid therapy, plasmapheresis over corticosteroid or IVIG treatment.
IVIG, plasmapheresis, rituximab, cyclophosphamide, Treatment typically proceeds as follows (Fig. 13.10).
mycophenolate, and other agents. The results seen in a Initially, a single first-line agent is selected and treatment
consecutive group of patients with autoimmune epilep- response assessed during weeks #6–12 of therapy. If treat-
sies of a variety of types seen in our Autoimmune Neu- ment is ineffective or not tolerated, one of the other first-
rology and Epilepsy clinics are shown in Figure 13.6. The line treatments is usually tried instead. Once a first-line
treatment strategy we use in patients with a newly diag- therapy is deemed successful, the patient is reassessed
nosed autoimmune epilepsy syndrome is shown in during weeks 6–12 to judge whether there has been a
Figure 13.10. This strategy is more effective for certain response. If there is evidence of response, the dosage
antibody-mediated autoimmune epilepsy syndromes interval is gradually increased, and consideration is given
than others. In our experience and in that of other inves- to the initiation of oral chronic immunosuppressant ther-
tigators, patients with antibodies directed at cell surface apy, such as mycophenolate mofetil or azathioprine.
antigens generally fare better than those with antibodies Unfortunately, these disorders are uncommon, and at this
targeting intracellular antigens and those associated with time, there is not enough evidence to make a determina-
T-cell autoimmunity (Dalmau, 2008). tion as to the advisability of chronic immunosuppressive
240 J. BRITTON

Fig. 13.10. Treatment approach in autoimmune epilepsy and antibody-mediated encephalitis. These treatment options have not
been established with randomized control trials and are not approved by the US Food and Drug Administration for this indication.
ANNA-1, type 1 antineuronal nuclear antibody; CRMP5, collapsin response mediator protein-5; Abs, antibodies; AE, autoimmune
epilepsy; NMDAR, N-methyl-D-aspartate receptor; VGKC, voltage-gated potassium channel; LGI1, leucine-rich glioma inacti-
vated 1 protein; CASPR2, contactin-associated protein-2; AMPA, a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid;
GABA-B, gamma-aminobutyric acid B; mGluR, metabotropic glutamate receptor; VGCC, voltage-gated calcium channel;
gAChR, neuronal ganglionic nicotinic acetylcholine receptor antibody; DPPX, dipeptidyl-peptidase-like protein-6; GAD-65,
glutamic acid decarboxylase 65 kD isoform; IVIg, intravenous immunoglobulin. Used with permission of Mayo Foundation
for Medical Education and Research. All rights reserved.

treatment. However, relapses have been described in (Maloney, 2012). In treated patients, CD19 or CD20 lym-
some antibody-mediated autoimmune epilepsy syn- phocyte counts can be followed in order to ensure a full
dromes, providing a rationale for their use. response has been achieved. Initial doses of rituximab are
If first-line therapies fail, second-line therapies can be associated with infusion-related hypersensitivity reactions
considered. Second-line treatments typically used include in 50%, such as urticaria, fevers, and chills. Ten percent
rituximab and cyclophosphamide. Cyclophosphamide is a experience more serious reactions, including hypotension,
cytotoxic agent which lyses lymphocytes, but which may rigors, bronchospasm, and angioedema. Other potential
also affect myeloid cells. Potential side effects of cyclo- reactions include cardiac arrhythmia, acute coronary
phosphamide include alopecia, leukopenia, thrombocyto- syndrome, interstitial lung disease, bowel perforation,
penia, abdominal discomfort, hematuria, peripheral Stevens–Johnson syndrome, and infection. Fulminant
neuropathy, cardiotoxicity, interstitial lung disease, hepatitis B reactivation can occur, so screening for
opportunistic infection, and hematologic and solid tumor chronic hepatitis should be performed prior to treatment.
malignancy. Rituximab is a monoclonal antibody that tar- Neutropenia may occur in 3–27% of patients, and most
gets CD20 antigens, which serve as a specific marker for often develops 3–4 weeks posttreatment. Progressive mul-
antibody-producing B cells. Elimination of CD20 cells tifocal leukoencephalopathy has been described in associ-
leads to a significant reduction in antibody production ation with rituximab use as well.
AUTOIMMUNE EPILEPSY 241
SUMMARY Bien CG, Granata T, Antozzi C et al. (2005). Pathogenesis,
diagnosis and treatment of Rasmussen encephalitis:
The clinical spectrum and understanding of autoimmune a European consensus statement. Brain 128: 454–471.
epilepsy syndromes are expanding. Many challenges Bien CG, Tiemeier H, Sassen R et al. (2013). Rasmussen
remain with respect to diagnosis and treatment. Treat- encephalitis: incidence and course under randomized ther-
ment is highly efficacious in some autoimmune epilepsy apy with tacrolimus or intravenous immunoglobulins.
syndromes due to certain antibody types; however, it is Epilepsia 54: 543–550.
frustratingly ineffective in others. Improvements in the Boronat A, Sabater L, Saiz A et al. (2011). GABA(B) receptor
understanding of immune mechanisms underlying all antibodies in limbic encephalitis and anti-GAD-associated
neurologic disorders. Neurology 76: 795–800.
forms of autoimmune epilepsy are needed in order to
Boronat A, Gelfand JM, Gresa-Arribas N et al. (2013).
devise more uniformly effective therapies.
Encephalitis and antibodies to dipeptidyl-peptidase-like
protein-6, a subunit of Kv4.2 potassium channels. Ann
REFERENCES Neurol 73: 120–128.
ACR (1999). The American College of Rheumatology nomen- Bouquet F, Ventura A, Gobbi G et al. (1992). Coeliac disease, epi-
clature and case definitions for neuropsychiatric lupus syn- lepsy, and cerebral calcifications. The Lancet 340: 439–443.
dromes. Arthritis & Rheum 42: 599–608. Brain L, Jellinek EH, Ball K (1966). Hashimoto’s disease and
Alaedini A, Okamoto H, Briani C et al. (2007). Immune cross- encephalopathy. The Lancet 288: 512–514.
reactivity in celiac disease: anti-gliadin antibodies bind to Brenner T, Sills GJ, Hart Y et al. (2013). Prevalence of neuro-
neuronal synapsin I. J Immunol 178: 6590–6595. logic autoantibodies in cohorts of patients with new and
Amaral TN, Marques Neto JF, Lapa AT et al. (2012). established epilepsy. Epilepsia 54: 1028–1035.
Neurologic involvement in scleroderma en coup de sabre. Briani C, Zara G, Alaedini A et al. (2008). Neurological com-
Autoimmune Diseases 2012: 6. plications of celiac disease and autoimmune mechanisms: a
Andrews PI, Dichter MA, Berkovic SF et al. (2001). prospective study. J Neuroimmunol 195: 171–175.
Plasmapheresis in Rasmussen’s encephalitis. 1996. Brierley JB, Corsellis JAN, Hierons R et al. (1960). Subacute
Neurology 57: S37–41. encephalitis of later adult life mainly affecting the limbic
Antozzi C, Granata T, Aurisano N et al. (1998). Long-term areas. Brain 83: 357–368.
selective IgG immuno-adsorption improves Rasmussen’s Buckley C, Oger J, Clover L et al. (2001). Potassium channel
encephalitis. Neurology 51: 302–305. antibodies in two patients with reversible limbic encepha-
Arinuma Y, Yanagida T, Hirohata S (2008). Association of litis. Ann Neurol 50: 73–78.
cerebrospinal fluid anti-NR2 glutamate receptor antibodies Buenz EJ, Howe CL (2007). Mechanisms of action of IVIG in
with diffuse neuropsychiatric systemic lupus erythemato- adult onset Rasmussen’s encephalitis. Can J Neurol Sci 34:
sus. Arthritis & Rheumatism 58: 1130–1135. 108–109.
Armangue T, Leypoldt F, Malaga I et al. (2014). Herpes sim- Bürk K, Farecki M-L, Lamprecht G et al. (2009). Neurological
plex virus encephalitis is a trigger of brain autoimmunity. symptoms in patients with biopsy proven celiac disease.
Ann Neurol 75: 317–323. Mov Disord 24: 2358–2362.
Arts WFM, Aarsen FK, Scheltens-de Boer M et al. (2009). Burt RK, Traynor A, Statkute L et al. (2006).
Landau-Kleffner syndrome and CSWS syndrome: treatment Nonmyeloablative hematopoietic stem cell transplantation
with intravenous immunoglobulins. Epilepsia 50: 55–58. for systemic lupus erythematosus. JAMA 295: 527–535.
Arya R, Anand V, Chansoria M (2013). Hashimoto encepha- Butler CR, Miller TD, Kaur MS et al. (2014). Persistent anter-
lopathy presenting as progressive myoclonus epilepsy syn- ograde amnesia following limbic encephalitis associated
drome. Eur J Paediatr Neurol 17: 102–104. with antibodies to the voltage-gated potassium channel com-
Bauer J, Bien CG, Lassmann H (2002). Rasmussen’s enceph- plex. J Neurol Neurosurg Psychiatry 85: 387–391. 2014 Apr.
alitis: a role for autoimmune cytotoxic T lymphocytes. Curr Carvajal-Gonzalez A, Leite MI, Waters P et al. (2014). Glycine
Opin Neurol 15: 197–200. receptor antibodies in PERM and related syndromes: charac-
Beatrice G, Lovato L, Cianti R et al. (2008). Novel autoanti- teristics, clinical features and outcomes. Brain 137: 2178–2192.
gens recognized by CSF IgG from Hashimoto’s encephali- Caselli RJ, Boeve BF, Scheithauer BW et al. (1999).
tis revealed by a proteomic approach. J Neuroimmunol 196: Nonvasculitic autoimmune inflammatory meningoenceph-
153–158. alitis (NAIM): a reversible form of encephalopathy.
Bektas O, Yilmaz A, Kendirli T et al. (2012). Hashimoto Neurology 53: 1579–1581.
encephalopathy causing drug-resistant status epilepticus Castillo P, Woodruff B, Caselli R et al. (2006). Steroid-
treated with plasmapheresis. Pediatr Neurol 46: 132–135. responsive encephalopathy associated with autoimmune
Bien CG, Bauer J, Deckwerth TL et al. (2002a). Destruction thyroiditis. Arch Neurol 63: 197–202.
of neurons by cytotoxic T cells: a new pathogenic Chaigne B, Mercier E, Garot D et al. (2013). Hashimoto’s
mechanism in Rasmussen’s encephalitis. Ann Neurol 51: encephalopathy in the intensive care unit. Neurocrit Care
311–318. 18: 386–390.
Bien CG, Urbach H, Deckert M et al. (2002b). Diagnosis and Chapman RW, Laidlaw JM, Colin-Jones D et al. (1978).
staging of Rasmussen’s encephalitis by serial MRI and his- Increased prevalence of epilepsy in coeliac disease. BMJ
topathology. Neurology 58: 250–257. 2: 250–251.
242 J. BRITTON
Choi J, Nordli Jr DR, Alden TD et al. (2009). Cellular injury marker of Hashimoto’s encephalopathy. J Neuroimmunol
and neuroinflammation in children with chronic intractable 162: 130–136.
epilepsy. J Neuroinflammation 6: 38. Gelfand EW (2012). Intravenous immune globulin in autoim-
Chong JY, Rowland LP, Utiger RD (2003). Hashimoto mune and inflammatory diseases. New Engl J Med 367:
encephalopathy: syndrome or myth? Arch Neurol 60: 2015–2025.
164–171. Go CY, Mackay MT, Weiss SK et al. (2012). Evidence-based
Corsellis JA, Goldberg GJ, Norton AR (1968). “Limbic enceph- guideline update: Medical treatment of infantile spasms:
alitis” and its association with carcinoma. Brain 91: 481–496. Report of the Guideline Development Subcommittee of
Cury RG, Hobi Moreira C (2014). Occipital calcification and the American Academy of Neurology and the Practice
celiac disease. New Engl J Med 370: e26. Committee of the Child Neurology Society. Neurology
Dalmau J (2008). Limbic encephalitis and variants related to 78: 1974–1980.
neuronal cell membrane autoantigens. Rinsho Gono T, Takarada T, Fukumori R et al. (2011). NR2-reactive
Shinkeigaku - Clinical Neurology 48: 871–874. antibody decreases cell viability through augmentation of
Dalmau J, Graus F, Rosenblum MK et al. (1992). Anti-Hu- Ca(2+) influx in systemic lupus erythematosus.
associated paraneoplastic encephalomyelitis/sensory neu- Arthritis & Rheumatism 63: 3952–3959.
ronopathy: a clinical study of 71 patients. Medicine González-Duarte A, Cantú-Brito CG, Ruano-Calderón L et al.
(Baltimore) 71: 59–72. (2008). Clinical description of seizures in patients with sys-
Dalmau J, Tüzün E, Wu H-Y et al. (2007). Paraneoplastic anti- temic lupus erythematosus. Eur Neurol 59: 320–323.
N-methyl-D-aspartate receptor encephalitis associated with Granata T, Fusco L, Gobbi G et al. (2003). Experience with
ovarian teratoma. Ann Neurol 61: 25–36. immunomodulatory treatments in Rasmussen’s encephali-
DeGiorgio LA, Konstantinov KN, Lee SC et al. (2001). tis. Neurology 61: 1807–1810.
A subset of lupus anti-DNA antibodies cross-reacts with Graus F, Saiz A (2008). Limbic encephalitis: an expanding
the NR2 glutamate receptor in systemic lupus erythemato- concept. Neurology 70: 500–501.
sus. Nat Med 7: 1189–1193. Graus F, Elkon KB, Cordon-Cardo C et al. (1986). Sensory neu-
Devinsky O, Petito CK, Alonso DR (1988). Clinical and neu- ronopathy and small cell lung cancer: antineuronal antibody
ropathological findings in systemic lupus erythematosus: that also reacts with the tumor. Am J Med 80: 45–52.
the role of vasculitis, heart emboli, and thrombotic throm- Graus F, Saiz A, Lai M et al. (2008). Neuronal surface antigen
bocytopenic purpura. Ann Neurol 23: 380–384. antibodies in limbic encephalitis: clinical–immunologic
England MJ, Liverman CT, Schultz AM et al. (Eds.), (2012). associations. Neurology 71: 930–936.
Epilepsy Across the Spectrum: Promoting Health and Graus F, Boronat A, Xifró X et al. (2010). The expanding clin-
Understanding. The National Academies Press, ical profile of anti-AMPA receptor encephalitis. Neurology
Washington, D. C. 74: 857–859.
Fasano A, Catassi C (2012). Celiac disease. New Engl J Med Hanly JG, Urowitz MB, Su L et al. (2012). Seizure disorders in
367: 2419–2426. systemic lupus erythematosus results from an international,
Fattouch J, Di Bonaventura C, Di Gennaro G et al. (2008). prospective, inception cohort study. Ann Rheum Dis 71:
Electrical status epilepticus “invisible” to surface EEG in 1502–1509.
late-onset Rasmussen encephalitis. Epileptic Disord 10: Hansen HC, Klingbeil C, Dalmau J et al. (2013). Persistent
219–222. intrathecal antibody synthesis 15 years after recovering
Fauser S, Talazko J, Wagner K et al. (2005). FDG-PET and from anti-N-methyl-D-aspartate receptor encephalitis.
MRI in potassium channel antibody-associated non- JAMA Neurol 70: 117–119.
paraneoplastic limbic encephalitis: correlation with clini- Hart YM, Cortez M, Andermann F et al. (1994). Medical treat-
cal course and neuropsychology. Acta Neurol Scand 111: ment of Rasmussen’s syndrome (chronic encephalitis and
338–343. epilepsy): effect of high-dose steroids or immunoglobulins
Fenalti G, Law RHP, Buckle AM et al. (2007). GABA produc- in 19 patients. Neurology 44: 1030–1036.
tion by glutamic acid decarboxylase is regulated by a Hiraga A, Kuwabara S, Hayakawa S et al. (2006). Voltage-
dynamic catalytic loop. Nat Struct Mol Biol 14: 280–286. gated potassium channel antibody-associated encephalitis
Ferracci F, Moretto G, Candeago RM et al. (2003). Antithyroid with basal ganglia lesions. Neurology 66: 1780–1781.
antibodies in the CSF: their role in the pathogenesis of Hoftberger R, Titulaer MJ, Sabater L et al. (2013).
Hashimoto’s encephalopathy. Neurology 60: 712–714. Encephalitis and GABAB receptor antibodies: novel find-
Finke C, Kopp UA, Scheel M et al. (2013). Functional and ings in a new case series of 20 patients. Neurology 81:
structural brain changes in anti-N-methyl-D-aspartate 1500–1506.
receptor encephalitis. Ann Neurol 74: 284–296. Hughes EG, Peng X, Gleichman AJ et al. (2010). Cellular and
Fois A, Vascotto M, Di Bartolo R et al. (1994). Celiac disease synaptic mechanisms of anti-NMDA receptor encephalitis.
and epilepsy in pediatric patients. Childs Nerv Syst 10: J Neurosci 30: 5866–5875.
450–454. Iizuka T, Sakai F, Ide T et al. (2008). Anti-NMDA receptor
Fujii A, Yoneda M, Ito T et al. (2005). Autoantibodies against encephalitis in Japan: long-term outcome without tumor
the amino terminal of alpha-enolase are a useful diagnostic removal. Neurology 70: 504–511.
AUTOIMMUNE EPILEPSY 243
Irani SR, Alexander S, Waters P et al. (2010). Antibodies to Leach JP, Chadwick DW, Miles JB et al. (1999). Improvement
Kv1 potassium channel-complex proteins leucine-rich, gli- in adult-onset Rasmussen’s encephalitis with long-term
oma inactivated 1 protein and contactin-associated immunomodulatory therapy. Neurology 52: 738–742.
protein-2 in limbic encephalitis, Morvan’s syndrome and Li Y, Uccelli A, Laxer KD et al. (1997). Local-clonal expan-
acquired neuromyotonia. Brain 133: 2734–2748. sion of infiltrating T lymphocytes in chronic encephalitis of
Irani SR, Michell AW, Lang B et al. (2011). Faciobrachial dys- Rasmussen. J Immunol 158: 1428–1437.
tonic seizures precede Lgi1 antibody limbic encephalitis. Liimatainen S, Peltola M, Sabater L et al. (2010). Clinical sig-
Ann Neurol 69: 892–900. nificance of glutamic acid decarboxylase antibodies in
Irani SR, Gelfand JM, Bettcher BM et al. (2014). Effect of patients with epilepsy. Epilepsia 51: 760–767.
rituximab in patients with leucine-rich, glioma-inactivated Liou HH, Wang CR, Chen CJ et al. (1996). Elevated levels of
1 antibody–associated encephalopathy. JAMA Neurol 71: anticardiolipin antibodies and epilepsy in lupus patients.
896–900. Lupus 5: 307–312.
Jarius S, Hoffmann L, Clover L et al. (2008). CSF findings Longaretti F, Dunkley C, Varadkar S et al. (2012). Evolution of
in patients with voltage gated potassium channel anti- the EEG in children with Rasmussen’s syndrome. Epilepsia
body associated limbic encephalitis. J Neurol Sci 268: 53: 1539–1545.
74–77. Lu XY, Chen XX, Huang LD et al. (2010). Anti-alpha-
Jeffery OJ, Lennon VA, Pittock SJ et al. (2013). GABAB internexin autoantibody from neuropsychiatric lupus
receptor autoantibody frequency in service serologic eval- induce cognitive damage via inhibiting axonal elongation
uation. Neurology 81: 882–887. and promote neuron apoptosis. PLoS ONE [Electronic
Kang EH, Shen GQ, Morris R et al. (2008). Flow cytometric Resource] 5: e11124.
assessment of anti-neuronal antibodies in central nervous Ludvigsson JF, Zingone F, Tomson T et al. (2012). Increased
system involvement of systemic lupus erythematosus and risk of epilepsy in biopsy-verified celiac disease:
other autoimmune diseases. Lupus 17: 21–25. a population-based cohort study. Neurology 78: 1401–1407.
Kashihara K, Ohno M, Takahashi Y (2010). Twenty-one-year Mahad DJ, Staugaitis S, Ruggieri P et al. (2005). Steroid-
course of adult-onset Rasmussen’s encephalitis and bilat- responsive encephalopathy associated with autoimmune
eral uveitis: case report. J Neurol Sci 294: 127–130. thyroiditis and primary CNS demyelination. J Neurol Sci
Khan NL, Jeffree MA, Good C et al. (2009). Histopathology of 228: 3–5.
VGKC antibody-associated limbic encephalitis. Neurology Majoie HJ, de Baets M, Renier W et al. (2006). Antibodies to
72: 1703–1705. voltage-gated potassium and calcium channels in epilepsy.
Kirkpatrick MP, Clarke CD, Sonmezturk HH et al. (2011). Epilepsy Res 71: 135–141.
Rhythmic delta activity represents a form of nonconvulsive Maloney DG (2012). Anti-CD20 antibody therapy for B-cell
status epilepticus in anti-NMDA receptor antibody enceph- lymphomas. New Engl J Med 366: 2008–2016.
alitis. Epilepsy Behav 20: 392–394. Malter MP, Helmstaedter C, Urbach H et al. (2010).
Klein CJ, Lennon VA, Aston PA et al. (2012). Chronic pain as Antibodies to glutamic acid decarboxylase define a form
a manifestation of potassium channel-complex autoimmu- of limbic encephalitis. Ann Neurol 67. 470–8.
nity. Neurology 79: 1136–1144. Malter MP, Frisch C, Schoene-Bake JC et al. (2014).
Klein CJ, Lennon VA, Aston PA et al. (2013). Insights from Outcome of limbic encephalitis with VGKC-complex
LGI1 and CASPR2 potassium channel complex autoanti- antibodies: relation to antigenic specificity. J Neurol
body subtyping. JAMA Neurol 70: 229–234. 261: 1695–1705.
Kotsenas AL, Watson R, Pittock S et al. (2014). MRI findings Mantegazza R, Bernasconi P, Baggi F et al. (2002). Antibodies
in autoimmune voltage-gated potassium channel complex against GluR3 peptides are not specific for Rasmussen’s
encephalitis with seizures: one potential etiology for mesial encephalitis but are also present in epilepsy patients with
temporal sclerosis. AJNR. severe, early onset disease and intractable seizures.
Krauss GL, Campbell ML, Roche KW et al. (1996). J Neuroimmunol 131: 179–185.
Chronic steroid-responsive encephalitis without autoanti- Marras CE, Granata T, Franzini A et al. (2010).
bodies to glutamate receptor GluR3. Neurology 46: 247–249. Hemispherotomy and functional hemispherectomy: indica-
Lai M, Hughes EG, Peng X et al. (2009). AMPA receptor anti- tions and outcome. Epilepsy Res 89: 104–112.
bodies in limbic encephalitis alter synaptic receptor loca- Matsunaga A, Ikawa M, Fujii A et al. (2013). Hashimoto’s
tion. Ann Neurol 65: 424–434. encephalopathy as a treatable adult-onset cerebellar ataxia
Lalic T, Pettingill P, Vincent A et al. (2011). Human limbic mimicking spinocerebellar degeneration. Eur Neurol 69:
encephalitis serum enhances hippocampal mossy fiber- 14–20.
CA3 pyramidal cell synaptic transmission. Epilepsia 52: McKeon A, Robinson MT, McEvoy KM et al. (2012). Stiff-
121–131. man syndrome and variants: clinical course, treatments,
Lancaster E, Lai M, Peng X et al. (2010). Antibodies to the and outcomes. Arch Neurol 69: 230–238.
GABA(B) receptor in limbic encephalitis with seizures: McLachlan RS, Diosy D, Levin S (2011). Early treatment of a
case series and characterisation of the antigen. Lancet progressive Rasmussen’s like syndrome with ganciclovir.
Neurol 9: 67–76. Can J Neurol Sci 38: 296–298.
244 J. BRITTON
Mikdashi J, Krumholz A, Handwerger B (2005). Factors Rasmussen T, Olszewski J, Lloyd-Smith D (1958). Focal sei-
at diagnosis predict subsequent occurrence of seizures zures due to chronic localized encephalitis. Neurology 8:
in systemic lupus erythematosus. Neurology 64: 435–455.
2102–2107. Rey C, Koric L, Guedj E et al. (2012). Striatal hypermetabo-
Moscato EH, Peng X, Jain A et al. (2014). Acute mechanisms lism in limbic encephalitis. J Neurol 259: 1106–1110.
underlying antibody effects in anti-N-methyl-D-aspartate Rhodes RH, Lehman RM, Wu BY et al. (2007). Focal chronic
receptor encephalitis. Ann Neurol 76: 108–119. inflammatory epileptic encephalopathy in a patient with
Mundiyanapurath S, Jarius S, Probst C et al. (2013). GABA-B- malformations of cortical development, with a review of
receptor antibodies in paraneoplastic brainstem encephali- the spectrum of chronic inflammatory epileptic encepha-
tis. J Neuroimmunol 259: 88–91. lopathy. Epilepsia 48: 1184–1202.
Muscal E, Brey RL (2010). Neurologic manifestations of sys- Rogers SW, Andrews PI, Gahring LC et al. (1994).
temic lupus erythematosus in children and adults. Neurol Autoantibodies to glutamate receptor GluR3 in
Clin 28: 61–73. Rasmussen’s encephalitis. Science 265: 648–651.
Olmez I, Moses H, Sriram S et al. (2013). Diagnostic and ther- Rudzinski LA, Pittock SJ, McKeon A et al. (2011).
apeutic aspects of Hashimoto’s encephalopathy. J Neurol Extratemporal EEG and MRI findings in ANNA-1 (anti-
Sci 331: 67–71. Hu) encephalitis. Epilepsy Res 95: 255–262.
Ong M, Kohane IS, Cai T et al. (2014). Population-level evi- Saiz A, Blanco Y, Sabater L et al. (2008). Spectrum of neuro-
dence for an autoimmune etiology of epilepsy. JAMA logical syndromes associated with glutamic acid decarbox-
Neurol 71: 569–574. ylase antibodies: diagnostic clues for this association. Brain
Pari E, Rinaldi F, Premi E et al. (2014). A follow-up 18 F-FDG 131: 2553–2563.
brain PET study in a case of Hashimoto’s encephalopathy Salvarani C, Brown RD, Calamia KT et al. (2007). Primary
causing drug-resistant status epilepticus treated with plas- central nervous system vasculitis: analysis of 101 patients.
mapheresis. J Neurol 261: 663–667. Ann Neurol 62: 442–451.
Peltola J, Kulmala P, Isojarvi J et al. (2000). Autoantibodies to Schauble B, Castillo PR, Boeve BF et al. (2003). EEG findings
glutamic acid decarboxylase in patients with therapy- in steroid-responsive encephalopathy associated with auto-
resistant epilepsy. Neurology 55: 46–50. immune thyroiditis. Clin Neurophysiol 114: 32–37.
Petersen JS, Hejnaæs KR, Moody A et al. (1994). Detection of Schmitt SE, Pargeon K, Frechette ES et al. (2012). Extreme
GAD65 antibodies in diabetes and other autoimmune dis- delta brush: a unique EEG pattern in adults with anti-
eases using a simple radioligand assay. Diabetes 43: NMDA receptor encephalitis. Neurology 79: 1094–1100.
459–467. Seo SW, Lee BI, Lee JD et al. (2003). Thyrotoxic autoimmune
Petit-Pedrol M, Armangue T, Peng X et al. (2014). encephalopathy: a repeat positron emission tomography
Encephalitis with refractory seizures, status epilepticus, study. J Neurol Neurosurg Psychiatry 74: 504–506.
and antibodies to the GABAA receptor: a case series, char- Sinclair DB, Snyder TJ (2005). Corticosteroids for the treat-
acterisation of the antigen, and analysis of the effects of ment of Landau-Kleffner syndrome and continuous spike-
antibodies. Lancet Neurol 13: 276–286. wave discharge during sleep. Pediatr Neurol 32: 300–306.
Pettingill P, Kramer HB, Coebergh JA et al. (2015). Antibodies Song YM, Seo DW, Chang GY (2004). MR findings in
to GABAA receptor a1 and g2 subunits: clinical and sero- Hashimoto encephalopathy. Am J Neuroradiol 25: 807–808.
logic characterization. Neurology 84: 1233–1241. Spatola M, Stojanova V, Prior JO et al. (2014). Serial brain
Pittock SJ, Yoshikawa H, Ahlskog JE et al. (2006). Glutamic 8FDG-PET in anti-AMPA receptor limbic encephalitis.
acid decarboxylase autoimmunity with brainstem, extrapy- J Neuroimmunol 271: 53–55.
ramidal, and spinal cord dysfunction. Mayo Clin Proc 81: Stagg CJ, Lang B, Best JG et al. (2010). Autoantibodies to glu-
1207–1214. tamic acid decarboxylase in patients with epilepsy are associ-
Plantone D, Renna R, Grossi D et al. (2013). Teaching ated with low cortical GABA levels. Epilepsia 51: 1898–1901.
NeuroImages: Basal ganglia involvement in facio-brachial Stamelou M, Plazzi G, Lugaresi E et al. (2012). The distinct
dystonic seizures associated with LGI1 antibodies. movement disorder in anti-NMDA receptor encephalitis
Neurology 80: e183–184. may be related to status dissociatus: a hypothesis. Mov
Pozo-Rosich P, Clover L, Saiz A et al. (2003). Voltage-gated Disord 27: 1360–1363.
potassium channel antibodies in limbic encephalitis. Ann Striano P, Pagliuca M, Andreone V et al. (2006). Unfavourable
Neurol 54: 530–533. outcome of Hashimoto encephalopathy due to status epi-
Pruss H, Finke C, Holtje M et al. (2012a). N-methyl-D- lepticus. J Neurol 253: 248–249.
aspartate receptor antibodies in herpes simplex encephali- Takahashi Y, Mori H, Mishina M et al. (2005). Autoantibodies
tis. Ann Neurol 72: 902–911. and cell-mediated autoimmunity to NMDA-type
Pruss H, Holtje M, Maier N et al. (2012b). IgA NMDA receptor GluRepsilon2 in patients with Rasmussen’s encephalitis
antibodies are markers of synaptic immunity in slow cog- and chronic progressive epilepsia partialis continua.
nitive impairment. Neurology 78: 1743–1753. Epilepsia 46: 152–158.
Quek AM, Britton JW, McKeon A et al. (2012). Autoimmune Tan EM, Feltkamp TEW, Smolen JS et al. (1997). Range of
epilepsy: clinical characteristics and response to immuno- antinuclear antibodies in “healthy” individuals.
therapy. Arch Neurol 69: 582–593. Arthritis & Rheumatism 40: 1601–1611.
AUTOIMMUNE EPILEPSY 245
Tang Y, Xing Y, Lin MT et al. (2012). Hashimoto’s encephalop- Vianello M, Bisson G, Dal Maschio M et al. (2008).
athy cases: Chinese experience. BMC Neurol 12: 60. Increased spontaneous activity of a network of hippocam-
Tao JX, Baldwin M, Hawes-Ebersole S et al. (2007). Cortical pal neurons in culture caused by suppression of inhibitory
substrates of scalp EEG epileptiform discharges. J Clin potentials mediated by anti-gad antibodies. Autoimmunity
Neurophysiol 24: 96–100. 41: 66–73.
Tekgul H, Polat M, Kitis O et al. (2005). T-cell subsets and Villani F, Spreafico R, Farina L et al. (2001). Positive response
interleukin-6 response in Rasmussen’s encephalitis. to immunomodulatory therapy in an adult patient with
Pediatr Neurol 33: 39–45. Rasmussen’s encephalitis. Neurology 56: 248–250.
Tengah DSNAP, Holmes GKT, Wills AJ (2004). The preva- Vincent A, Crino PB (2011). Systemic and neurologic autoim-
lence of epilepsy in patients with celiac disease. mune disorders associated with seizures or epilepsy.
Epilepsia 45: 1291–1293. Epilepsia 52: 12–17.
Thieben MJ, Lennon VA, Boeve BF et al. (2004). Potentially Vincent A, Irani SR, Lang B (2010). The growing recognition
reversible autoimmune limbic encephalitis with neuronal of immunotherapy-responsive seizure disorders with auto-
potassium channel antibody. Neurology 62: 1177–1182. antibodies to specific neuronal proteins. Curr Opin Neurol
Titulaer MJ, McCracken L, Gabilondo I et al. (2013a). 23: 144–150.
Treatment and prognostic factors for long-term outcome Vining EPG, Freeman JM, Pillas DJ et al. (1997). Why would
in patients with anti-NMDA receptor encephalitis: an you remove half a brain? The outcome of 58 children after
observational cohort study. Lancet Neurol 12: 157–165. hemispherectomy – the Johns Hopkins experience: 1968 to
Titulaer MJ, McCracken L, Gabilondo I et al. (2013b). Late-onset 1996. Pediatrics 100: 163–171.
anti-NMDA receptor encephalitis. Neurology 81: 1058–1063. Watson R, Jiang Y, Bermudez I et al. (2004). Absence of anti-
Toledano M, Britton JW, McKeon A et al. (2014). Utility of an bodies to glutamate receptor type 3 (GluR3) in Rasmussen
immunotherapy trial in evaluating patients with presumed encephalitis. Neurology 63: 43–50.
autoimmune epilepsy. Neurology 82: 1578–1586. Wiendl H, Bien CG, Bernasconi P et al. (2001). GluR3
Tsai J-DL, Cheng-Li Lin, Cheng-Chieh et al. (2014). Risk of antibodies: prevalence in focal epilepsy but no specificity
epilepsy in patients with systemic lupus erythematosus – a for Rasmussen’s encephalitis. Neurology 57: 1511–1514.
retrospective cohort study. Neuropsychiatr Dis Treat 10: Yoshio T, Hirata D, Onda K et al. (2005). Antiribosomal
1635–1643. P protein antibodies in cerebrospinal fluid are associated
Tsutsui K, Kanbayashi T, Tanaka K et al. (2012). Anti- with neuropsychiatric systemic lupus erythematosus.
NMDA-receptor antibody detected in encephalitis, schizo- J Rheumatol 32: 34–39.
phrenia, and narcolepsy with psychotic features. BMC Zandman-Goddard G, Blank M, Shoenfeld Y (2009).
Psychiatry 12: 37. Intravenous immunoglobulins in systemic lupus eryt-
Tüzün E, Dalmau J (2007). Limbic encephalitis and variants: hematosus: from the bench to the bedside. Lupus 18:
classification, diagnosis and treatment. Neurologist 13: 884–888.
261–271. Zhang Q, Tanaka K, Sun P et al. (2012). Suppression of
Tüzün E, Erdağ E, Durmus H et al. (2011). Autoantibodies to synaptic plasticity by cerebrospinal fluid from anti-
neuronal surface antigens in thyroid antibody-positive and NMDA receptor encephalitis patients. Neurobiol Dis 45:
-negative limbic encephalitis. Neurol India 59: 47–50. 610–615.
Varadkar S, Bien CG, Kruse CA et al. (2014). Rasmussen’s Zuliani L, Ferlazzo E, Andrigo C et al. (2014). Glycine recep-
encephalitis: clinical features, pathobiology, and treatment tor antibodies in 2 cases of new, adult-onset epilepsy.
advances. Lancet Neurol 13: 195–205. Neurol Neuroimmunol Neuroinflamm 1: e16.

You might also like