Journal Pre-proof Immune-mediated epilepsy with GAD65 antibodies
Xinxin Li, Qi Guo, Zhaoshi Zheng, Xiaoshuang Wang, Songyan Liu PII:
S0165-5728(20)30027-8
DOI:
https://doi.org/10.1016/j.jneuroim.2020.577189
Reference:
JNI 577189
To appear in:
Journal of Neuroimmunology
Received date:
11 January 2020
Revised date:
11 February 2020
Accepted date:
11 February 2020
Please cite this article as: X. Li, Q. Guo, Z. Zheng, et al., Immune-mediated epilepsy with GAD65 antibodies, Journal of Neuroimmunology (2019), https://doi.org/10.1016/ j.jneuroim.2020.577189
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2019 Published by Elsevier.
Journal Pre-proof Immune-mediated epilepsy with GAD65 antibodies
Xinxin Lia, Qi Guoa, Zhaoshi Zhenga, Xiaoshuang Wanga, Songyan Liub,* a
Department of Neurology, China–Japan Union Hospital of Jilin University,
Changchun, Jilin Province, China b
Department of Neurology, China–Japan Union Hospital of Jilin University, No 126,
oo
Corresponding author.
pr
*
f
Xian tai Street, Changchun, 130000 Jilin Province, China.
e-
Abstract
Pr
Anti-GAD65 antibodies have been identified in both acute/subacute seizures (limbic encephalitis and extralimbic encephalitis) and chronic isolated epilepsy. The evidence of
al
high serum titers and intrathecal synthesis play a fundamental role in diagnosis but poorly
rn
correlate with disease severity or response to therapies. It remains controversial whether
Jo u
anti-GAD65 Abs are the pathogenic entity or only serve as a surrogate marker for autoimmune disorders mediated by cytotoxic T cells. Unlike other immune-mediated epilepsy, although multiple combinations of therapeutics are used, the efficacy and prognosis of patients with GAD65-epilepsy patients are poor. Besides, GAD65-epilepsy is more prone to relapse and potentially evolve into a more widespread CNS inflammatory disorder. This article reviews the recent advances of GAD65-epilepsy, focusing on the diagnosis, epidemiology, pathophysiology, clinical features, and treatment, to better promote the recognition and provide proper therapy for this condition. 1.Introduction
Journal Pre-proof Glutamic acid decarboxylase (GAD) is a key enzyme in the dynamic regulation of neural network excitability that converts glutamate into Gamma-aminobutyric acid (GABA). There are two isoforms of this enzyme: GAD65 and GAD67, which are widely distributed within the central nervous system, pancreas, and other organs[1, 2] . In patients with neurological symptoms, antibodies can be detected against GAD65, but also less frequently against GAD67. Although the significance of anti-GAD65 antibodies(anti-GAD65 Abs) has been a
oo
f
focus of debate, several neurological phenotypes of anti-GAD65 associated neurological
pr
disease have been described, such as stiff-person syndrome (SPS), cerebellar ataxia(CA),
e-
ocular movement disorder and myelitis[3-6] .Lately, anti-GAD65 Abs were found in patients with seizures in the context of encephalitis, and in patients with chronic epileptic
Pr
syndromes without clinical or MRI evidence of active CNS inflammation [7-9]. In some cases,
al
seizures may be the main or the only clinical presentation in encephalitis associated with
rn
anti-GAD65 Abs; thus, “subacute” encephalitis and “chronic” epilepsy seem to represent
Jo u
a continuum of a rare neurological syndrome [10] . To make consistency in terminology, we collectively refer to these diseases as "immune-mediated epilepsy with anti-GAD65 Abs " (GAD65-epilepsy) [11,
12] .
This article reviews the recent advances of GAD65-epilepsy,
focusing on the diagnosis, epidemiology, pathophysiology, clinical features, and treatment, to better promote the recognition and provide proper therapy for this condition. 2.Diagnosis The diagnosis of GAD65-epilepsy is based on clinical grounds plus the presence of high anti-GAD65 Abs titers or the detection of intrathecal synthesis (IS) in CSF. Currently, several recognized high anti-GAD65 Abs levels are defined as follows: 1) usually> 20
Journal Pre-proof nmol/L or >2000 U/mL when determined by radioimmunoassay(RIA);2)> 20 nmol/L or >1000 IU/mL by Enzyme-linked immunosorbent assay (ELISA);3) screened by immunohistochemistry(IHC) and confirm the positive cases by RIA or Western Blot (WB)[10,
12-17] .
High anti-GAD65 Abs titers have high specificity for neurological
autoimmunity, and only 0.8% of patients with T1DM detected anti-GAD65 Abs titers above 2000 U/mL [13] . 20% of patients with Abs were only present in serum, and the serum Abs
oo
f
titers are often higher than CSF [10, 18] . However, in some cases, the serum Ab titers might
pr
be low, but still show clear evidence of IS in CSF. Therefore, Clinically, patients with highly
e-
suspected GAD65-epilepsy should be simultaneously screened for the titer of anti-GAD65 Abs in serum and CSF. The IS index of anti-GAD65 Abs was calculated using the values
Pr
obtained by immunohistochemistry, based on the following formula: [CSF anti-GAD65
al
Abs titer/serum anti-GAD65 Abs titer]/ [CSF albumin/serum albumin], Values higher than
19] .IS
suggests active and ongoing CNS inflammation that could identify patients
Jo u
IgG[13,
rn
the IgG index, particularly higher than 1, are strong indicators for IS of antibody-specific
who might be warranted an immunosuppressive therapy [1, 10] . 3.Epidemiology
Up to 80% of type I diabetes(T1DM) have low-positive anti-GAD65 Abs (< 100 U/mL); furthermore, such low-positive antibodies may have been detected in 1 to 2% of healthy people and are relatively non-specific
[20, 21] .
The true incidence of GAD65-epilepsy
remains unknown. In the existing studies, due to the method of determination and the levels of antibodies considered relevant have been variable, discrepant results do exist , and most studies are limited by lack of CSF data. Recently, a population-based study from
Journal Pre-proof Olmsted County, USA, estimated the prevalence of autoimmune encephalitis to be13.7/100,000 people,of which anti-GAD65 Abs was found to be one of
the most
common autoantibodies, with a prevalence of 1.9/100,000 [22] .A recent hospital-based prospective study reported that 8% of new-onset or established epilepsy of unknown etiology had high titers of anti-GAD65 Abs [7] .Other studies reported that high anti-GAD65 Abs titers have been detected in 2.1 to 5.4% of adult-onset focal epilepsy(more frequently
research on children, high anti-GAD65 Abs titers have been detected in 0.5% to
pr
23] .Less
[10, 14, 18,
oo
f
exhibiting drug-resistant seizures) and 6.5%-17% of limbic encephalitis (LE)
encephalopathies
[24-26]. Moreover,
e-
0.8% in pediatric epilepsy, and 2% in pediatric patients with cryptogenic forms of epileptic anti-GAD65 Abs were detected 8.7% in pediatric
Pr
epileptic patients with an undetermined etiology and up to 35.3% in patients with
28] .
However, the diagnostic value of these low anti-GAD65 Abs titers to
rn
levels[27,
al
encephalitis and childhood status epilepticus, but none of them reached the high titer
Jo u
neurological disease remains unclear. 4.Pathophysiology
4.1Structural and functional of the GAD The neuroendocrine enzyme GAD catalyzes the synthesis of Gamma-aminobutyric acid (GABA), which is the prime inhibitory neurotransmitter that is necessary to the control of synaptic excitation/inhibition and neural oscillation[29]. GAD involves two isoforms that catalyze GABA synthesis named according to their respective molecular weights: GAD65 and GAD67[30] . The two isoforms have the same fold and general overall sequence similarity, each having an amino (N)-terminal domain, a middle PLP-binding domain
Journal Pre-proof containing the active catalytic site of the enzyme, and a carboxy (C) -terminal domain, with the middle and C-terminal domains having 74% identity, 25% identity for the N-terminal domain[4] . GAD67 is scattered all over the cytoplasm, is an active element and constantly active to produce basal levels of GABA, whereas GAD65 is mainly located in synaptic vesicles, is temporarily activated, undergoes auto-inactivation during enzyme activity and occurs in the cell primarily as apoenzyme, providing for a pulse in production under
oo
f
circumstances that demand a rapid surge of GABA synthesis and release [30,
31] .The
inhibitory neurotransmission
subtype-specific
knockdown
animals
at
the
e-
subsequent
pr
activity and expression of the two isoforms are highly correlated with GABA levels and
have
synapses[32] .GAD67
shown
that
the
reduced
interneuron inhibitory
Pr
neurotransmission caused by GAD67 deficiency results in behavioral changes related to
al
anxiety and schizophrenia[33] .In GAD65 knockout mice, the level of GABA is a greater
only the GAD65 functions as an autoantigen, the GAD67 is rarely
Jo u
35] .However,
rn
than 50% decrease, and the mice appear to be more vulnerable to stress and seizures [34,
autoantigenic in isolation[36,
37] .
Comparison of GAD65 and GAD67 reveals that the
binding site of GAD65 for antibodies displays greater polarity and more negatively charged amino acid residues on its surface, and the C-terminal and catalytic loop residues of GAD65 display more flexibility and mobility, which may enhance the propensities of antigenicity[4, 30, 38]. 4.2 Epitope features of Anti-GAD65 Abs Anti-GAD65 Abs autoimmunity is associated with a variety of syndromes, from T1DM to neurological diseases such as SPS, CA, as well as limbic encephalitis (LE) and epilepsy.
Journal Pre-proof Immunodominant epitope-specific recognition is considered to be one of the causes of different clinical symptoms. Studies disclosed that antibodies epitopes were tightly clustered in the region surrounding the C-terminal flexible region, and segregated into two distinct C-terminal clusters (b78 and 96.11) located to opposite faces of the C-terminal domain[4, 39] . Anti-GAD65 Abs from patients with neurological disorders have been shown to preferentially recognize the b78 epitope specificity, which tended to be enzyme
oo
f
inhibitory, while that in T1DM patients recognize more commonly the b96.11 epitope
pr
specificity, which tended to be non-enzyme inhibitory [40-42] . Antibodies against the b78
e-
epitope have demonstrated to interferes with the association of GAD65 with the cytosolic face of GABA-containing synaptic vesicles, inhibit the enzymatic activity of GAD65.
Pr
Besides, cerebellar slice experiments suggested a gradual but sustained inhibitory effect
al
of the b78 epitope on the GABAergic synaptic transmission between basket cells and
In neurological disorders linked to anti-GAD65 Abs, researches on specific epitopes
Jo u
44].
rn
Purkinje cells. These effects were not observed with Abs against the b96.11 epitope[1, 41, 43,
recognition was contradictory. Vianello et al.
[45] used
immunofluorescence on cultured
hippocampal neurons, and staining patterns differed among patients with SPS, CA,and epilepsy-related to anti-GAD65 Abs; the tentative suggestion was that differences in GAD65 epitope recognition differentiated clinical phenotypes. But in two other studies of GAD65 epitope mapping, no differences in specific epitopes recognized was found among various symptoms, and only reported that the serum of patients with LE was more likely to react with the N-terminal domain, whereas epileptic patients showed more reactivity against the C-terminal domain, but this difference of epitope recognition was insignificant
Journal Pre-proof and not observed in the CSF [36,
46] .
A recent study showed that SPS patients could
recognize a linear epitope at the N-terminal region of GAD65 ,preferably, and inhibited GAD65 enzyme activity better compared to epilepsy patients
[47] ,
but these are not
sufficient, and further specific identification studies of the epitope of GAD65 are necessary. 4.3 Do Anti-GAD65 Abs Play a Pathogenic Role in GAD65-epilepsy?
oo
f
It has been postulated that the humoral immune response to anti-GAD65 Abs could
pr
result in reduced amounts of presynaptic available GABA content through reducing GABA
[41, 48-51] .
e-
synthesis in nerve terminals and interfering with exocytosis of GABA in synaptic vesicles In an MRI-spectroscopy study, low cortical GABA levels were reported in epileptic
Pr
patients with high levels of serum anti-GAD65 Abs
[52] .
Nevertheless, it remains
al
controversial that anti-GAD65 Abs are the pathogenic entity of GAD65-epilepsy. Firstly,
rn
the results of studies on the direct pathogenic influence of anti-GAD65 Abs affecting
Jo u
inhibitory synaptic transmission in central GABAergic pathways are inconsistent . Increased spontaneous activity of a network of hippocampal neurons in culture caused by registering on cultured hippocampal neurons following application of serum from epileptic patients with anti-GAD65 Abs while no effect was noted using sera from negative controls [53] .However,
stereotactic injection of human CSF containing anti-GAD65 Abs into the
hippocampus of rats in vivo did not alter evoked and spontaneous GABAergic synaptic transmission intact in the recent two studies[54,
55] .
Secondly, GAD65 is cytosolic and,
therefore, not readily accessible for circulating antibodies. It generally acts as a biomarker for cytotoxic T-cell mediated neuronal damage
[56] .
Nowadays, the effector functions of T
Journal Pre-proof cells in GAD65-epilepsy have attracted attention. In pathological studies of LE and unilateral TLE who underwent amygdalohippocampectomy, patchy inflammation by T cells was observed in the hippocampi. Instead, immunoglobulin and complement deposition was completely absent
[57-59] .
Experimental studies have shown that cytotoxic
t-lymphocyte attack on neurons may lead to perforin-dependent electrical silencing, thereby causing the cluster of differentiation CD8+ T cells to play a major role in neuronal
attacks
against
GAD65-expressing
pr
neurons[60] . Thus, these nonlethal cytotoxic
oo
f
damage in CNS inflammation, but this damage does not necessarily cause the death of
e-
GABAergic interneurons and ensuing cellular silence/dysfunction could lead to chronic epilepsy[12, 57].Thirdly, if anti-GAD65 Abs are pathogenic, they need to be internalized and
Pr
reach the intracellular targets. Some experiments have confirmed the internalization of
al
human monoclonal anti-GAD65Ab b78 in cultured cells or rat cerebral cortex, and have
rn
observed b78 in CA1 interneurons and Purkinje neurons in the medial septum/diagonal
Jo u
band and ipsilateral interpositus nucleus [41, 50, 61, 62] . However, it was not confirmed in a study involving patients with LE and epilepsy [46] .Furthermore, GAD65 can become membrane-associated or anchored to synaptic vesicles through protein complex formation with the heat shock protein 70 families
[63] .
Thus, GAD65 could be transiently
exposed on the cell surface during exocytosis from GABAergic neurons. Alternatively, the antibodies may be endocytosed and transported internally to their cytoplasmic antigen , allowing a pathogenic antibody-antigen interaction to occur[64-66] . Moreover, anti-GAD65 Abs may coexist with additional cell-surface antibody, such as anti-NMDA-R, anti-VGKC, anti-GABAA-R, or GABAB-R[67-6 9] .These antibodies might alter neuronal function by
Journal Pre-proof receptor competition and internalization and are directly pathogenic to cells. At this time, anti-GAD65 Abs might rather be a bystander effect generated by blood-brain barrier permeability and widespread neuronal destruction [68] . 5.Clinical Features GAD65-epilepsy may be found in two different settings: acute/subacute onset of seizures or chronic epilepsy, and the clinical manifestations are heterogeneous with a
oo
f
wide clinical spectrum ranging from mild non-pharmacoresistant epilepsy to refractory
pr
TLE, LE, and also extralimbic encephalitis (ELE). Most cases were young adult patients,
males have a slight predominance [27, 70] .
Pr
5.1chronic epilepsy
e-
and women were more frequently affected than men; however, in the pediatric population,
al
Historically, Giometto was the first to describe the presence of anti-GAD65 Abs in
rn
patients with chronic epilepsy in 1998[71] .Most patients were focal seizures with clinical
Jo u
localization of the seizure focus to the temporal lobe, and onset in adult life with no history of antecedent cerebral insults and no epileptogenic lesions identified on MRI scanning using appropriate imaging protocols[70].The most common focal seizures were psychic, followed by somatosensorial, motor, and visual symptoms
[72] .Moreover,
recent studies
reported that 9% of to15%of patients with GAD65 epilepsy presented music-induced reflex seizures (MRS), it seems to be a distinctive seizure type of GAD65 epilepsy [72, 73] . Also, some patients experienced different seizure types during disease evolution. 75% of patients found no epileptogenic lesions identified on the first MRI scanning [72] . During the follow-up, the most common imaging abnormality was disproportionate cerebral or
Journal Pre-proof cerebellar volume loss for age. Although a minority of patients had some degree of hippocampal involvement, hippocampal sclerosis (HS) seemed to be not a dominant feature[11,
74] .In
the FDG-PET study, most patients presented hypometabolism in MTLE
and insular structures, and insular involvement could be an important diagnostic clue for GAD65-epilepsy, especially in patients with MTLE epilepsy of unknown origin [72] . 5.2 limbic encephalitis (LE) and extralimbic encephalitis (ELE)
oo
f
LE is subacute onset with progression in less than 3 months,defined by pathological
pr
temporal structural abnormalities on neuroimaging, temporal lobe seizures, and in part, 76] .
Sub-forms of LE is
e-
rapidly progressive memory and affective disturbances[75,
differentiated based on the underlying antigen. One form of LE based on neuronal
Pr
cell-surface antibodies such as voltage-gated potassium channel (VGKC) complex Abs
al
(currently known to react with LGI-1 and CASPR-2 antigens), and the other form of LE
rn
with antibodies targeting intracellular antigens like GAD65 [77] .The main clinical
Jo u
manifestation with LE associated with anti-GAD65 Abs are epilepsy and memory impairment, and psychiatric disorders, orientation, executive functions, and language disorders were also possible[8] .But a “craniofacial dystonia or orofacial dyskinesia ” pattern, as described in VGKC or NMDAR antibodies receptor encephalitis, is lacking in anti-GAD65 Abs-related LE[8] . Compared to patients with LE associated with anti-VGKC complex Abs, those with anti-GAD65 Abs were younger, present first with seizures rather than cognitive symptoms, had oligoclonal bands in the CSF, along with intrathecal synthesis, and showed an unremitting chronic disease course with antibodies persistence[18,
78, 79].Rare
cases may present with prominent dysautonomia, severe
Journal Pre-proof neuropsychological impairments,dementia ,ictal asystole, and may have drug-resistant TLE for several years prior to diagnosis
[80-84] .
Cases involving mixed LE and ELE
subtypes have also been described, but the seizure features were mesial temporal despite ELE involvement[85] .Isolated ELE subtype is uncommon, and only a few have been reported. ELE subtype comprises, clinically and radiographically, a more heterogeneous syndrome, varying according to the location and extent of ELE
[85-88] .
In the setting of LE, brain MRI usually shows MRI T2-weighted
pr
status epilepticus
oo
f
involvement, some patients developed non-convulsive status epilepticus and convulsive
e-
hyperintensity and "swelling" in mesial temporal structure (bilaterally in most cases), and automated mesiotemporal volumetry only displays amygdala enlargement[79, 89] . Follow up
and atrophy[59] . The
brain MRI of
ELE present
with
hyperintense
al
sclerosis
Pr
MRI in patients with prolonged and frequent seizures may demonstrate hippocampal
rn
cortical/subcortical lesions in T2W/FLAIR MRI sequences, progressive changes
Jo u
predominantly affecting the frontal lobes, the occipito-parietal region with some extension into the temporal lobes and other cortical regions, and usually without contrast enhancement[86,
87] .
FDG-PET may show hypermetabolism corresponding to the MRI
lesions, and DTI may show widespread white matter damage already at relatively early disease stages[90, 91] . 5.3Co-existing autoimmune antibodies and diseases Patients with GAD65-epilepsy frequently coexist one or more systemic autoimmune disorders, including diabetes ,thyroiditis,psoriasis, celiac disease, vitiligo, common variable immune deficiency, and others[8] . In addition, some patients are more likely to
Journal Pre-proof have serological evidence of multiple autoantibodies, but without any clinical signs of autoimmune
diseases,
such
antiphospholipid antibodies[10,
as 92] .
anti-TPO,
anti-gliadin,
antinuclear
antibody,
In Gagnon's report, almost half of cases have
co-existing systemic autoimmune conditions, mainly T1DM, followed by autoimmune thyroiditis[8]. These systemic autoimmune conditions and high anti-GAD65 Abs levels can precede the clinical manifestations of GAD65-epilepsy for several years[93] .Furthermore, it
oo
f
is necessary to screen and address endocrinologic abnormalities, particularly in patients
pr
with GAD65-epilepsy that are less responsive to immunosuppressive therapy. One case
e-
reported that exogenous testosterone replacement had played an important role in improving the seizures in a patient with GAD65-epilepsy and low testosterone [94] . Rare
Pr
cases have reported the co-existence of anti-GAD65 Abs and cell-surface antibodies, and
example,
multifocal
asynchronous
sub-cortical/cortical
hyperintensities
are
rn
For
al
this co-existence may result in the variability seen in the clinical and radiographic features.
Jo u
concerning for co-existing anti-GABAA-R antibodies[67] . Also, GAD65-epilepsy may co-exist with visual disturbances or movement disorder when anti-GlyR antibodies are present, or warning a paraneoplastic and refractory disease in the presence of anti-GABAB-R antibodies[69, 95, 96] . Nevertheless, in some cases, the titer of anti-GAD65 Abs was present at a much lower than that in neurologic syndromes and was not related to disease activity[68] . Moreover, some patients with GAD65-epilepsy may evolve into a more widespread CNS inflammatory disorder at some point in their illness, such as SPS, CA, optic neuritis and transverse myelitis, these syndromes may represent a continuum of anti-GAD65 Abs -associated CNS disease [70, 97-99] .
Journal Pre-proof 5.4Paraneoplastic associations Previous studies have suggested that GAD65-epilepsy often associated with non-paraneoplastic forms
[12] ,
little is known of anti-GAD65 Abs in the paraneoplastic
context. However, anti-GAD65 Abs have been detected in patients with lung carcinomas (small-cell and non-small cell), thymoma, breast cancer, and testicular seminoma [100-102] . Although this antibody has less sensitivity and specificity for the presence of malignancy,
oo
f
and are not included as a “classic” paraneoplastic antibody, anti-GAD65 Abs are already
pr
included in commercially available paraneoplastic line-blots[103] .Paraneoplastic etiology in
e-
anti-GAD65 Abs associated isolate epilepsy is extremely rare [104] . The probability of malignancy with anti-GAD65 Abs increases if the presentation matches a classic
Pr
paraneoplastic syndrome or typical of LE with rapid disease progression[101] . Furthermore,
al
it has been shown that the probability of an underlying malignancy was a 7-fold increased
rn
risk in patients with anti-GAD65 Abs and co-existing neuronal cell-surface antibodies,
Jo u
particularly in male patients older than 50 years of age with concomitant anti-GABAB receptor Abs[95,
101, 102] .In
these cases, appropriate and targeted malignancy screening
should be performed. paraneoplastic antibodies may be earlier than any evidence of primary malignancy, so continued, regular malignancy screening is necessary , it is recommended to be at least once every six months for four years [105, 106] . 6.Treatment The treatment of patients with GAD65-epilepsy is comprised of symptomatic therapy (including antiepileptic drugs), immunotherapy, and surgical therapies. Most patients received multiple therapeutic interventions in varying combinations, but the response and
Journal Pre-proof prognosis were poor. Moreover, current treatment recommendations lack randomized controlled trials and are mainly based on case series and clinical experience . Recently, a special study emphasis on treatment timing and the relationship between immunotherapy and anti-epileptic drugs GAD65-epilepsy
into three main
[107] .They
stages:
In
the
divided the clinical course of first
stage, acute reversible
immune-activation causes the first seizure, there is no permanent brain damage, and no
oo
f
visible brain MRI changes [108] . The main focus of treatment should be on immunotherapy,
pr
and early initiation of immunotherapy can prevent from developing refractory epilepsy
e-
even provides complete seizure freedom. In the second stage, there is already subtle irreversible brain damage, which causes MRI abnormalities and refractory epilepsy[58] .
Pr
Immunotherapy is still effective and can resolve edema and abnormal signal changes of
al
MRI. However, after that, focus management should shift to refractory epilepsy. In the
rn
third stage, there was post-inflammatory astrogliosis causing secondary hippocampal
Jo u
sclerosis or to more diffuse brain damage, and it was often considered the underlying “structural” cause of the seizures. Immunotherapy seems to be ineffective, and treatment should focus on seizure control
[91, 109] .
However, for patients with pharmacoresistant
epilepsy without an acute onset, the mentioned above three stages do not seem to explain the clinical course, and the routine use of immunotherapy has not been efficacious [70, 110] .
6.1Immunotherapy regimens The immunotherapy protocol may be subdivided into first-line (immunomodulatory treatment) and second-line therapies (immunosuppressive treatment).First-line therapies
Journal Pre-proof include high-dose intravenous methylprednisolone (IVMP), intravenous immune globulin (IVIG), or plasma exchange (PLEX) [111] . High-dose IVMP (1g per day for 5 days, followed by monthly pulsatile treatment or oral prednisone) were the most frequent applied first-line therapies, with 45% of patients achieving seizure response (50% reduction) . However, relevant side effects were observed in 50% of the patients. IVIG (0.4 g/kg per day for 5 consecutive days) and immunoadsorption were less effective in seizure response but with In some cases, PLEX (5–7 cycles; 1
f
112] .
oo
better tolerability than corticosteroids [11,
[113, 114] .
For patients with improvement, gradual
e-
antibody titers and improve seizures
pr
exchange every other day spread over 10–14 days)can effectively diminish anti-GAD65
reduction in dosing may be required, including a slow taper of oral prednisone, or a
Pr
gradual lengthening of the intervals between intravenous treatments [102,
111] .
In some
al
scenarios, such as refractory status epilepticus, aggressive progression or preventing
rn
relapses, although variable combination immunotherapy of IVMP, PE and IVIG are utilized,
Jo u
there is no or only moderate and short-lived effect for the clinical. Previous studies have demonstrated that multimodality immunotherapy of combination of both first- and second-line therapies are effective
[99, 115-117] .But
it remains not unequivocal which
immunosuppressant may have provided the greatest impact on the outcome since all of the treatments were administered over a short time. Second-line therapy commonly consists
of
Mycophenolate
Cyclophosphamide [12] .Azathioprine
mofetil, and
Azathioprine, mycophenolate
Rituximab, mofetil,
as
and mild
immunosuppressants, are mainly used to maintain remission and reduce dependence on steroids, with varying results. Saidha described two patients with GAD65-epilepsy who
Journal Pre-proof had a partial response to IVIG and prednisolone and successfully improved symptoms with mycophenolate mofetil[116] . In one case report, the patient's condition remained unchanged after 5 years of azathioprine use[118] .In another case report, azathioprine was effective but had a lower clinical response, and was suspended after about 40 days due to severe anemia [114] . Rituximab and cyclophosphamide can be used as first aid therapy in the rapid progression of symptoms, and in some cases, remarkable and lasting
oo
f
improvement or remission were observed[80, 115, 117, 119] . But compared to rituximab, it is not
pr
welcome given the adverse effects of cyclophosphamide. Besides, Triplett reported a
fulminant
encephalopathy
and
e-
case of GAD65-epilepsy presenting with epilepsia partialis continua progressing to a responding
to
rituximab
combined
with
Pr
cyclophosphamide [87]. Other immunosuppressive agents such as Natalizumab and
al
Basiliximab have also been tried without long-term improvement[11, 57] .
rn
6.2Titer of anti-GAD65 value in guiding immunomodulatory treatment
Jo u
anti-GAD65 Abs levels/titers showed poor correlation with the clinical presentation of GAD65-epilepsy, and the value of the change of anti-GAD65 Abs titer in guiding long-term treatment is still unclear. In some cases, serum antibodies remained high titer during and after adequate treatment; however, patients became rapidly asymptomatic without the need for long-term immunomodulatory therapy
[120] .In
some cases, sustained high levels
of anti-GAD65 Abs titers are often associated with clinical adverse effect, and reduced antibody titers are often observed in patients who clinically respond to treatment, but antibodies titers rarely disappear, and often elevated again with clinical symptom recurrence during follow-up.[64,
121] .
Thus, Di Giacomo et al. propose that clinical
Journal Pre-proof responses and "relative" trends in antibodies titers overtime should be used instead of "absolute" values to guide treatment decisions[120] . Furthermore, Gagnon et al. [8] reviewed a total of 58 published cases and did not find a significant difference in outcome between cases whether or not positive anti-GAD65 Abs were reported in CSF. We advocate that the value of anti-GAD65 Abs titer needs to be interpreted in the overall clinical context of the patient under evaluation, patients with positive anti-GAD65 Abs in CSF or high serum
oo
f
titers have a higher risk of clinical adverse effect or recurrence. So long-term follow-up
pr
examinations are necessary and increase the effectiveness of treatment by closely
e-
monitoring the correlation between changes in clinical symptoms and antibody titers. 6.3Antiepileptic Drugs
Pr
Even though seizures in GAD65-epilepsy are resistant to antiepileptic drugs (AEDs),
al
some patients may respond adequately to treatment with only AEDs from the beginning of
rn
the inflammatory disease or isolated seizures, and some patients can benefit from AEDs
Jo u
after Immunotherapy failure[11, 110] . For this reason, AEDs should be used throughout the GAD65-epilepsy. In the Cabezudo-Garcia, et al. Systematic review, 8% of patients with GAD65-epilepsy responded to AEDs and usually required a combination of various AEDs[122] . There is no randomized clinical trial data to support one AED over another ; theoretically, they should be more responsive to AEDs that enhance GABA function . 6.4Epilepsy Surgery The surgical outcomes of GAD65-epilepsy seem to be worse than with other etiologies of refractory epilepsy. In a retrospective study,8 patients with GAD65-associated TLE underwent anterior temporal lobectomy (ATL); 5 patients improve in seizure frequency but
Journal Pre-proof not seizure freedom[123] .In another study, selective amygdalohippocampectomy (SAH) resection for this disease reduced seizures in 2 of 3 patients[11] , but it should be noted that SAH combined with immunotherapy may lead to worse performance of graphics and language memory compared with immunotherapy alone [124] . But, as mentioned above, In the third stage of GAD65-epilepsy, refractory epilepsy appears to be related to underlying "structural" etiology rather than ongoing inflammatory processes. In these conditions,
oo
f
especially when FDG-PET scanning revealed no hypermetabolic areas or serological and
pr
CSF markers have returned to normal, epilepsy surgery can still be considered as a useful
e-
treatment option to reduce seizure frequency and improved quality of life. Additionally, cases successfully treated with vagal stimulation (VNS) or Deep brain stimulation (DBS)
Pr
have been reported. [107, 109] .
anti-GAD65
Abs
levels
have
been
identified
in
many
patients
with
rn
High
al
7.Conclusions
Jo u
acute/subacute-onset, chronic, or cryptogenic epilepsy. Anti-GAD65 Abs are directed against the rate-limiting enzyme for the synthesis of GABA. Although anti-GAD65 Abs have been demonstrated to inhibit GAD65 enzyme activity and reduce amounts of available GABA content in some experiments, the pathogenic role of anti-GAD65 Abs remains unclear due to the intracellular location of the antigen and the controversial evidence of Abs internalization into neurons. Patients with GAD65-epilepsy frequently coexist multiple autoimmune disorders or auto-antibodies and may evolve into a more widespread CNS inflammatory disorder at some point in their illness. This condition often associated with not paraneoplastic, but if co-existing neuronal cell-surface antibodies are
Journal Pre-proof present, the risk of an underlying malignancy will greatly increase. Although patients with GAD65-epilepsy received multiple therapeutic interventions in varying combinations, the efficacy and prognosis were poor. Early identification of anti-GAD65 Abs and aggressive immunosuppressive treatment has the potential to prevent the development of permanent brain tissue damage, but it is usually only a temporary therapeutic success, so more aggressive and long-term immunosuppressive therapies are needed to prevent disease
oo
f
progression and relapse. Improvement of clinical symptoms may parallel a "relative"
pr
reduction in antibody titers, and patients with positive anti-GAD65 Abs in CSF or
e-
extremely high serum titers have a higher risk of clinical adverse effect or recurrence, so monitoring antibodies titers may be used to guide immunotherapy. In the future,
Pr
prospective studies recruiting patients with GAD65-epilepsy are needed to elucidate
al
better the spectrum of epilepsy-related to anti-GAD65 Abs, and further studies on
Jo u
Contributors
rn
underlying pathophysiology may influence therapeutic management strategies.
All authors planned the manuscript, did the literature search, contributed to the f igures, and wrote, edited, and approved the manuscript. F unding This work was supported by the Jilin Province Science and technology development plan project in China (Grant No.20180311075yy) Disclosure of conflicts of interest None of the authors has any conflict of interest to disclose.
Journal Pre-proof
. 1.
Baizabal-Carvallo, J.F., The neurological syndromes associated with glutamic acid
decarboxylase antibodies. J Autoimmun, 2019. 101: p. 35-47. 2.
Huang, J., et al., Clinical Heterogeneity in Patients with Glutamat e Decarboxylase
Antibody. Neuroimmunomodulation, 2019. 26(5): p. 234-238.
f
Liu, Z., et al., Glutamic Acid Decarboxylase Antibody in a Patient with Myelitis: A
oo
3.
Ali, F., et al., Stiff-person syndrome (SPS) and anti-GAD-related CNS degenerations:
e-
4.
pr
Retrospective Study. Neuroimmunomodulation, 2018. 25(2): p. 68-72.
protean additions to the autoimmune central neuropathies. J Autoimmun, 2011. 37(2):
Burbelo, P.D., et al., High definition profiling of autoantibodies to glutamic acid
al
5.
Pr
p. 79-87.
rn
decarboxylases GAD65/GAD67 in stiff-person syndrome. Biochem Biophys Res
6.
Jo u
Commun, 2008. 366(1): p. 1-7. Dubbioso, R., et al., Anti-GAD antibody ocular flutter: expanding the spectrum of
autoimmune ocular motor disorders. J Neurol, 2013. 260(10): p. 2675-7. 7.
Dubey, D., et al., Neurological Aut oantibody Prevalence in Epilepsy of Unk nown
Etiology. JAMA Neurol, 2017. 74(4): p. 397-402. 8.
Gagnon, M.M. and M. Savard, Limbic Encephalitis Associated With GAD65 Antibodies:
Brief Review of the Relevant literature. Can J Neurol Sci, 2016. 43(4): p. 486-93. 9.
Spatola, M. and J. Dalmau, Seizures and risk of epilepsy in autoimmune and other
inflammatory encephalitis. Curr Opin Neurol, 2017. 30(3): p. 345-353.
Journal Pre-proof 10.
Liimatainen, S., et al., Clinical significance of glutamic acid decarboxylase antibodies
in patients with epilepsy. Epilepsia, 2010. 51(5): p. 760-7. 11.
Malter, M.P., et al., Treatment of immune-mediated temporal lobe epilepsy with GAD
antibodies. Seizure, 2015. 30: p. 57-63. 12.
Daif, A., et al., Antiglutamic acid decarboxylase 65 (GAD65) antibody -associated
epilepsy. Epilepsy Behav, 2018. 80: p. 331-336.
f
Saiz, A., et al., Spectrum of neurological syndromes associated with glutamic acid
oo
13.
pr
decarboxylase antibodies: diagnostic clues for this association. Brain, 2008. 131(Pt
14.
e-
10): p. 2553-63.
Peltola, J., et al., Autoantibodies to glutamic acid decarboxylase in patients with
Walikonis, J.E. and V.A. Lennon, Radioimmunoassay for glutamic acid decarboxylase
al
15.
Pr
therapy-resistant epilepsy. Neurology, 2000. 55(1): p. 46-50.
rn
(GAD65) autoantibodies as a diagnostic aid f or stiff-man syndrome and a correlate of
16.
Jo u
susceptibility to type 1 diabetes mellitus. Mayo Clin Proc, 1998. 73(12): p. 1161-6. Vianello, M., et al., Antigenic differences between neurological and diabetic patients
with anti-glutamic acid decarboxylase antibodies. Eur J Neurol, 2005. 12(4): p. 294-9. 17.
Pittock, S.J., et al., Glutamic acid decarboxylase autoimmunity with brainstem,
extrapyramidal, and spinal cord dysfunction. Mayo Clinic proceedings, 2006. 81(9): p. 1207-1214. 18.
Malter, M.P., et al., Antibodies to glutamic acid decarboxylase define a form of limbic
encephalitis. Ann Neurol, 2010. 67(4): p. 470-8. 19.
Dalak as, M.C., et al., Stiff person syndrome: quantification, specificity, and intrat hecal
Journal Pre-proof synthesis of GAD65 antibodies. Neurology, 2001. 57(5): p. 780-4. 20.
Meinck, H.M., et al., Antibodies against glutamic acid decarboxylase: prevalence in
neurological diseases. J Neurol Neurosurg Psychiatry, 2001. 71(1): p. 100-3. 21.
Brooking, H., et al., A sensitive non-isot opic assay for GAD65 autoantibodies. Clin Chim Acta, 2003. 331(1-2): p. 55-9.
22.
Dubey, D., et al., Autoimmune encephalitis epidemiology and a comparison to
McKnight, K., et al., Serum antibodies in epilepsy and seizure -associated disorders.
24.
e-
Neurology, 2005. 65(11): p. 1730-6.
pr
23.
oo
f
infectious encephalitis. Annals of neurology, 2018. 83(1): p. 166-177.
Borusiak, P., et al., Autoantibodies to neuronal antigens in children with focal epilepsy
Veri, K., et al., Newly-diagnosed pediatric epilepsy is associated with elevated
al
25.
Pr
and no prima facie signs of encephalitis. Eur J Paediatr Neurol, 2016. 20(4): p. 573-9.
rn
autoantibodies to glutamic acid decarboxylase but not cardiolipin. Epilepsy Res, 2013.
26.
Jo u
105(1-2): p. 86-91.
Tekturk, P., et al., Investigation of neuronal auto-antibodies in children diagnosed with
epileptic encephalopathy of unknown cause. Brain Dev, 2018. 40(10): p. 909-917. 27.
Lin, J.J., et al., Antiglutamic acid decarboxylase antibodies in children with
encephalitis and status epilepticus. Pediatr Neurol, 2012. 47(4): p. 252-8. 28.
Bektas, O., et al., Epilepsy and autoimmunity in pediatric patients. Neuropediatrics, 2015. 46(1): p. 13-9.
29.
Hwang, S., et al., Hypoxia regulates the level of glutamic acid decarboxylase enzymes
and interrupts inhibitory synapse stability in primary cultured neurons . Neurotoxicology,
Journal Pre-proof 2018. 65: p. 221-230. 30.
Kass, I., et al., Cofactor-dependent conformational heterogeneity of GAD65 and its
role in autoimmunity and neurotransmitter homeostasis. Proc Natl Acad Sci U S A, 2014. 111(25): p. E2524-9. 31.
Fenalti, G., et al., GABA production by glutamic acid decarboxylase is regulat ed by a
dynamic catalytic loop. Nat Struct Mol Biol, 2007. 14(4): p. 280-6.
f
Lee, S.E., Y. Lee, and G.H. Lee, The regulation of glutamic acid decarboxylases in
oo
32.
Lazarus, M.S., K. Krishnan, and Z.J. Huang, GAD67 deficiency in parvalbumin
e-
33.
pr
GABA neurotransmission in the brain. Arch Pharm Res, 2019.
interneurons produces deficits in inhibitory transmission and net work disinhibition in
Qi, J., et al., Enhanced susceptibility to stress and seizures in GAD65 deficient mice.
al
34.
Pr
mouse prefrontal cortex. Cereb Cortex, 2015. 25(5): p. 1290-6.
Muller, I., G. Caliskan, and O. Stork, The GAD65 k nock out mouse - a model f or
Jo u
35.
rn
PLoS One, 2018. 13(1): p. e0191794.
GABAergic processes in fear- and stress-induced psychopathology. Genes Brain Behav, 2015. 14(1): p. 37-45. 36.
Fouka, P., et al., GAD65 epitope mapping and searc h for novel aut oantibodies in
GAD-associated neurological disorders. J Neuroimmunol, 2015. 281: p. 73-7. 37.
Arafat, Y., et al., Structural determinants of GAD antigenicity. Mol Immunol, 2009. 47(2-3): p. 493-505.
38.
Fenalti, G. and A.M. Buckle, Structural biology of the GAD autoantigen. Autoimmun Rev, 2010. 9(3): p. 148-52.
Journal Pre-proof 39.
Fenalti, G. and M.J. Rowley, GAD65 as a prototypic autoantigen. J Autoimmun, 2008. 31(3): p. 228-32.
40.
Ganelin-Cohen, E., et al., Epilepsy and behavioral changes, type 1 diabetes mellitus
and a high titer of glutamic acid decarboxylase antibodies. Pediatr Diabetes, 2016. 17(8): p. 617-622. 41.
Manto, M., et al., Disease-s pecific monoclonal antibodies targeting glutamate
oo
f
decarboxylase impair GABAergic neurotransmission and affect motor learning and
Cheramy, M., et al., Characteristics of in-vitro phenotypes of glutamic acid
e-
42.
pr
behavioral functions. Front Behav Neurosci, 2015. 9: p. 78.
decarboxylase 65 autoantibodies in high -titre individuals. Clin Exp Immunol, 2013.
Manto, M., H. Mitoma, and C.S. Hampe, Anti-GAD Antibodies and the Cerebellum:
al
43.
Pr
171(3): p. 247-54.
Hampe, C.S., et al., Monoclonal antibodies to 65kDa glutamate decarboxylase induce
Jo u
44.
rn
Where Do We Stand? Cerebellum, 2019. 18(2): p. 153-156.
epitope specific effects on motor and cognitive functions in rats. Orphanet J Rare Dis, 2013. 8: p. 82. 45.
Vianello, M., et al., Peculiar labeling of cultured hippocampal neurons by different sera
harboring anti-glutamic acid decarboxylase autoantibodies (GAD -Ab). Exp Neurol, 2006. 202(2): p. 514-8. 46.
Gresa-Arribas, N., et al., Antibodies to inhibitory synaptic proteins in neurological
syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS One, 2015. 10(3): p. e0121364.
Journal Pre-proof 47.
Liimatainen, S., et al., GAD65 autoantibody characteristics in patients with
co-occurring type 1 diabetes and epilepsy may help identify underlying epilepsy etiologies. Orphanet J Rare Dis, 2018. 13(1): p. 55. 48.
Vega-Flores, G., et al., The GABAergic septohippocampal pathway is directly involved
in internal processes related to operant reward learning. Cereb Cortex, 2014. 24(8): p. 2093-107.
f
Manto, M.U., et al., Effects of anti-glutamic acid decarboxylase antibodies associated
oo
49.
Manto, M.U., et al., Respective implications of glut amate decarboxylase antibodies in
e-
50.
pr
with neurological diseases. Ann Neurol, 2007. 61(6): p. 544-51.
stiff person syndrome and cerebellar ataxia. Orphanet J Rare Dis, 2011. 6: p. 3. Ishida, K., H. Mitoma, and H. Mizusawa, Reversibility of cerebellar GABAergic
Pr
51.
al
synapse impairment induc ed by anti -glutamic acid decarboxylase autoantibodies. J
Stagg, C.J., et al., Autoantibodies to glutamic acid decarboxylase in patients with
Jo u
52.
rn
Neurol Sci, 2008. 271(1-2): p. 186-90.
epilepsy are associated with low cortical GABA levels. Epilepsia, 2010. 51(9): p. 1898-901. 53.
Vianello, M., et al., Increased spontaneous activity of a network of hippocampal
neurons in culture caused by suppression of inhibitory potentials mediated by anti -gad antibodies. Autoimmunity, 2008. 41(1): p. 66-73. 54.
Hackert, J.K., et al., Anti-GAD65 Containing Cerebros pinal Fluid Does not Alter
GABAergic Transmission. Front Cell Neurosci, 2016. 10: p. 130. 55.
Stemmler, N., et al., Serum from a Patient with GAD65 Antibody-Associated Limbic
Journal Pre-proof Encephalitis Did Not Alter GABAergic Neurotransmission in Cultured Hippocampal Networks. Front Neurol, 2015. 6: p. 189. 56.
Dalmau, J. and F. Graus, Antibody-Mediated Encephalitis. N Engl J Med, 2018. 378(9): p. 840-851.
57.
Widman, G., et al., Treating a GAD65 Antibody-Associated Limbic Encephalitis with
Basiliximab: A Case Study. Front Neurol, 2015. 6: p. 167.
f
Bien, C.G., et al., Immunopathology of autoantibody-associated enc ephalitides: clues
oo
58.
Kumar, G., et al., Histopathological evidence that hippocampal atrophy following
e-
59.
pr
for pathogenesis. Brain, 2012. 135(Pt 5): p. 1622-38.
status epilepticus is a result of neuronal necrosis. J Neurol Sci, 2013. 334(1-2): p.
Meuth, S.G., et al., Cytotoxic CD8+ T cell-neuron interactions: perforin-dependent
al
60.
Pr
186-91.
rn
electrical silencing precedes but is not causally linked to neuronal cell death. J
61.
Jo u
Neurosci, 2009. 29(49): p. 15397-409. Chang, T., et al., Immunization against GAD induces antibody binding to
GAD-independent antigens and brainstem GABAergic neuronal loss. PLoS One, 2013. 8(9): p. e72921. 62.
Chang, T., et al., Neuronal surface and glutamic acid decarboxylase autoantibodies in
Nonparaneoplastic stiff person syndrome. JAMA Neurol, 2013. 70(9): p. 1140-9. 63.
Hsu, C.C., et al., Association of L-glutamic acid decarboxylase to the 70 -kDa heat
shock protein as a potential anchoring mechanism to synaptic vesicles. J Biol Chem, 2000. 275(27): p. 20822-8.
Journal Pre-proof 64.
Errichiello, L., et al., Autoantibodies to glutamic acid decarboxylase (GAD) in focal and
generalized epilepsy: A study on 233 patients. J Neuroimmunol, 2009. 211(1-2): p. 120-3. 65.
Alexopoulos, H. and M.C. Dalakas, The immunobiology of autoimmune encephalitides. J Autoimmun, 2019. 104: p. 102339.
66.
Dalak as, M.C., Progress and stiff challenges in understanding the role of
Gagnon, M.M., M. Savard, and K. Mourabit Amari, Refractory status epilepticus and
pr
67.
oo
f
GAD-antibodies in stiff-person syndrome. Exp Neurol, 2013. 247: p. 303-7.
e-
autoimmune encephalitis with GABAAR and GAD65 antibodies: A case report. Seizure, 2016. 37: p. 25-7.
Kammeyer, R. and A.L. Piquet, Multiple co-existing antibodies in aut oimmune
Pr
68.
al
encephalitis: A case and review of the literature. J Neuroimmunol, 2019. 337: p.
Hoftberger, R., et al., Encephalitis and GABAB receptor antibodies: novel findings in a
Jo u
69.
rn
577084.
new case series of 20 patients. Neurology, 2013. 81(17): p. 1500-6. 70.
Lilleker, J.B., V. Biswas, and R. Mohanraj, Glutamic acid decarboxylase (GAD)
antibodies in epilepsy: diagnostic yield and therapeutic implications. Seizure, 2014. 23(8): p. 598-602. 71.
Giometto,
B.,
et
al.,
Temporal-lobe
epilepsy
associated
with
glutamic-acid-decarboxylase autoantibodies. Lancet, 1998. 352(9126): p. 457. 72.
Falip, M., et al., Hippocampus and Ins ula Are Targets in Epileptic Patients With
Glutamic Acid Decarboxylase Antibodies. Front Neurol, 2018. 9: p. 1143.
Journal Pre-proof 73.
Falip, M., et al., Musicogenic reflex seizures in epilepsy with glutamic acid
decarbocylase antibodies. Acta Neurol Scand, 2018. 137(2): p. 272-276. 74.
Fredriksen, J.R., et al., MRI findings in glut amic acid decarboxylase associated
autoimmune epilepsy. Neuroradiology, 2018. 60(3): p. 239-245. 75.
Graus, F., et al., A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol, 2016. 15(4): p. 391-404.
f
Bien, C.G. and C.E. Elger, Limbic encephalitis: a cause of temporal lobe epilepsy with
oo
76.
Hans en, N., et al., Seizure control and cognitive improvement via immunotherapy in
late
onset
epilepsy
e-
77.
pr
onset in adult life. Epilepsy Behav, 2007. 10(4): p. 529-38.
patients
with
paraneoplastic
versus
GAD65
Feyissa, A.M., A.S. Lopez Chiriboga, and J.W. Britton, Antiepileptic drug therapy in
al
78.
Pr
autoantibody-associated limbic encephalitis. Epilepsy Behav, 2016. 65: p. 18-24.
79.
Jo u
p. e353.
rn
patients with autoimmune epilepsy. Neurol Neuroimmunol Neuroinflamm, 2017. 4(4):
Wagner, J., et al., Automated volumetry of the mesiotemporal structures in
antibody-associated limbic encephalitis. J Neurol Neurosurg Psychiatry, 2015. 86(7): p. 735-42. 80.
Ben Achour, N., et al., Severe dysautonomia as a main feature of anti-GAD
encephalitis: Report of a paediatric case and literature review. Eur J Paediatr Neurol, 2018. 22(3): p. 548-551. 81.
D'Souza, C.E. and A.M. Feyissa, GAD65 antibody-associated autoimmune epilepsy
with unique independent bitemporal -ons et ictal asystole. Epileptic Disord, 2018. 20(3):
Journal Pre-proof p. 204-208. 82.
Witt, J.A., et al., Loss of Autonoetic Awareness of Recent Autobiographical Episodes
and Accelerated Long-Term Forgetting in a Patient wit h Previously Unrecognized Glutamic Acid Decarboxylase Antibody Related Limbic Encephalitis. Front Neurol, 2015. 6: p. 130. 83.
Finelli, P.F., Autoimmune Limbic Encephalitis With GAD Antibodies. Neurohospitalist,
oo
Takagi, M., et al., Cognitive decline in a patient wit h anti-glut amic acid decarboxylase
pr
84.
f
2011. 1(4): p. 178-81.
85.
e-
autoimmunity; case report. BMC neurology, 2011. 11: p. 156. Najjar, S., et al., Extralimbic autoimmune encephalitis associated wit h glut amic acid
Pr
decarboxylase antibodies: an underdiagnosed entity? Epilepsy Behav, 2011. 21(3): p.
Incecik, F., et al., Autoimmune encephalitis associated wit h glut amic acid
rn
86.
al
306-13.
87.
Jo u
decarboxylase antibodies: a case series. Acta Neurol Belg, 2018. 118(3): p. 411-414. Triplett, J., et al., Fulminant Anti-GAD antibody enc ephalitis pres enting with status
epilepticus requiring aggressive immunosuppression. J Neuroimmunol, 2018. 323: p. 119-124. 88.
Cikrikcili, U., et al., Non-convulsive status epilepticus associated with glutamic acid
decarboxylase antibody. Clin EEG Neurosci, 2013. 44(3): p. 232-6. 89.
Wagner, J., B. Weber, and C.E. Elger, Early and chronic gray matter volume changes
in limbic encephalitis revealed by voxel-based morphomet ry. Epilepsia, 2015. 56(5): p. 754-61.
Journal Pre-proof 90.
Kojima, G., M. Inaba, and M.K. Bruno, PET-positive extralimbic presentation of
anti-glutamic acid decarboxylase antibody-associated encephalitis. Epileptic Disord, 2014. 16(3): p. 358-61. 91.
Wagner, J., et al., Distinct white matter integrity in glutamic acid decarboxylase and
voltage-gated potassium channel-c omplex antibody-associated limbic encephalitis. Epilepsia, 2016. 57(3): p. 475-83.
f
Quek, A.M., et al., Autoimmune epilepsy: clinical characteristics and response to
oo
92.
Fauser, S., et al., Long latency between GAD-antibody detection and development of
e-
93.
pr
immunotherapy. Arch Neurol, 2012. 69(5): p. 582-93.
limbic encephalitis--a case report. BMC Neurol, 2015. 15: p. 177. Heiry, M., et al., Improv ement of GAD65-associated autoimmune epilepsy with
Pr
94.
Boronat, A., et al., GABA(B) receptor antibodies in limbic encephalitis and
Jo u
95.
rn
e142.
al
testosterone replacement therapy. Neurol Neuroimmunol Neuroinflamm, 2015. 2(5): p.
anti-GAD-associated neurologic disorders. Neurology, 2011. 76(9): p. 795-800. 96.
Piquet, A.L., et al., Novel clinical features of glycine receptor antibody syndrome: A
series of 17 cases. Neurol Neuroimmunol Neuroinflamm, 2019. 6(5): p. e592. 97.
Schaefer, S.M. and J.J. Moeller, Anti-GAD antibody syndrome with concomitant
cerebellar ataxia, stiff person syndrome, and limbic encephalitis. Neurol Clin Pract, 2015. 5(6): p. 502-504. 98.
Flores -Cantu, H., et al., Anti-GAD Antibody, Seizures, Cerebellar Ataxias and Vitiligo:
A Diagnostic Challenge. Cerebellum, 2015. 14(3): p. 375-7.
Journal Pre-proof 99.
Dubey, D., et al., Effectiveness of multimodality treatment for autoimmune limbic
epilepsy. Epileptic Disord, 2014. 16(4): p. 494-9. 100.
Zekeridou, A., et al., Paraneoplastic autoimmunity and small-cell lung cancer:
Neurological and serological accompaniments. Thorac Cancer, 2019. 10(4): p. 1001-1004. 101.
Arino, H., et al., Paraneoplastic Neurological Syndromes and Glutamic Acid
McKeon, A. and J.A. Tracy, GAD65 neurological aut oimmunity. Muscle Nerve, 2017.
pr
102.
oo
f
Decarboxylase Antibodies. JAMA Neurol, 2015. 72(8): p. 874-81.
103.
e-
56(1): p. 15-27.
Olberg, H.K., G.E. Eide, and C.A. Vedeler, Can serum GAD65 antibody levels predict
Pr
neurological disease or cancer? J Neuroimmunol, 2019. 336: p. 577025. Serafini, A., et al., Paraneoplastic epilepsy. Epilepsy Behav, 2016. 61: p. 51-58.
105.
Kannoth, S., Paraneoplastic neurologic syndrome: A practical approach. Ann Indian
rn
al
104.
106.
Jo u
Acad Neurol, 2012. 15(1): p. 6-12. Pittock, S.J., T.J. Kryzer, and V.A. Lennon, Paraneoplastic antibodies coexist and
predict cancer, not neurological syndrome. Ann Neurol, 2004. 56(5): p. 715-9. 107.
Makela, K.M., et al., Clinical Management of Epilepsy With Glutamic Acid
Decarboxylase Antibody Positivity: The Interplay Between Immunot herapy and Anti-epileptic Drugs. Front Neurol, 2018. 9: p. 579. 108.
Liimatainen, S., et al., Immunological perspectives of temporal lobe seizures. J Neuroimmunol, 2013. 263(1-2): p. 1-7.
109.
Gillinder, L., et al., Refractory epilepsy secondary to anti-GAD encephalitis treat ed
Journal Pre-proof with DBS post SEEG evaluation: a novel case report based on stimulation findings. Epileptic Disord, 2018. 20(5): p. 451-456. 110.
Falip, M., et al., Prevalence and immunological spectrum of temporal lobe epileps y
with glutamic acid decarboxylase antibodies. Eur J Neurol, 2012. 19(6): p. 827-33. 111.
Husari, K.S. and D. Dubey, Autoimmune Epilepsy. Neurotherapeutics, 2019. 16(3): p. 685-702.
f
Dogan Onugoren, M., et al., Immunoadsorption therapy in autoimmune encephalitides.
oo
112.
Farooqi, M.S., et al., Therapeutic plasma exchange and immunosuppressive therapy
e-
113.
pr
Neurol Neuroimmunol Neuroinflamm, 2016. 3(2): p. e207.
in a patient with anti-GAD antibody -related epilepsy: quantification of the antibody
Mazzi, G., et al., Plasma exchange for anti GAD associated non paraneoplastic limbic
al
114.
Pr
response. J Clin Apher, 2015. 30(1): p. 8-14.
Kanter, I. C., et al., Cyclophosphamide for anti-GAD antibody-positive refractory status
Jo u
115.
rn
encephalitis. Transfus Apher Sci, 2008. 39(3): p. 229-33.
epilepticus. Epilepsia, 2008. 49(5): p. 914-20. 116.
Saidha, S., et al., Treatment of anti-glutamic acid decarboxylase antibody-associated
limbic encephalitis with mycophenolate mofetil. J Neurol, 2010. 257(6): p. 1035-8. 117.
Korff, C.M., et al., Encephalitis associated with glutamic acid decarboxylase
autoantibodies in a child: a treatable condition? Arch Neurol, 2011. 68(8): p. 1065-8. 118.
Millet, C., V. van Pesch, and C.J. Sindic, Idiopat hic limbic encephalitis associated with
antibodies to glutamic acid decarboxylase. Acta Neurol Belg, 2015. 115(2): p. 165-7. 119.
Grilo, E., et al., Type 1 diabetes and GAD65 limbic encephalitis: a case report of a
Journal Pre-proof 10-year-old girl. J Pediatr Endocrinol Metab, 2016. 29(8): p. 985-90. 120.
Di Giacomo, R., et al., Predictive value of high titer of GAD65 antibodies in a case of
limbic encephalitis. J Neuroimmunol, 2019. 337: p. 577063. 121.
Kwan, P., et al., Glutamic acid decarboxylase autoantibodies in controlled and
uncontrolled epilepsy: a pilot study. Epilepsy Res, 2000. 42(2-3): p. 191-5. Cabezudo-Garcia, P., et al., Efficacy of antiepileptic drugs in autoimmune epilepsy: A
Carreno, M., et al., Epilepsy surgery in drug resistant temporal lobe epilepsy
pr
123.
oo
systematic review. Seizure, 2018. 59: p. 72-76.
f
122.
N.,
et
Pre-
al.,
hippocampus-associated
memory
and
long-term
impairment
Pr
Hans en,
postoperative in
epilepsy
courses patients
of with
al
antibody-associated limbic encephalitis and selective amygdalohippocampectomy.
rn
Epilepsy Behav, 2018. 79: p. 93-99.
Jo u
124.
e-
associated with neuronal antibodies. Epilepsy Res, 2017. 129: p. 101-105.
pr
oo
f
Journal Pre-proof
comprise
three
domains:
N-terminal,
Pr
GAD67,
e-
Fig. 1. A) Each GAD monomeric unit , GAD65 and
PLP-binding, and C-terminal domain. 74% identity for the
al
middle and C-terminal domains, 25% identity for the
rn
N-terminal domain. B ) Dimeric structure ofGAD65
Jo u
showing the structural locations of the three domains, colored as in Fig. 1(A) Note that ctc1 includes prototypically mAb b78 and some residues from the N-terminal domain, ctc2 includes prototypically mAb 96.11 and some residues from the PLP-domain. (Figure.1. from
Stiff-person
syndrome
(SPS) and
anti-GAD-related CNS degenerations: Protean additions to the autoimmune central neuropathies. Journal of Autoimmunity. 37 (2011) 79-87. copyright 2011; Elsevier. This material is reproduced with permission from Elsevier)
Jo u
rn
al
Pr
e-
pr
oo
f
Journal Pre-proof
Journal Pre-proof
A
B
C
e-
pr
oo
f
D
Pr
Fig. 2. Brain MRI of GAD65-epilepsy. Patient 1. A 61-year-old man experienced left lower
al
limb paroxysmal convulsion and numbness, associated with brief heart palpitations, tongue
rn
stiffness, and sweats several times per day, for two months. Brain MRI: FLAIR sequen ce demonstrating bilateral medial temporal hyperintensities on axial (A) and sagittal (B) sections. Patient 2. A 23-year-old woman presented with left face and tongue twitching,
Jo u
therapies
behavioral changes for 20 days, and progressed to confusion and bilateral tonic-clonic seizures 3 days. FLAIR sequence(C) and DWI image (D) demonstrating right temporoparietal lobe slightly hyperintensities on axial.
Table 1. Review of recommending dose, mechanism of action, adverse effects of various immune therapies: First-line and Second-line Recommended dose Fi rst-line therapies
Mechanism of action
Adverse effects
Journal Pre-proof
dose:
IV.
Modulates
methylprednisolone
and
(MP)
production,
pulses.
1000
on
3–5
mg/day
consecutive days ↓ 1000
mg/day
on
consecutive
chemokine cytokine adhesion
molecule
expression
Insomnia,
psychiatric
symptoms, hyperglycemia, hypertension
obesity,
electrolyte
imbalances,
and reduces migration
hypertension, peptic ulcer,
of leukocytes to the
Cushing
target tissue
cataracts,
syndrome,
3
f
Initial
oo
Corticosteroids
days
infection
osteoporosis,
once
weekly for 5 weeks or mg/day
consecutive
days by
3 ,
once
al
followed
on
necrosis
Pr
1000
pr
by
avascular
e-
followed
and
rn
weekly for 5 weeks,
Jo u
then every 2 weeks for 6 weeks.
Oral.
60–80
prednisone duration
↓
of
mg daily,
tapering
dose IVIG
Initial
dose:
0.4/kg
body-weight/day on 3–5
Modulates complement
Anaphylactic
activation,
(IgA-deficient patients) acute
suppress
reactions
Journal Pre-proof
consecutive days
idiotypic
↓
antibodies,
renal failure, thromboembolic
saturate Fc receptors
events, hemolytic anemia,
on macrophages, and
aseptic meningitis,headache
0.4/kg body-weight/day on 3 consecutive days suppress followed
by
cytokines,
once chemokines,
and
weekly for 5 weeks or metalloproteinases
oo
f
0.4/kg body-weight/day on 3 consecutive days once
weekly for 5 weeks,
Pr
then every 2 weeks for 6 weeks.
An extracorporeal blood
Hypotension and electrolyte
day spread over 5–7
purification technique,
imbalance,
cycles,10–14 days
removes
hemorrhage,
al
1 exchange every other
Jo u
rn
Plasmapheresis
pr
by
e-
followed
large
molecular-weight molecules,
such
as
complex,
complements and filters out plasma membrane proteins
thrombosis,
and pneumothorax
immunoglobulins, immune
infection,
Journal Pre-proof
Se cond-line therapies 500–800
mg/m2/
An
alkylating
interferes with
symptoms,
which
Oral: 1–2 mg/kg/day
DNA synthesis, causing
failure,
neutropenia,
cell death by DNA–RNA
infections,
hemorrhagic
cross-linking
cystitis
and
of
alopecia, mucositis, ovarian
protein
IV: monthly single day
A humanized anti–α4
Progressive
infusions of 300 mg
e-
pr
synthesis
leukoencephalopathy (PML),
integrin
monoclonal
gastrointestinal
entry of T and B cells
allergic
al
Pr
antibody that prevents
rn
into the CNS.
multifocal
symptoms,
reaction,
liver
problems, headache, tired feeling, cold symptoms, joint
Jo u Rituximab
Gastrointestinal
month for 3–6 months
inhibition
Natalizumab
agent
f
de
IV:
oo
Cyclophosphami
pain,
vaginal
itching
or
discharge
IV: 1000 mg followed
A
humanized
Allergic reaction, infections,
by once weekly for 2
monoclonal IgG against
reactivation of tuberculosis
weeks, or 375 mg/m 2
CD20-positive B cells
infection,
weekly for 4 weeks.
which leads to B cell
infection,
depletion and reduces
neutropenia.
autoantibody
or
hepatitis
B
late-onset
Journal Pre-proof
generation Mycophenolate
Oral: Initially 500 mg
Inhibition
of
mofetil
twice daily, target 1000
monophosphate
hypertension,
mg twice daily.
dehydrogenase
hypercholesterolemia,
de
novo
Gastrointestinal
symptoms,
hepatotoxicity,
peripheral
synthesis of guanosine
edema,
infections,
nucleotides, leading to
myelosuppression,
selective inhibition of
lymphoma,
pr
oo
f
mediated
inosine
other
malignancies
e-
lymphocyte
and
Oral:
Initially
1.5
rn
mg/kg/ day
Converted to cytotoxic
Gastrointestinal
symptoms,
6-thioguanine
hypersensitivity
reactions,
al
mg/kg/day, target 2.5–3
Jo u
Azathioprine
Pr
proliferation.
nucleotides
which
intercalates
into
lymphopenia, hepatotoxicity,
and
opportunistic infection.
replicating
DNA
can block the de novo pathway
of
purine
synthesis lymphocyte hair
loss,
inducing apoptosis cytopenia,
hepatotoxicity, lymphoma,
and
alopecia,
fatigue,
Journal Pre-proof
infections Basiliximab
IV :20 mg/month
An
interleukin-2
Allergic
receptor Ab that inhibits
lymphopenia,
T
infection
cell-mediated
reaction, opportunistic
immune reactions.
oo
Anti-GAD65 Abs were found in patients with seizures in the context of encephalitis,
pr
f
Highlights:
e-
and patients with chronic epileptic syndromes.
The pathogenic role of anti-GAD65 Abs remains unclear
Immune-mediated epilepsy with GAD65 antibodies frequently coexist multiple
Pr
al
autoimmune disorders or auto-antibodies and may evolve into a more widespread
The evidence of high serum titers and intrathecal synthesis play a fundamental role in
Jo u
rn
CNS inflammatory disorder.
diagnosis but poorly correlate with disease severity or response to therapies.
Early immunosuppressive therapy can achieve temporary success, more aggressive and long-term immunosuppressive therapies may be needed to prevent disease progression and relapse.
Table 1. Review of recommending dose, mechanism of action, adverse effects of various immune therapies: First -line and Second-line therapies