ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Autoimmune (including paraneoplastic) encephalitis: Clinical features and diagnosis

Autoimmune (including paraneoplastic) encephalitis: Clinical features and diagnosis
Literature review current through: Jan 2024.
This topic last updated: Dec 11, 2023.

INTRODUCTION — Encephalitis is an inflammatory condition of the brain with many etiologies. There are several types of encephalitis that are immune mediated (autoimmune), including the encephalitis syndromes associated with antibodies against neuronal cell-surface/synaptic proteins and those associated with antibodies against intracellular neuronal proteins.

Although both categories of autoimmune encephalitis can occur as paraneoplastic manifestations of an underlying cancer, the frequency of cancer association varies depending on the autoantibodies (table 1). Most neuronal antibodies typically associated with paraneoplastic encephalitis (eg, >70 percent of cases occur with a cancer association) are directed against intracellular neuronal proteins (onconeuronal proteins).

Thus, many autoimmune encephalitides associated with antibodies against neuronal cell-surface proteins can occur in the presence or absence of cancer, and a few encephalitides associated with antibodies against intracellular neuronal proteins (eg, glutamic-acid decarboxylase 65 kilodalton isoform [GAD65] or adenylate kinase 5 [AK5]) rarely or almost never occur with cancer.

Clinical features, diagnosis, and natural history of autoimmune encephalitis, including paraneoplastic encephalitis, will be reviewed here. Initial and long-term management are reviewed separately. (See "Autoimmune (including paraneoplastic) encephalitis: Management".)

An overview of paraneoplastic syndromes and other paraneoplastic disorders are discussed separately. (See "Overview of paraneoplastic syndromes of the nervous system" and "Paraneoplastic syndromes affecting spinal cord, peripheral nerve, and muscle" and "Paraneoplastic cerebellar degeneration" and "Opsoclonus-myoclonus-ataxia syndrome".)

Other inflammatory disorders of the central nervous system (CNS) covered in separate topics include:

Acute disseminated encephalomyelitis (ADEM) (see "Acute disseminated encephalomyelitis (ADEM) in children: Pathogenesis, clinical features, and diagnosis" and "Acute disseminated encephalomyelitis (ADEM) in children: Treatment and prognosis" and "Acute disseminated encephalomyelitis (ADEM) in adults")

Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) (see "Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD): Clinical features and diagnosis" and "Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD): Treatment and prognosis")

Neuromyelitis optica spectrum disorders (NMOSD) (see "Neuromyelitis optica spectrum disorder (NMOSD): Clinical features and diagnosis" and "Neuromyelitis optica spectrum disorder (NMOSD): Treatment and prognosis")

Hashimoto encephalopathy/steroid-responsive encephalopathy associated with autoimmune thyroiditis (SREAT) (see "Hashimoto encephalopathy")

Chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS) (see "Evaluation and diagnosis of multiple sclerosis in adults", section on 'CLIPPERS')

NEURONAL CELL-SURFACE PROTEIN ANTIBODY SYNDROMES — These syndromes occur in patients of all ages, some with a preferential age distribution, and may or may be associated with cancer. For many syndromes, the antibodies have been shown to be pathogenic. (See "Overview of paraneoplastic syndromes of the nervous system", section on 'Pathogenic effects of antibodies'.)

Anti-NMDA receptor encephalitis

Clinical features — Anti-N-methyl-D-aspartate (NMDA) receptor encephalitis is a disorder that usually affects young adults and children, with a female predominance in young adults and a less strong female predominance in children and patients older than 45 years of age. The disorder is associated with a predictable set of symptoms that combine to make up a characteristic syndrome [1-5].

Many patients present with prodromal headache, fever, or a viral-like process, followed in a few days by a multistage progression of symptoms that include:

Prominent psychiatric manifestations (anxiety, agitation, bizarre behavior, hallucinations, delusions, disorganized thinking, psychosis). In very rare instances the disease can be monosymptomatic (eg, manifesting as isolated psychosis), or psychiatric symptoms can be the only manifestation of a recurrence [6,7].

Sleep disorders, including sleep reduction at disease onset and hypersomnia during recovery [8].

Memory deficits.

Seizures.

Decreased level of consciousness, stupor with catatonic features.

Frequent dyskinesias: orofacial, choreoathetoid movements, dystonia, rigidity, opisthotonic postures.

Autonomic instability: hyperthermia, fluctuations of blood pressure, tachycardia, bradycardia, cardiac pauses, and sometimes hypoventilation requiring mechanical ventilation.

Language dysfunction: diminished language output, mutism, echolalia.

Children as young as eight months have been reported with this syndrome [3,9-12]; in the authors' experience, children as young as two months may be affected. In children, the symptoms are similar to those of the adults, with prominent early behavioral or psychiatric symptoms in most patients; dysautonomia and hypoventilation are less frequent and severe. Presenting symptoms usually include acute behavioral change, seizures, language dysfunction, and prominent dyskinesias, including dystonia and chorea [13-15]. Although rare, approximately 5 percent of patients are >45 years of age. In this group, the disease is less severe but outcomes tend to be worse, possibly due to delay in diagnosis and treatment [16].

Diagnosis and differential diagnosis — The disorder should be suspected in adults or children that develop the above clinical symptoms, usually accompanied by:

Cerebrospinal fluid (CSF) lymphocytic pleocytosis or oligoclonal bands (although basic CSF parameters can be normal initially).

Electroencephalography (EEG) with infrequent epileptic activity but frequent slow, disorganized activity that does not correlate with most abnormal movements. An infrequent (<30 percent of cases; less frequent in children) unique electrographic pattern called extreme delta brush can assist in the diagnosis and is associated with a more prolonged illness [17].

Brain magnetic resonance imaging (MRI) that is often normal or shows transient fluid-attenuated inversion recovery (FLAIR) or contrast-enhancing abnormalities in cortical (brain, cerebellum) or subcortical (hippocampus, basal ganglia, white matter) regions [2,18]. While not routinely performed, positron emission tomography (PET) reportedly shows a characteristic increase in the frontal-occipital gradient of cerebral glucose metabolism, which correlates with disease severity [19].

The diagnosis of anti-NMDA receptor encephalitis is confirmed by the detection of immune globulin G (IgG) antibodies against the GluN1 (also known as NR1) subunit of the NMDA receptor (table 2) [20]. Initial antibody studies should be done in CSF since CSF antibodies are always present at the time of presentation; most patients have intrathecal synthesis of antibodies [21]. False-positive and -negative results may occur when testing only serum [22]. However, results of antibody testing can be delayed.

A set of criteria based on the clinical examination and commonly available diagnostic tests (eg, MRI, EEG, and routine CSF studies) has been proposed to support the diagnosis of probable anti-NMDA encephalitis [23]. Patients meeting these criteria should initiate treatment. The diagnosis and treatment approach can then be re-evaluated when antibody testing results are available. A critical caveat to the use of these criteria is that they are not applicable to patients in the very early stages of the disease when they may have few symptoms or an isolated symptom (eg, only seizures), as the differential diagnosis is too broad.

Immune globulin M (IgM) and immune globulin A (IgA) antibodies against the NMDA receptor, which have been described in some patients with chronic schizophrenia and many other chronic neurologic or psychiatric disorders, are nonspecific and have no additional value in the diagnosis of NMDA receptor encephalitis [24,25].

After treatment or in advanced stages of the disease, the CSF antibodies usually remain elevated if there is no clinical improvement, while serum antibodies may be substantially decreased by treatments [18,26,27]. The titer of CSF antibodies appears to correlate more closely with the clinical outcome than serum titers [2,18,22,26].

The differential diagnosis includes primary psychiatric disorders (acute psychosis or schizophrenia), malignant catatonia, neuroleptic malignant syndrome [28], viral encephalitis [29], and encephalitis lethargica [30], among others [31]. There have been several case reports of anti-NMDA receptor encephalitis in patients with current or recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [32-34]; the pathogenic link between both diseases is unclear. (See "COVID-19: Neurologic complications and management of neurologic conditions", section on 'Other acute neurologic manifestations'.)

Association with ovarian teratoma and other tumors — The detection of an ovarian teratoma is age dependent; approximately 45 percent of female patients older than 18 years have uni- or bilateral ovarian teratomas, while less than 9 percent of females younger than 14 years have a teratoma [5]. A review of 400 cases showed that African American patients have a mild predominance to have ovarian teratomas compared with patients from other ethnic groups [4]. Ovarian teratomas are often revealed by MRI and computed tomography (CT) of the abdomen and pelvis, along with abdominal or transvaginal ultrasound (table 3) [18]. (See "Ovarian germ cell tumors: Pathology, epidemiology, clinical manifestations, and diagnosis", section on 'Teratomas'.)

In male patients, the detection of a tumor is rare. Cases with associated tumors other than ovarian teratoma include testicular germ cell tumor [35], teratoma of the mediastinum, small cell lung cancer (SCLC) [2], Hodgkin lymphoma [36], ovarian cystadenofibroma [37], and neuroblastoma [38]. The frequency of underlying tumors in older patients (>45 years) is low, and when present, tumors are more often carcinomas instead of teratomas [16]. Anti-NMDA receptor encephalitis has been described in a few patients with metastatic cancer treated with checkpoint inhibitor immunotherapy [39]. (See "Toxicities associated with immune checkpoint inhibitors".)

Association with HSVE — Although preceding infections have been suspected to play a role in triggering autoimmune encephalitis, to date this has only been demonstrated for herpes simplex viral encephalitis (HSVE). Studies have shown that approximately 25 percent of patients who are NMDA receptor antibody-negative in serum and CSF at the time of HSVE infection seroconvert to positive NMDA receptor antibodies (or less commonly other neuronal antibodies) in the setting of relapsing symptoms not attributable to HSVE relapse [40-44]. A smaller proportion develops NMDA receptor or other antibodies in the absence of clinical symptoms [43].

Symptoms of anti-NMDA receptor encephalitis in these cases begin at a median of four to six weeks after initial viral infection and may occur in contiguity with or after recovery from the HSVE [41,43-48]. In a series of 58 patients with antibody-confirmed autoimmune encephalitis after HSVE (74 percent with NMDA receptor antibodies), the most common symptoms were change of behavior (93 percent), decreased level of consciousness (57 percent), choreoathetosis (47 percent, all in children four years of age or younger), seizures (38 percent), and dysautonomia (27 percent) [43]. In most pediatric cases, symptoms have included choreoathetosis and/or orofacial dyskinesias [41]; teenagers and young adults are more likely to develop behavioral and psychiatric symptoms [42]. Prompt diagnosis and treatment with immunomodulatory therapy improve symptoms and outcome despite persistence of deficits from the HSVE, especially in older children and adults [41,43].

In addition to NMDA receptor antibodies, antibodies to gamma-aminobutyric acid A (GABA-A), dopamine 2 receptor, and unknown neuronal cell-surface antigens have been reported in patients with autoimmune encephalitis after HSVE [42,43].

Treatment — Treatment of anti-NMDA receptor encephalitis consists of immunomodulatory therapy and tumor resection, when appropriate (algorithm 1). Progressive neurologic deterioration and death can occur without treatment, and early initiation of immunomodulatory therapy is associated with improved outcomes. Treatment is reviewed in detail separately. (See "Autoimmune (including paraneoplastic) encephalitis: Management".)

Long-term outcomes — After recovery of the acute phase, when patients are able to return home, they continue to have residual cognitive, behavioral and psychiatric alterations that can last more than one year [49]. In a study that included 28 patients with anti-NMDA receptor encephalitis, 89 percent of patients at visit 1 (median 4 months from disease onset) had deficits in at least one cognitive domain (most commonly executive function), including 54 percent with deficits in one to two domains, 18 percent with deficits in three to four domains, and 18 percent with deficits in five to six domains [50]. Only 11 percent of patients had normal cognitive function. Features of this postacute stage resemble the alterations found in patients with stable symptoms of schizophrenia spectrum disorders.

The strongest risk factors for poor one-year functional outcomes are lack of clinical improvement within the first four weeks of treatment, requirement for intensive care unit admission, treatment delay >4 weeks, abnormal MRI, and CSF white blood cell count >20 cells/microL [51]. Additional risk factors include extremes of age (≤2 or ≥65 years of age) and extreme delta brush pattern on EEG [52]. In children, an abnormal brain MRI and sensorimotor deficits at presentation have also been associated with worse outcomes [53].

Patients with anti-NMDA receptor encephalitis are at risk for relapse. Relapse occurs in 15 to 24 percent of patients, sometimes after several years [2,18,54]. Relapse may occur in the absence of a tumor or in association with an occult or recurrent teratoma. In several series, relapses were more common among those who did not receive immunomodulatory therapy with the initial presentation [5,54]. Adolescent presentation has also been associated with increased risk for relapse [52]. Relapses are typically treated similarly to the approach in newly diagnosed patients, with a lower threshold to initiate second-line therapies early in the course of the relapse. (See "Autoimmune (including paraneoplastic) encephalitis: Management", section on 'Relapsed disease'.)

Pregnancy and fetal effects — Transplacental transfer of IgG anti-NMDA receptor antibodies has been documented in serum of babies born to mothers with anti-NMDA receptor encephalitis. However, newborn outcomes appear to be good in most cases.

In a review of 21 previously published cases and 11 new cases of females who either developed anti-NMDA receptor encephalitis while pregnant or became pregnant while recovering from the encephalitis, the majority of newborns were healthy and had no evidence of neurodevelopmental delay with follow-up ranging from 6 to 36 months [55-61]. Transient respiratory distress or neuromuscular deficits at delivery rarely occurred. Such complications spontaneously recovered and were considered secondary to antiseizure and sedative medications given to the mothers. Adverse outcomes included one infant with cortical dysplasia, developmental disorder, and seizures who was the result of a complicated pregnancy with uteroplacental insufficiency [56]. There was one neonatal death reported [55]. In this case, the mother had recovered from anti-NMDA receptor encephalitis 18 months before delivery and presented with eclampsia without prior knowledge of the pregnancy or prenatal care.

Obstetric complications were reported in approximately one-third of the females who developed anti-NMDA receptor encephalitis during pregnancy, mostly due to pathologic pregnancy or spontaneous miscarriage, supporting close monitoring in intensive care units focused on high-risk pregnancies [61].

Anti-LGI1 encephalitis — Patients with anti-leucine-rich glioma inactivated 1 (LGI1) encephalitis develop memory disturbances, confusion, and seizures [62-65]. Memory and cognitive deficits may be preceded by short faciobrachial dystonic seizures that can be mistaken for myoclonus or dystonia and are often poorly responsive to antiseizure medication therapy. Patients may develop hyponatremia and rapid eye movement (REM) sleep behavior disorder. Some patients present with rapidly progressive cognitive decline without signs of encephalitis in CSF or on MRI [66].

MRI usually shows findings typical of limbic encephalitis (eg, medial temporal lobe hyperintensity) (image 1) [67]. CSF may show pleocytosis but is often normal or only shows oligoclonal bands [68]. Approximately 5 percent of cases are associated with tumors; the most common associated tumor is thymoma. The association with other tumors may be coincidental [66].

The associated antibodies target the LGI1 protein, a secreted neuronal protein that functions as a ligand for two epilepsy-related proteins, ADAM22 and ADAM23 [69]. The binding of the antibodies to LGI1 disrupts pre- and postsynaptic LGI1 signaling, resulting in neuronal hyperexcitability [70].

Treatment with immunomodulatory therapy results in substantial clinical improvement in 70 to 80 percent of patients [62,71-74]. Observational data suggest that early initiation of immunomodulatory therapy in patients with faciobrachial dystonic seizures may prevent the development of cognitive impairment and improve long-term outcomes [64]. (See "Autoimmune (including paraneoplastic) encephalitis: Management".)

Relapses occur in up to one-third of patients, often but not exclusively during the first six months of the disease, and are associated with worse outcome. In one study, the median time to relapse after initial presentation was 35 months [75]. Despite substantial recovery, cognitive deficits and disability persist in many patients, along with evidence of hippocampal atrophy on MRI [66,76]. The risk of long-term epilepsy appears to be low in the absence of disease relapse [77,78].

Anti-Caspr2-associated encephalitis — Anti-contactin-associated protein-like 2 (Caspr2)-associated encephalitis can manifest as limbic encephalitis, as Morvan syndrome (neuromyotonia, memory loss and confusion, sleep disturbances, autonomic instability), and in a small number of patients with isolated neuromyotonia [68,79-81]. The target antigen is Caspr2, which plays a role in maintaining the normal function of voltage-gated potassium channels (VGKC) [79]. Caspr2 autoantibodies inhibit cell adhesion interactions between Caspr2 and contactin-2, reduce levels of Caspr2 and related proteins, and cause reversible memory impairment in mice [82,83].

This disorder predominantly affects older males with a median age of 65 years, although rare pediatric cases have been described in children as young as two years old [65,81,84,85]. Most patients do not have cancer. Patients with a tumor (usually thymoma) are more likely to develop Morvan syndrome than isolated central or peripheral symptoms.

Although the clinical syndrome is varied, almost 80 percent of patients develop three or more of the following core symptoms: cognitive changes, cerebellar symptoms, peripheral nerve hyperexcitability, autonomic dysfunction, insomnia, neuropathic pain, and weight loss [84]. Seizures occur in approximately one-third of patients, mostly focal nonmotor seizures with impaired awareness [86]. Approximately one-third of patients have one or more movement disorders, including ataxia, myoclonus, and tremor [87]. Rare but unusual manifestations include continuous segmental spinal myoclonus, orthostatic myoclonus of the legs, and paroxysmal cerebellar ataxia [87,88].

The presentation and disease course are slower than those of other autoimmune encephalitis syndromes. In one study, the median time to disease nadir was four months, but in 30 percent of cases it was >12 months. Most patients have responses to immunomodulatory therapy, and approximately 25 percent will have relapses.

Anti-AMPA receptor encephalitis — Encephalitis associated with antibodies against the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor affects predominantly females, with a median age of onset of 50 to 60 years [89-93]. In a case series of 22 patients with anti-AMPA receptor antibodies, limbic encephalitis with or without seizures was the most common clinical presentation (55 percent); other presentations included limbic dysfunction along with multifocal or diffuse encephalopathy (36 percent), motor deficits followed by limbic encephalitis (one patient), and psychosis with bipolar features (one patient) [92].

An underlying neoplasm is identified in approximately two-thirds of the patients, most commonly lung, thymus, or breast [89,92,94]. A lymphocytic pleocytosis in the CSF has been reported in 50 to 90 percent of patients (range 5 to 164 cells) [89,92]. MRI is usually abnormal, with FLAIR signal abnormality in the medial temporal lobes.

In one series, 9 of 10 patients responded to treatment of the underlying neoplasm and/or immunomodulatory therapy, but subsequent relapses occurred in five (in the absence of tumor recurrence) and were associated with an incomplete treatment response and death from status epilepticus in one patient [89]. In another series, 15 of 21 patients had a full or partial response to treatment of the underlying neoplasm and/or immunomodulatory therapy [92].

The binding of patients' antibodies to AMPA receptors causes a reversible decrease of these receptors. In an animal model of passive transfer of patients' antibodies, these caused a reorganization of AMPA receptor subunits and memory dysfunction [95,96].

Anti-GABA-A receptor encephalitis — Patients with encephalitis due to antibodies against the GABA-A receptor develop a rapidly progressive encephalitis with refractory seizures, status epilepticus, and/or epilepsia partialis continua [97]. Nearly half of reported cases are children [98]. In these patients, the development of dyskinesias can suggest the diagnosis of anti-NMDA receptor encephalitis.

The CSF often shows lymphocytic pleocytosis with increased protein concentration. Unlike other causes of autoimmune encephalitis, in which the MRI is normal or shows abnormalities limited to the limbic system, the MRI in anti-GABA-A receptor encephalitis often shows multifocal cortical/subcortical and widespread FLAIR and T2 signal abnormalities [98]. Tumors (mostly thymoma) occur in 40 percent of patients, almost all adults; isolated cases with other tumors have been reported [97,98]. In children, anti-GABA-A receptor encephalitis may develop as a postviral encephalitis and coexist with NMDA receptor antibodies. (See 'Association with HSVE' above.)

Patients respond to immunomodulatory therapy but often require pharmacologic-induced coma for prolonged seizures. (See "Autoimmune (including paraneoplastic) encephalitis: Management", section on 'Intensive care management'.)

Anti-GABA-B receptor encephalitis — Encephalitis due to antibodies against the B1 subunit of the gamma-aminobutyric acid B (GABA-B) receptor has been described primarily in adults presenting with limbic encephalitis, although presentations may vary and include limbic encephalitis with seizures, rapidly progressive dementia, status epilepticus, ataxia, or opsoclonus-myoclonus-ataxia syndrome (OMAS) [99,100]. In one report, anti-GABA-B antibodies were the most common antibodies found in limbic encephalitis syndromes associated with SCLC that were previously considered seronegative [101]. One pediatric case was characterized by encephalopathy, refractory seizures, and a mixed movement disorder (opsoclonus, ataxia, and chorea) [102].

Approximately 50 percent of cases are paraneoplastic and are almost always associated with SCLC [99,103]. Co-occurring antibodies against potassium channel tetramerization domain-containing 16 (KCTD16) increase the likelihood of a paraneoplastic etiology [100]. MRI shows FLAIR and T2 signal abnormalities consistent with limbic encephalitis in approximately one-half of patients, and over one-half of patients have CSF pleocytosis and/or elevated protein levels [99].

Most patients have substantial improvement with treatment (ie, immunomodulatory therapy and tumor treatment when indicated). However, the outcome is dictated by the presence of a tumor [99,104].

Anti-IgLON5 disease — Patients with antibodies against IgLON family member 5 (IgLON5), a neuronal cell adhesion protein, manifest a neurologic disease with shared features of autoimmunity and neurodegeneration. Although REM and non-REM sleep parasomnias were highlighted in early case descriptions, subsequent studies have shown that abnormal movements and gait and balance problems are usually present in addition to sleep problems [105-109].

In a series of 72 patients with anti-IgLON5 disease, the median age at symptom onset was 62 years (range 42 to 91 years) [109]. In approximately three-quarters of patients, symptoms progressed over months to years leading up to diagnosis. In the remaining quarter, the presentation was more rapid, with progression over weeks to a few months.

Five main clinical syndromes have been described at diagnosis: a sleep disorder with REM and non-REM parasomnias and sleep-disordered breathing; movement disorders, which in some cases resemble progressive supranuclear palsy (PSP); a bulbar syndrome including dysarthria, dysphagia, vocal cord paralysis (stridor), or acute respiratory insufficiency; cognitive decline with or without chorea; and neuromuscular manifestations with muscle weakness, atrophy, or fasciculations [108-110]. Almost all patients eventually develop both abnormal movements and sleep problems, such as parasomnias, sleep apnea, insomnia, or excessive daytime sleepiness.

Video-polysomnography is essential to define the complex sleep disorder and identify characteristic features of the disorder [111] (see "Polysomnography in the evaluation of parasomnias and epilepsy"). CSF and imaging studies tend to be normal aside from the presence of anti-IgLON5 antibodies in CSF and serum. There appears to be a strong association with the human leucocyte antigen (HLA) DRB1*10:01 allele [108]. Prognosis is poor, with few patients responding to immunomodulatory therapy and a high mortality rate [108,109]. Early initiation of immunomodulatory therapy prior to advanced symptoms may be associated with disease stabilization and better outcomes [110].

Neuropathologic evaluation of some patients at autopsy revealed neuronal loss and extensive deposits of hyperphosphorylated tau, mainly involving the brainstem tegmentum and hypothalamus. Such deposits have also been visualized in patients using tau-specific PET tracers [112]. Neuropathologic criteria of anti-IgLON5-related tauopathy have been proposed [113].

Anti-DPPX encephalitis — Patients with antibodies against dipeptidyl-peptidase-like protein-6 (DPPX) develop severe prodromal symptoms of loss of weight, diarrhea, or other gastrointestinal symptoms followed within a few months (average four months) by the development of encephalitis with central hyperexcitability such as hyperekplexia, agitation, myoclonus, tremor, and seizures [114,115]. Some patients have symptoms suggestive of progressive encephalomyelitis with rigidity and myoclonus (PERM) [115,116]. Most patients have a protracted course, although subacute presentations have been reported [114,117]. Tumors are rare, and when they do occur, most are B cell neoplasms [117].

Other common features included CSF lymphocytic pleocytosis, but studies may be normal; the MRI is usually not specific. Patients usually have an initial benefit from immunomodulatory therapy but often relapse when therapy is tapered.

Anti-GlyR encephalopathy — Antibodies to the alpha-1 subunit of the glycine receptor (GlyR) have been associated with a syndrome of PERM, acquired hyperekplexia, and stiff-person syndrome in some patients without glutamic acid decarboxylase (GAD) antibodies [118-121]. A few patients have been reported with limbic or other encephalitis without brainstem or spinal cord features. In a series of 45 patients, five had a past history and successful treatment of a tumor, and in four patients, the tumor diagnosis was concurrent with the neurologic disease [122]. Most patients have responses to immunomodulatory therapy. Antibodies to alpha-1 GlyR have also been reported in serum (and at low titers) of some patients with isolated seizures, optic neuritis, multiple sclerosis, OMAS, and cerebellar ataxia without stiff-person syndrome or PERM [123-126].

Anti-mGluR5 encephalitis — Antibodies against the metabotropic glutamate receptor 5 (mGluR 5) have been associated with encephalitis, often including movement disorders, sleep dysfunction, and seizures [127-130]. The clinical spectrum includes prominent neuropsychiatric features (eg, progressive mood and personality changes, anterograde amnesia, disorientation), prosopagnosia, headaches, involuntary movements, and seizures. The CSF often shows pleocytosis, and in approximately half of the cases, the MRI shows FLAIR abnormalities in limbic or extralimbic regions [130].

The syndrome is most commonly associated with Hodgkin lymphoma (Ophelia syndrome) or SCLC but can also occur in the absence of a tumor. The disorder is highly responsive to immunomodulatory therapy and treatment of the tumor, but relapses can occur.

Anti-mGluR1 encephalitis — Patients with antibodies against the mGluR1 develop cerebellar ataxia that remains isolated in less than 15 percent of patients [131]. In the majority, the ataxia is accompanied by cognitive changes, seizures, or psychiatric symptoms [131-133]. Both sexes are affected, with a slight predominance of males and a median age of 55 years [131].

The CSF is abnormal is approximately 75 percent of patients, including pleocytosis, oligoclonal bands, or increased IgG index. Brain MRI is normal in most patients but can show cerebellar T2 hyperintensities or, less frequently, leptomeningeal enhancement. Cerebellar atrophy may develop over time.

The disorder is usually not associated with cancer; reported associations have involved a history of Hodgkin lymphoma or other hematologic malignancy and one patient with a concurrent cutaneous T cell lymphoma. The disorder can improve with early immunomodulatory therapy. Relapses can occur, usually in the context of immunomodulatory therapy discontinuation.

Anti-neurexin-3 alpha encephalitis — Antibodies to neurexin-3 alpha have been described in a series of five patients who developed a severe encephalitis with rapid decline in consciousness, orofacial dyskinesias, and central hypoventilation that in some patients resembled anti-NMDA receptor encephalitis [134]; however, NMDA receptor antibodies were absent in all cases. Patients were relatively young (median age 44 years) and all had prodromal symptoms before the rapid decline. The CSF was abnormal in all (four with pleocytosis and one with elevated IgG index); brain MRI was normal in four and showed FLAIR abnormalities in the temporal lobes in one. All five patients received immunomodulatory therapy, and three had partial recovery. One patient died from the disease and the other from sepsis. Four of the patients had evidence of systemic autoimmunity, but none had a cancer history. A more complete clinical spectrum of this syndrome remains to be described.

Encephalopathy with antibodies against GFAP — Antibodies against glial fibrillary acidic protein (GFAP, an astrocytic intracellular protein) have been described as a marker for a relapsing autoimmune meningoencephalitis or encephalitis, with or without myelitis [135,136]. Although these antibodies do not target a neuronal protein, they are included in this section because many of the reported patients had other relevant autoimmune responses (eg, anti-NMDA receptor antibodies, anti-GAD antibodies, and aquaporin-4 [AQP4] antibodies, among others) that may have driven the clinical syndrome. The patients had a range of clinical symptoms including headache, optic disc edema and optic papillitis, encephalopathy, myelopathy, tremor, autonomic dysfunction, and psychiatric disturbances [135-137]. Approximately one-third of the patients had a past or current systemic tumor. The CSF often showed a leukocytic pleocytosis. A variety of MRI features have been described including diffuse T2 abnormalities in periventricular white matter, leptomeningeal or perivascular enhancement (sometimes in a radial distribution), and longitudinally extensive T2 hyperintensity in the spinal cord [138]. Most of the patients responded to glucocorticoids [135,136]. GFAP-antibody-associated neurologic disorders can also occur as adverse events of checkpoint inhibitor immunotherapy.

Other encephalitides

Anti-mGluR2 encephalitis – Antibodies against mGluR2 were described in two patients with paraneoplastic cerebellar ataxia [139]. One patient was 78 years of age with SCLC who had a relapsing-remitting course and the other was three years old with an alveolar rhabdomyosarcoma who improved with corticosteroids. In both cases, the tumor was shown to express mGluR2.

Anti-SEZ6L2 encephalitis – Antibodies against seizure-related 6 homolog like 2 (SEZ6L2) were initially described in two patients who developed subacute cerebellar ataxia with retinopathy [140]. In a study of four additional patients, median age was 62 years, half were females, and the cerebellar syndrome was associated with extrapyramidal symptoms in three [141]. None of the reported patients had cancer.

Anti-GluK2 encephalitis – Antibodies against the glutamate kainate receptor subunit 2 (GluK2) have been described in eight patients with encephalitis and prominent cerebellar involvement [142]. The median age was 28 years (range 14 to 75), and five were males. The most common clinical presentation was rapidly progressive cerebellitis or encephalitis with cerebellar ataxia often accompanied by alterations of cognition and memory, decreased level of consciousness, myoclonus, or movement disorders. There was a remote history of Hodgkin lymphoma in one patient. Immunotherapy resulted in improvement in three of four patients with assessable follow-up.

INTRACELLULAR NEURONAL PROTEIN ANTIBODY SYNDROMES

High-risk antibodies (>70 percent cancer association) — These syndromes are almost always paraneoplastic and usually occur in older patients. The associated antibodies target intracellular neuronal proteins, and the pathogenic mechanisms are mediated by cytotoxic T cells that lead to irreversible neuronal degeneration and a poor response to treatment. (See "Overview of paraneoplastic syndromes of the nervous system", section on 'Immunologic mechanisms'.)

Anti-Hu encephalomyelitis — Anti-Hu paraneoplastic syndromes are often multifocal, resulting from involvement of cortical gray matter, temporal lobes, brainstem, cerebellum, dorsal root ganglia, and/or autonomic nervous system [143-146]. In many patients, symptoms begin with, and may remain restricted to, the dorsal root ganglia, causing a subacute sensory neuronopathy [144,145] (see "Paraneoplastic syndromes affecting spinal cord, peripheral nerve, and muscle", section on 'Subacute motor neuronopathy'). Other patients with anti-Hu antibodies present with a more restricted brainstem or limbic encephalitis [144,147].

Small cell lung cancer (SCLC) is found in most patients with anti-Hu encephalitis [143,145,146]. Some patients have other cancer types; no cancer is found in approximately 15 percent of patients [145,146]. Some children with anti-Hu antibodies have opsoclonus-myoclonus-ataxia syndrome (OMAS) and neuroblastoma, but more commonly, the encephalitis is not paraneoplastic. In one referral-based cohort, six of eight children with anti-Hu encephalitis were cancer free compared with none of 243 adults [148].

Early recognition of anti-Hu encephalomyelitis and prompt antitumor treatment are important in stabilizing or sometimes improving the neurologic symptoms. In one series of 200 patients with paraneoplastic encephalomyelitis and anti-Hu antibodies, antitumor treatment was associated with a significantly greater likelihood of neurologic improvement or stabilization (odds ratio [OR] 4.6) and a lower mortality rate (relative risk of death with no antitumor treatment 2.6) [145].

Immunomodulatory therapy (eg, glucocorticoids, immune globulin, therapeutic plasma exchange), either given with antitumor treatment or alone, has also been associated with stabilization in a smaller proportion of patients [146,149]. One report describes improvement with rituximab in two patients with paraneoplastic encephalomyelitis associated with the anti-Hu antibody [150]. In children, the syndrome appears to be more treatment resistant and associated with long-term sequelae of intractable epilepsy and cognitive impairment [148].

Neurologic dysfunction is a frequent cause of death, particularly when there is brainstem and autonomic dysfunction [144]. However, prolonged survival can occur in association with regression of the tumor [145,151].

Ma2-associated encephalitis — Ma2-associated encephalitis (also called anti-Ta) is usually associated with testicular germ-cell tumors, mainly seminoma [152]. The Ma2 antigen is selectively expressed in neurons and the testicular tumor. Ma2 shares homology with Ma1, a protein that is associated with other paraneoplastic neurologic syndromes, particularly brainstem and cerebellar dysfunction (table 1) [153,154].

The clinical presentation of anti-Ma2-associated encephalitis differs from classic paraneoplastic limbic encephalitis. In a series of 38 patients with anti-Ma2 encephalitis, 34 (89 percent) developed isolated or combined limbic (n = 27), diencephalic (n = 13), or brainstem encephalopathy (n = 25) [153]. MRI abnormalities were frequent in these brain regions, and inflammatory changes were typically present in the cerebrospinal fluid (CSF). Four patients developed other syndromes, including two with a predominant cerebellar ataxia that remained stable for several years and two with myelopathy.

Other prominent neurologic features included excessive daytime sleepiness and eye movement abnormalities, particularly a vertical gaze paresis (mainly upward gaze) that sometimes evolved to total external ophthalmoplegia. Three patients developed atypical parkinsonism, and two developed a severe hypokinetic syndrome. An additional case report describes an individual whose syndrome included amyotrophy of the upper extremities with an associated cervical cord lesion [155].

Testicular germ cell tumors are the most common associated neoplasms, identified in 18 of 34 patients found to have cancer in the above described series [153]. Orchiectomy may be required to reveal microscopic testicular germ cell neoplasia, as occurred in six males (age 26 to 40 years) with Ma2-associated encephalitis and no evidence of other cancer by rigorous testing [156]. All six males were identified to be at risk for testicular cancer because of new testicular enlargement, testicular microcalcifications, and/or cryptorchidism. Extragonadal germ cell tumors have also been described in association with this syndrome [157,158]. The diagnosis of testicular germ cell tumors is discussed separately. (See "Clinical manifestations, diagnosis, and staging of testicular germ cell tumors".)

Coexisting antibodies to Ma1 were identified in 15 of 34 patients with anti-Ma2 encephalitis and were significantly more common in patients with tumors other than testicular cancer (usually lung cancer) [153]. Compared with patients with antibodies only to Ma2, these patients are also more likely to develop ataxia and to have a worse prognosis.

Anti-Ma2-associated encephalitis is more responsive to oncologic and immunomodulatory therapy than other types of paraneoplastic encephalomyelitis [157]. In the series cited above, neurologic improvement or stabilization occurred in 18 of 33 patients with outcome information [153]. Features significantly associated with improvement or stabilization included male sex, age 45 years or less, testicular tumor with complete response to treatment, absence of anti-Ma1 antibodies, and limited central nervous system (CNS) involvement. However, a positive treatment response can occur in other patients as well [159].

Anti-CRMP5 encephalomyelitis — Antibodies to collapsin-responsive mediator protein 5 (CRMP5) have been associated with paraneoplastic encephalitis. Cortical symptoms are often not confined to the limbic system; distinctive symptoms including cerebellar ataxia, chorea, optic neuropathy, cranial neuropathies, and loss of olfaction and taste have been described [160-164]. Other paraneoplastic syndromes associated with these antibodies include axonal sensorimotor neuropathy, uveitis, and optic neuritis.

The most common associated cancers are SCLC and thymoma. Improvement with anticancer therapy and glucocorticoids has been described.

Others

Stiff-person syndrome – Stiff-person syndrome may occur in isolation or as part of encephalomyelitis. Paraneoplastic stiff-person syndrome is associated with antiamphiphysin antibodies and rarely with antibodies to glutamic acid decarboxylase 65 kilodalton isoform (GAD65) or glycine receptor (GlyR) that are more commonly associated with the nonparaneoplastic syndrome [123,165-167]. (See "Paraneoplastic syndromes affecting spinal cord, peripheral nerve, and muscle", section on 'Stiff-person syndrome'.)

Anti-Ri encephalitis – Antibodies to Ri are found in patients with paraneoplastic OMAS. These antibodies have also been described in some patients with paraneoplastic brainstem encephalitis with eye movement disorders, dysphagia, ptosis, laryngospasm, and/or jaw dystonia [168]. While most patients do not respond to therapy, as with other paraneoplastic neurologic disorders, there are reports of responses to early initiation of immunomodulatory therapy and tumor-directed treatment. (See "Opsoclonus-myoclonus-ataxia syndrome", section on 'Pathogenesis'.)

Anti-KLHL11 encephalitis – Antibodies to Kelch-like protein 11 (KLHL11) have been described in patients with a predominant brainstem/cerebellar encephalitis, often with sensorineural hearing loss, and are frequently but not invariably associated with tumors, most commonly benign teratoma or a testicular tumor [169-171]. Other symptoms may include limbic encephalitis, myelitis, hypersomnia, and seizures [172]. Both males and females can be affected, and patients ranging in age from 9 to 76 years have been reported. These antibodies have also been described in patients with other types of autoimmune encephalitis, including anti-N-methyl-D-aspartate (NMDA) receptor and anti-Ma2 encephalitis [171]. In these patients, the presence of KLHL11 antibodies did not confer distinct clinical features.

Low-risk antibodies (<30 percent cancer association)

Anti-GAD65 encephalitis High-titer antibodies against GAD65 can be associated with three distinct syndromes, sometimes with overlapping symptoms: stiff-person syndrome and variants, cerebellar ataxia, and encephalitis with pharmacoresistent temporal lobe seizures [173]. Patients with GAD65 antibody-associated neurologic syndromes always have high-titer GAD65 antibodies in CSF, and more than 80 percent have evidence of intrathecal antibody synthesis. Since low titers of GAD65 antibodies can be found in the serum of patients with type 1 diabetes mellitus, healthy subjects, and patients with a variety of nonimmunologic neurologic disorders [174], the determination that a neurologic syndrome is GAD antibody-associated should be based in CSF antibody testing and determination of intrathecal GAD65 antibody synthesis [175]. To date, there is no clear evidence that GAD antibodies are pathogenic.

Anti-AK5 encephalitis – Antibodies to adenylate kinase 5 (AK5) were initially described in two patients with the subacute onset of a severe limbic encephalitis refractory to therapy [176], and additional patients have been encountered [177-179]. In a review of 26 patients, the median age of onset was 66 years, and 77 percent of patients were male [179]. Predominant symptoms were severe episodic amnesia, depression, anorexia, and weight loss. MRI demonstrated bilateral medial temporal T2-weighted signal abnormalities in most patients, which evolved to severe atrophy. AK5 is an intracellular protein, and the pathogenesis of anti-AK5 encephalitis appears to be related to T cell-mediated cytotoxicity resulting in extensive neuronal loss. The response to immunomodulatory therapy is generally poor.

DIAGNOSTIC APPROACH — The diagnosis of autoimmune encephalitis is established by detection of specific antineuronal antibodies in patients with a characteristic clinical syndrome. However, not all patients with autoimmune encephalitis have antibodies. For these patients, the suspicion of an autoimmune etiology is based on [180]:

Acute or subacute presentation

Clinical features, which in some cases are highly suggestive of a specific autoimmune encephalitis syndrome

Clinical history, including cancer risk factors

Supportive laboratory and radiologic findings

In all patients, it is critical to consider alternative etiologies for the encephalitis syndrome (table 4), as initial treatment decisions are often made before confirmatory antibody test results are available, and infectious etiologies in particular should be excluded before embarking on immunomodulatory therapies.

Differential diagnosis — The differential diagnosis of autoimmune (including paraneoplastic) encephalitis includes a variety of alternative causes of encephalitis and encephalopathy. Broad categories include infection, toxic and metabolic disturbances, vascular disorders, neoplastic disorders, demyelinating and inflammatory disorders, psychiatric disease, neurodegenerative dementias, and rare heritable or metabolic disorders (table 4).

Syndromes such as anti-N-methyl-D-aspartate (NMDA) receptor encephalitis have a characteristic pattern of symptoms that are easily recognizable. By contrast, other syndromes such as limbic encephalitis may initially have a wide differential diagnosis. For example, metastatic disease affecting the brain or leptomeninges, viral encephalitis, Creutzfeldt-Jakob disease, ischemic and hemorrhagic cerebrovascular disease, Whipple disease, psychiatric disease, toxic-metabolic encephalopathy, Wernicke encephalopathy, and primary degenerative dementia have all been reported to cause a clinical syndrome for which the diagnosis of autoimmune limbic encephalitis was considered [20,31,143,181-183]. The differential diagnosis in patients with predominant brainstem encephalitis also includes certain additional disorders with a predilection for the brainstem, such as multiple sclerosis, Behçet syndrome, Listeria infection, enterovirus, and tuberculosis infection [184]. Some of the autoimmune encephalitides have early and prominent psychiatric manifestations that overlap with primary psychiatric diseases and substance use disorders. Careful examination may reveal subtle neurologic deficits at this stage, findings that should lead to the consideration of autoimmune encephalitis. Most patients develop additional features of the syndrome within days or a few weeks [5,185].

Clinical evaluation and imaging — Patients with suspected autoimmune encephalitis (table 5) should have neuroimaging, EEG, lumbar puncture, and serologic testing for appropriate biomarkers to confirm the diagnosis and exclude alternative etiologies.

Neuroimaging – A brain MRI is helpful in this clinical setting to exclude a cerebrovascular event or metastatic disease, among others. Characteristic MRI findings in patients with autoimmune encephalitis include signal hyperintensities on fluid-attenuated inversion recovery (FLAIR) or T2-weighted images in affected brain regions (eg, medial temporal lobes and/or brainstem); subcortical regions and the cerebellum are sometimes affected as well. Contrast enhancement is variable. Although MRI findings are neither sensitive nor specific for these disorders, in the appropriate clinical setting they can be highly suggestive of specific syndromes (eg, multifocal FLAIR and T2 signal abnormalities in gamma-aminobutyric acid A [GABA-A] receptor encephalitis) [97]. (See 'Anti-GABA-A receptor encephalitis' above.)

EEG – An EEG should be performed in most cases to exclude nonconvulsive seizures. In patients with autoimmune encephalitis, nonspecific EEG abnormalities are common and include focal or generalized slowing, epileptiform activity, and periodic lateralized epileptiform discharges (PLEDs) [186]. Less than one-third of patients with NMDA receptor encephalitis have an EEG pattern called extreme delta brush that is considered characteristic for the disorder [17].

Lumbar puncture – Cerebrospinal fluid (CSF) examination should be performed and should include cell count, protein, and glucose concentrations; inflammatory markers (immune globulin G [IgG] index, oligoclonal bands); cell-surface and/or intracellular neuronal antibodies; broad viral studies including herpes simplex virus (HSV) 1/2 and varicella zoster virus (VZV) polymerase chain reaction (PCR), and bacterial and fungal cultures when appropriate (see "Viral encephalitis in adults", section on 'Diagnosis'); and cytology to exclude leptomeningeal metastasis [187]. (See "Clinical features and diagnosis of leptomeningeal disease from solid tumors", section on 'Diagnostic evaluation'.)

Patients with autoimmune encephalitis may have normal or abnormal CSF findings. Abnormalities include modest elevation of protein (<100 mg/dL), mild to moderate lymphocytic pleocytosis, elevated IgG index, and/or the presence of oligoclonal bands [186,188]. However, these findings are variable, and some syndromes, such as anti-leucine-rich glioma inactivated 1 (LGI1) encephalitis, often have only mildly abnormal or normal CSF studies [189]. A lack of inflammatory findings on CSF and MRI may be particularly common in older adults [190].

Oncologic evaluation – If the patient does not have a known cancer diagnosis, evaluation for occult malignancy should also ensue, with the syndrome often providing a clue as to the tumor location (table 1 and table 3). For example, as small cell lung cancer (SCLC) comprises the majority of underlying neoplasms in older patients with limbic encephalitis, chest imaging and 18-fluorodeoxyglucose-positron emission tomography (FDG-PET) should have a high priority in patients with this syndrome, whereas young females with anti-NMDA receptor encephalitis should be thoroughly investigated for an ovarian teratoma. (See "Overview of paraneoplastic syndromes of the nervous system", section on 'Search for occult malignancy'.)

Antibody testing — Autoimmune encephalitis antibody testing should initially always include CSF and serum (table 1) [20]. Not all biomarkers have commercially available testing, and some antigens remain to be characterized. Thus, negative results do not exclude an autoimmune disorder [188]. A sample of a patient's CSF and serum can be sent to a research laboratory for examination in these cases. Other caveats regarding antibody testing are discussed separately. (See "Overview of paraneoplastic syndromes of the nervous system", section on 'Antibody screening'.)

Of note, some forms of autoimmune cortical encephalitis are associated with myelin oligodendrocyte glycoprotein (MOG) antibodies; for MOG antibodies, the optimal sample is serum. MOG antibody-associated cases predominantly involving cortical gray matter can occur in adults and children; however, serum MOG antibody testing should be considered in most children with suspected autoimmune encephalitis. Studies suggest that in children with autoimmune encephalitis, MOG and NMDA receptor antibodies are by far the most frequent autoantibodies [191]. (See "Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD): Clinical features and diagnosis".)

General principles to follow when assessing and interpreting neuronal antibody testing in a patient with symptoms suggesting autoimmune encephalitis include the following:

Test for antibodies in CSF and serum. The approach of first testing serum and then, if negative, testing CSF is not recommended, as it delays diagnosis and may lead to false-positive results.

If serum antibody testing is positive but the CSF is negative, consider that the serum result may be a false-positive diagnosis. Contact the laboratory and request retesting of the samples.

If the clinical picture does not fit with the antibody identified, the antibody may be a false-positive result [192], particularly if only serum has been examined, or if the antibodies were only identified in serum. In this case, contact the clinical laboratory for retesting or a research laboratory for guidance.

Clinical decisions should be based on clinical assessment rather than on antibody titers. Although the antibody titers may correlate with the clinical course, this correlation is imperfect, and antibody titers often remain detectable even after recovery.

Among over 100 patients referred to tertiary care centers with a diagnosis of autoimmune encephalitis who were ultimately diagnosed with an alternative disorder, the most common misdiagnoses were functional neurological symptom disorder (conversion disorder), neurodegenerative disease, primary psychiatric disease, cognitive deficits from comorbidities, and cerebral neoplasm [193]. In retrospect, approximately half of the misdiagnoses were caused by overinterpretation of nonspecific positive serum antibody testing. Misdiagnosis based on CSF testing was much less common. These results emphasize the primacy of clinical judgment and the importance of careful consideration of the differential diagnosis, understanding of the nuances of antibody testing, and, in particular when CSF is not investigated for antibodies, adherence to diagnostic criteria [194].

Diagnostic criteria — A clinical approach to the diagnosis of autoimmune encephalitis has been developed by expert panels [23,195]. Because autoantibody test results (table 1) and response to antitumor and immunomodulatory therapy are not available at disease onset, these criteria rely on neurologic assessment and conventional neuroimaging and CSF testing reviewed above [23]. Misdiagnosis of autoimmune encephalitis and probable antibody-negative autoimmune encephalitis can be mitigated by adherence to these criteria and comprehensive antibody testing [180].

Diagnostic criteria for definite autoimmune limbic encephalitis require all four of the following (table 6):

Subacute onset (rapid progression of <3 months) of working memory deficits (short-term memory loss), seizures, or psychiatric symptoms suggesting involvement of the limbic system

Bilateral brain abnormalities on T2-weighted FLAIR MRI highly restricted to the medial temporal lobes

At least one of the following:

CSF pleocytosis (>5 white blood cells per mm3)

EEG with epileptic or slow-wave activity involving the temporal lobes

Reasonable exclusion of alternative causes

If one of the first three criteria is not met, a diagnosis of definite limbic encephalitis can be made only with the detection of antibodies against cell-surface or onconeuronal proteins [23]. In addition to limbic encephalitis, specific criteria for probable and definite anti-NMDA receptor encephalitis are also available (table 2). (See 'Anti-NMDA receptor encephalitis' above.)

Proposed criteria for probable antibody-negative autoimmune encephalitis require the following [180]:

Rapid progression (<3 months) of working memory deficit (short-term memory loss), altered mental status, or psychiatric symptoms

Exclusion of well-defined syndromes of autoimmune encephalitis (limbic encephalitis, acute disseminated encephalomyelitis, Bickerstaff brainstem encephalitis, among others)

Absence of well-characterized autoantibodies in serum and CSF, and at least two of the following:

MRI abnormalities suggesting autoimmune encephalitis

CSF pleocytosis, CSF-specific oligoclonal bands, or elevated CSF IgG index

Brain biopsy showing inflammatory infiltrates and excluding other disorders (eg, vasculitis or tumor)

Reasonable exclusion of alternative causes (table 4)

Diagnostic criteria for paraneoplastic neurologic syndromes are reviewed separately [195]. (See "Overview of paraneoplastic syndromes of the nervous system", section on 'Diagnostic criteria'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Paraneoplastic neurologic disorders".)

SUMMARY AND RECOMMENDATIONS

Terminology – Autoimmune encephalitis includes the encephalitis syndromes associated with antibodies against neuronal cell-surface proteins and the paraneoplastic encephalitis syndromes that are usually associated with antibodies against intracellular neuronal (onconeuronal) proteins. (See 'Introduction' above.)

Clinical syndromes

Associated with antibodies against neuronal cell-surface proteins – A large number of autoimmune encephalitis syndromes have been identified in association with antibodies against neuronal cell-surface or synaptic proteins (table 1). The most common is anti-N-methyl-D-aspartate (NMDA) receptor encephalitis (table 2). (See 'Anti-NMDA receptor encephalitis' above.)

These syndromes occur in patients of all ages, some with a preferential age distribution, and may or may not be associated with the presence of a cancer. For many syndromes, the antibodies have been shown to be pathogenic via antibody-mediated effects. (See 'Neuronal cell-surface protein antibody syndromes' above.)

Associated with antibodies against intracellular neuronal proteins (onconeural proteins), high cancer association – These disorders are almost always paraneoplastic and can involve the limbic system (limbic encephalitis), brainstem (brainstem encephalitis), spinal cord (myelitis), or the entire neuraxis (encephalomyelitis). In most cases, symptoms have an acute to subacute onset and are accompanied by evidence of inflammation in the cerebrospinal fluid (CSF). Pathogenesis is mediated by cytotoxic T cells and neuronal degeneration. (See 'High-risk antibodies (>70 percent cancer association)' above.)

Examples of classic paraneoplastic encephalitis syndromes include anti-Hu encephalomyelitis, often associated with small cell lung cancer (SCLC), Ma2-associated encephalitis related to testicular cancer, and anti-collapsin-responsive mediator protein 5 (CRMP5) encephalomyelitis related to SCLC or thymoma (table 1).

Associated with antibodies against intracellular neuronal proteins, low cancer association – These antibody-associated disorders are rarely associated with cancer, but they appear to be mediated by cytotoxic T cell mechanisms rather than direct antibody-mediated effects. Examples include anti-glutamic acid decarboxylase 65 kilodalton isoform (GAD65) and anti-adenylate kinase 5 (AK5) encephalitis. (See 'Low-risk antibodies (<30 percent cancer association)' above.)

Diagnostic approach – Patients with suspected autoimmune encephalitis (table 5) should have neuroimaging (image 1), EEG, lumbar puncture, and CSF and serum neuronal antibody testing (table 1). Detection of specific antineuronal antibodies establishes the diagnosis of autoimmune encephalitis in patients with a characteristic clinical syndrome. (See 'Diagnostic approach' above and 'Diagnostic criteria' above.)

However, initial treatment decisions are often required while antibody results are pending. A provisional diagnosis of autoimmune encephalitis can be made based on the medical history and clinical features, laboratory and radiologic evidence of central nervous system (CNS) inflammation, and exclusion of infection and other alternative etiologies. (See 'Clinical evaluation and imaging' above.)

Differential diagnosis – In all patients, it is critical to consider alternative etiologies for the encephalitis syndrome (table 4). The differential diagnosis includes a variety of alternative causes of encephalitis and encephalopathy. Broad categories include infection, toxic and metabolic disturbances, vascular disorders, neoplastic disorders, demyelinating and inflammatory disorders, psychiatric disease, neurodegenerative dementias, and rare heritable or metabolic disorders. (See 'Differential diagnosis' above.)

  1. Dalmau J, Tüzün E, Wu HY, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 2007; 61:25.
  2. Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008; 7:1091.
  3. Florance-Ryan N, Dalmau J. Update on anti-N-methyl-D-aspartate receptor encephalitis in children and adolescents. Curr Opin Pediatr 2010; 22:739.
  4. Dalmau J, Lancaster E, Martinez-Hernandez E, et al. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 2011; 10:63.
  5. Titulaer MJ, McCracken L, Gabilondo I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013; 12:157.
  6. Kayser MS, Titulaer MJ, Gresa-Arribas N, Dalmau J. Frequency and characteristics of isolated psychiatric episodes in anti–N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2013; 70:1133.
  7. Guasp M, Giné-Servén E, Maudes E, et al. Clinical, Neuroimmunologic, and CSF Investigations in First Episode Psychosis. Neurology 2021; 97:e61.
  8. Ariño H, Muñoz-Lopetegi A, Martinez-Hernandez E, et al. Sleep disorders in anti-NMDAR encephalitis. Neurology 2020; 95:e671.
  9. Luca N, Daengsuwan T, Dalmau J, et al. Anti-N-methyl-D-aspartate receptor encephalitis: a newly recognized inflammatory brain disease in children. Arthritis Rheum 2011; 63:2516.
  10. Baizabal-Carvallo JF, Stocco A, Muscal E, Jankovic J. The spectrum of movement disorders in children with anti-NMDA receptor encephalitis. Mov Disord 2013; 28:543.
  11. Armangue T, Titulaer MJ, Málaga I, et al. Pediatric anti-N-methyl-D-aspartate receptor encephalitis-clinical analysis and novel findings in a series of 20 patients. J Pediatr 2013; 162:850.
  12. Gable M, Glaser C. Anti-N-Methyl-d-Aspartate Receptor Encephalitis Appearing as a New-Onset Psychosis: Disease Course in Children and Adolescents Within the California Encephalitis Project. Pediatr Neurol 2017; 72:25.
  13. Wright S, Hacohen Y, Jacobson L, et al. N-methyl-D-aspartate receptor antibody-mediated neurological disease: results of a UK-based surveillance study in children. Arch Dis Child 2015; 100:521.
  14. Gorman MP, Gombolay GY, Mehan WA Jr, Thibert RL. Case 27-2018: A 3-Year-Old Boy with Seizures. N Engl J Med 2018; 379:870.
  15. Qu XP, Vidaurre J, Peng XL, et al. Seizure Characteristics, Outcome, and Risk of Epilepsy in Pediatric Anti-N-Methyl-d-Aspartate Receptor Encephalitis. Pediatr Neurol 2020; 105:35.
  16. Titulaer MJ, McCracken L, Gabilondo I, et al. Late-onset anti-NMDA receptor encephalitis. Neurology 2013; 81:1058.
  17. Schmitt SE, Pargeon K, Frechette ES, et al. Extreme delta brush: a unique EEG pattern in adults with anti-NMDA receptor encephalitis. Neurology 2012; 79:1094.
  18. Florance NR, Davis RL, Lam C, et al. Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis in children and adolescents. Ann Neurol 2009; 66:11.
  19. Leypoldt F, Buchert R, Kleiter I, et al. Fluorodeoxyglucose positron emission tomography in anti-N-methyl-D-aspartate receptor encephalitis: distinct pattern of disease. J Neurol Neurosurg Psychiatry 2012; 83:681.
  20. Prüss H, Dalmau J, Harms L, et al. Retrospective analysis of NMDA receptor antibodies in encephalitis of unknown origin. Neurology 2010; 75:1735.
  21. Martinez-Hernandez E, Horvath J, Shiloh-Malawsky Y, et al. Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis. Neurology 2011; 77:589.
  22. Gresa-Arribas N, Titulaer MJ, Torrents A, et al. Antibody titres at diagnosis and during follow-up of anti-NMDA receptor encephalitis: a retrospective study. Lancet Neurol 2014; 13:167.
  23. Graus F, Titulaer MJ, Balu R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016; 15:391.
  24. Dahm L, Ott C, Steiner J, et al. Seroprevalence of autoantibodies against brain antigens in health and disease. Ann Neurol 2014; 76:82.
  25. Hara M, Martinez-Hernandez E, Ariño H, et al. Clinical and pathogenic significance of IgG, IgA, and IgM antibodies against the NMDA receptor. Neurology 2018; 90:e1386.
  26. Seki M, Suzuki S, Iizuka T, et al. Neurological response to early removal of ovarian teratoma in anti-NMDAR encephalitis. J Neurol Neurosurg Psychiatry 2008; 79:324.
  27. Okamoto S, Hirano T, Takahashi Y, et al. Paraneoplastic limbic encephalitis caused by ovarian teratoma with autoantibodies to glutamate receptor. Intern Med 2007; 46:1019.
  28. Sansing LH, Tüzün E, Ko MW, et al. A patient with encephalitis associated with NMDA receptor antibodies. Nat Clin Pract Neurol 2007; 3:291.
  29. Gable MS, Gavali S, Radner A, et al. Anti-NMDA receptor encephalitis: report of ten cases and comparison with viral encephalitis. Eur J Clin Microbiol Infect Dis 2009; 28:1421.
  30. Dale RC, Irani SR, Brilot F, et al. N-methyl-D-aspartate receptor antibodies in pediatric dyskinetic encephalitis lethargica. Ann Neurol 2009; 66:704.
  31. Vincent A, Bien CG, Irani SR, Waters P. Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol 2011; 10:759.
  32. Burr T, Barton C, Doll E, et al. N-Methyl-d-Aspartate Receptor Encephalitis Associated With COVID-19 Infection in a Toddler. Pediatr Neurol 2021; 114:75.
  33. Monti G, Giovannini G, Marudi A, et al. Anti-NMDA receptor encephalitis presenting as new onset refractory status epilepticus in COVID-19. Seizure 2020; 81:18.
  34. Panariello A, Bassetti R, Radice A, et al. Anti-NMDA receptor encephalitis in a psychiatric Covid-19 patient: A case report. Brain Behav Immun 2020; 87:179.
  35. Lancaster E, Martinez-Hernandez E, Dalmau J. Encephalitis and antibodies to synaptic and neuronal cell surface proteins. Neurology 2011; 77:179.
  36. Zandi MS, Irani SR, Follows G, et al. Limbic encephalitis associated with antibodies to the NMDA receptor in Hodgkin lymphoma. Neurology 2009; 73:2039.
  37. Sanmaneechai O, Song JL, Nevadunsky N, et al. Anti-N-methyl-d-aspartate encephalitis with ovarian cystadenofibroma. Pediatr Neurol 2013; 48:232.
  38. Lebas A, Husson B, Didelot A, et al. Expanding spectrum of encephalitis with NMDA receptor antibodies in young children. J Child Neurol 2010; 25:742.
  39. Williams TJ, Benavides DR, Patrice KA, et al. Association of Autoimmune Encephalitis With Combined Immune Checkpoint Inhibitor Treatment for Metastatic Cancer. JAMA Neurol 2016; 73:928.
  40. Leypoldt F, Titulaer MJ, Aguilar E, et al. Herpes simplex virus-1 encephalitis can trigger anti-NMDA receptor encephalitis: case report. Neurology 2013; 81:1637.
  41. Armangue T, Moris G, Cantarín-Extremera V, et al. Autoimmune post-herpes simplex encephalitis of adults and teenagers. Neurology 2015; 85:1736.
  42. Armangue T, Leypoldt F, Málaga I, et al. Herpes simplex virus encephalitis is a trigger of brain autoimmunity. Ann Neurol 2014; 75:317.
  43. Armangue T, Spatola M, Vlagea A, et al. Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: a prospective observational study and retrospective analysis. Lancet Neurol 2018; 17:760.
  44. Armangué T, Olivé-Cirera G, Martínez-Hernandez E, et al. Neurologic complications in herpes simplex encephalitis: clinical, immunological and genetic studies. Brain 2023; 146:4306.
  45. Prüss H, Finke C, Höltje M, et al. N-methyl-D-aspartate receptor antibodies in herpes simplex encephalitis. Ann Neurol 2012; 72:902.
  46. Hacohen Y, Deiva K, Pettingill P, et al. N-methyl-D-aspartate receptor antibodies in post-herpes simplex virus encephalitis neurological relapse. Mov Disord 2014; 29:90.
  47. Sutcu M, Akturk H, Somer A, et al. Role of Autoantibodies to N-Methyl-d-Aspartate (NMDA) Receptor in Relapsing Herpes Simplex Encephalitis: A Retrospective, One-Center Experience. J Child Neurol 2016; 31:345.
  48. Titulaer MJ, Leypoldt F, Dalmau J. Antibodies to N-methyl-D-aspartate and other synaptic receptors in choreoathetosis and relapsing symptoms post-herpes virus encephalitis. Mov Disord 2014; 29:3.
  49. Heine J, Kopp UA, Klag J, et al. Long-Term Cognitive Outcome in Anti-N-Methyl-D-Aspartate Receptor Encephalitis. Ann Neurol 2021; 90:949.
  50. Guasp M, Rosa-Justicia M, Muñoz-Lopetegi A, et al. Clinical characterisation of patients in the post-acute stage of anti-NMDA receptor encephalitis: a prospective cohort study and comparison with patients with schizophrenia spectrum disorders. Lancet Neurol 2022; 21:899.
  51. Balu R, McCracken L, Lancaster E, et al. A score that predicts 1-year functional status in patients with anti-NMDA receptor encephalitis. Neurology 2019; 92:e244.
  52. Nosadini M, Eyre M, Molteni E, et al. Use and Safety of Immunotherapeutic Management of N-Methyl-d-Aspartate Receptor Antibody Encephalitis: A Meta-analysis. JAMA Neurol 2021; 78:1333.
  53. Bartels F, Krohn S, Nikolaus M, et al. Clinical and Magnetic Resonance Imaging Outcome Predictors in Pediatric Anti-N-Methyl-D-Aspartate Receptor Encephalitis. Ann Neurol 2020; 88:148.
  54. Gabilondo I, Saiz A, Galán L, et al. Analysis of relapses in anti-NMDAR encephalitis. Neurology 2011; 77:996.
  55. Chourasia N, Watkins MW, Lankford JE, et al. An Infant Born to a Mother With Anti-N-Methyl-d-Aspartate Receptor Encephalitis. Pediatr Neurol 2018; 79:65.
  56. Jagota P, Vincent A, Bhidayasiri R. Transplacental transfer of NMDA receptor antibodies in an infant with cortical dysplasia. Neurology 2014; 82:1662.
  57. Hilderink M, Titulaer MJ, Schreurs MW, et al. Transient anti-NMDAR encephalitis in a newborn infant due to transplacental transmission. Neurol Neuroimmunol Neuroinflamm 2015; 2:e126.
  58. Magley J, Towner D, Taché V, Apperson ML. Pregnancy outcome in anti-N-methyl-D-aspartate receptor encephalitis. Obstet Gynecol 2012; 120:480.
  59. Kumar MA, Jain A, Dechant VE, et al. Anti-N-methyl-D-aspartate receptor encephalitis during pregnancy. Arch Neurol 2010; 67:884.
  60. Lamale-Smith LM, Moore GS, Guntupalli SR, Scott JB. Maternal-fetal transfer of anti-N-methyl-D-aspartate receptor antibodies. Obstet Gynecol 2015; 125:1056.
  61. Joubert B, García-Serra A, Planagumà J, et al. Pregnancy outcomes in anti-NMDA receptor encephalitis: Case series. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  62. Irani SR, Michell AW, Lang B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011; 69:892.
  63. Sen A, Wang J, Laue-Gizzi H, et al. Pathognomonic seizures in limbic encephalitis associated with anti-LGI1 antibodies. Lancet 2014; 383:2018.
  64. Thompson J, Bi M, Murchison AG, et al. The importance of early immunotherapy in patients with faciobrachial dystonic seizures. Brain 2018; 141:348.
  65. López-Chiriboga AS, Klein C, Zekeridou A, et al. LGI1 and CASPR2 neurological autoimmunity in children. Ann Neurol 2018; 84:473.
  66. Ariño H, Armangué T, Petit-Pedrol M, et al. Anti-LGI1-associated cognitive impairment: Presentation and long-term outcome. Neurology 2016; 87:759.
  67. Navarro V, Kas A, Apartis E, et al. Motor cortex and hippocampus are the two main cortical targets in LGI1-antibody encephalitis. Brain 2016; 139:1079.
  68. Irani SR, Alexander S, Waters P, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan's syndrome and acquired neuromyotonia. Brain 2010; 133:2734.
  69. Fukata Y, Lovero KL, Iwanaga T, et al. Disruption of LGI1-linked synaptic complex causes abnormal synaptic transmission and epilepsy. Proc Natl Acad Sci U S A 2010; 107:3799.
  70. Petit-Pedrol M, Sell J, Planagumà J, et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018; 141:3144.
  71. Jacob S, Irani SR, Rajabally YA, et al. Hypothermia in VGKC antibody-associated limbic encephalitis. J Neurol Neurosurg Psychiatry 2008; 79:202.
  72. Vincent A, Buckley C, Schott JM, et al. Potassium channel antibody-associated encephalopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain 2004; 127:701.
  73. Wong SH, Saunders MD, Larner AJ, et al. An effective immunotherapy regimen for VGKC antibody-positive limbic encephalitis. J Neurol Neurosurg Psychiatry 2010; 81:1167.
  74. Irani SR, Gelfand JM, Bettcher BM, et al. Effect of rituximab in patients with leucine-rich, glioma-inactivated 1 antibody-associated encephalopathy. JAMA Neurol 2014; 71:896.
  75. van Sonderen A, Thijs RD, Coenders EC, et al. Anti-LGI1 encephalitis: Clinical syndrome and long-term follow-up. Neurology 2016; 87:1449.
  76. Finke C, Prüss H, Heine J, et al. Evaluation of Cognitive Deficits and Structural Hippocampal Damage in Encephalitis With Leucine-Rich, Glioma-Inactivated 1 Antibodies. JAMA Neurol 2017; 74:50.
  77. de Bruijn MAAM, van Sonderen A, van Coevorden-Hameete MH, et al. Evaluation of seizure treatment in anti-LGI1, anti-NMDAR, and anti-GABABR encephalitis. Neurology 2019; 92:e2185.
  78. Smith KM, Dubey D, Liebo GB, et al. Clinical Course and Features of Seizures Associated With LGI1-Antibody Encephalitis. Neurology 2021; 97:e1141.
  79. Lancaster E, Huijbers MG, Bar V, et al. Investigations of caspr2, an autoantigen of encephalitis and neuromyotonia. Ann Neurol 2011; 69:303.
  80. Irani SR, Pettingill P, Kleopa KA, et al. Morvan syndrome: clinical and serological observations in 29 cases. Ann Neurol 2012; 72:241.
  81. Joubert B, Saint-Martin M, Noraz N, et al. Characterization of a Subtype of Autoimmune Encephalitis With Anti-Contactin-Associated Protein-like 2 Antibodies in the Cerebrospinal Fluid, Prominent Limbic Symptoms, and Seizures. JAMA Neurol 2016; 73:1115.
  82. Patterson KR, Dalmau J, Lancaster E. Mechanisms of Caspr2 antibodies in autoimmune encephalitis and neuromyotonia. Ann Neurol 2018; 83:40.
  83. Joubert B, Petit-Pedrol M, Planagumà J, et al. Human CASPR2 Antibodies Reversibly Alter Memory and the CASPR2 Protein Complex. Ann Neurol 2022; 91:801.
  84. van Sonderen A, Ariño H, Petit-Pedrol M, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology 2016; 87:521.
  85. Syrbe S, Stettner GM, Bally J, et al. CASPR2 autoimmunity in children expanding to mild encephalopathy with hypertension. Neurology 2020; 94:e2290.
  86. Garrido Sanabria ER, Zahid A, Britton J, et al. CASPR2-IgG-associated autoimmune seizures. Epilepsia 2022; 63:709.
  87. Gövert F, Abrante L, Becktepe J, et al. Distinct movement disorders in contactin-associated-protein-like-2 antibody-associated autoimmune encephalitis. Brain 2023; 146:657.
  88. Joubert B, Gobert F, Thomas L, et al. Autoimmune episodic ataxia in patients with anti-CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflamm 2017; 4:e371.
  89. Lai M, Hughes EG, Peng X, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 2009; 65:424.
  90. Bataller L, Galiano R, García-Escrig M, et al. Reversible paraneoplastic limbic encephalitis associated with antibodies to the AMPA receptor. Neurology 2010; 74:265.
  91. Graus F, Boronat A, Xifró X, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010; 74:857.
  92. Höftberger R, van Sonderen A, Leypoldt F, et al. Encephalitis and AMPA receptor antibodies: Novel findings in a case series of 22 patients. Neurology 2015; 84:2403.
  93. Joubert B, Kerschen P, Zekeridou A, et al. Clinical Spectrum of Encephalitis Associated With Antibodies Against the α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid Receptor: Case Series and Review of the Literature. JAMA Neurol 2015; 72:1163.
  94. Daneshmand A, Goyal G, Markovic S, et al. Autoimmune Encephalitis Secondary to Melanoma. Ann Intern Med 2019; 170:905.
  95. Peng X, Hughes EG, Moscato EH, et al. Cellular plasticity induced by anti-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor encephalitis antibodies. Ann Neurol 2015; 77:381.
  96. Haselmann H, Mannara F, Werner C, et al. Human Autoantibodies against the AMPA Receptor Subunit GluA2 Induce Receptor Reorganization and Memory Dysfunction. Neuron 2018; 100:91.
  97. Petit-Pedrol M, Armangue T, Peng X, et al. Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol 2014; 13:276.
  98. Spatola M, Petit-Pedrol M, Simabukuro MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017; 88:1012.
  99. Höftberger R, Titulaer MJ, Sabater L, et al. Encephalitis and GABAB receptor antibodies: novel findings in a new case series of 20 patients. Neurology 2013; 81:1500.
  100. van Coevorden-Hameete MH, de Bruijn MAAM, de Graaff E, et al. The expanded clinical spectrum of anti-GABABR encephalitis and added value of KCTD16 autoantibodies. Brain 2019; 142:1631.
  101. Boronat A, Sabater L, Saiz A, et al. GABA(B) receptor antibodies in limbic encephalitis and anti-GAD-associated neurologic disorders. Neurology 2011; 76:795.
  102. Kruer MC, Hoeftberger R, Lim KY, et al. Aggressive course in encephalitis with opsoclonus, ataxia, chorea, and seizures: the first pediatric case of γ-aminobutyric acid type B receptor autoimmunity. JAMA Neurol 2014; 71:620.
  103. Lancaster E, Lai M, Peng X, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 2010; 9:67.
  104. Zhao XH, Yang X, Liu XW, Wang SJ. Clinical features and outcomes of Chinese patients with anti-γ-aminobutyric acid B receptor encephalitis. Exp Ther Med 2020; 20:617.
  105. Sabater L, Gaig C, Gelpi E, et al. A novel non-rapid-eye movement and rapid-eye-movement parasomnia with sleep breathing disorder associated with antibodies to IgLON5: a case series, characterisation of the antigen, and post-mortem study. Lancet Neurol 2014; 13:575.
  106. Högl B, Heidbreder A, Santamaria J, et al. IgLON5 autoimmunity and abnormal behaviours during sleep. Lancet 2015; 385:1590.
  107. Simabukuro MM, Sabater L, Adoni T, et al. Sleep disorder, chorea, and dementia associated with IgLON5 antibodies. Neurol Neuroimmunol Neuroinflamm 2015; 2:e136.
  108. Gaig C, Graus F, Compta Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017; 88:1736.
  109. Gaig C, Compta Y, Heidbreder A, et al. Frequency and Characterization of Movement Disorders in Anti-IgLON5 Disease. Neurology 2021.
  110. Grüter T, Möllers FE, Tietz A, et al. Clinical, serological and genetic predictors of response to immunotherapy in anti-IgLON5 disease. Brain 2023; 146:600.
  111. Gaig C, Iranzo A, Cajochen C, et al. Characterization of the sleep disorder of anti-IgLON5 disease. Sleep 2019; 42.
  112. Theis H, Bischof GN, Brüggemann N, et al. In Vivo Measurement of Tau Depositions in Anti-IgLON5 Disease Using [18F]PI-2620 PET. Neurology 2023; 101:e2325.
  113. Gelpi E, Höftberger R, Graus F, et al. Neuropathological criteria of anti-IgLON5-related tauopathy. Acta Neuropathol 2016; 132:531.
  114. Boronat A, Gelfand JM, Gresa-Arribas N, et al. Encephalitis and antibodies to dipeptidyl-peptidase-like protein-6, a subunit of Kv4.2 potassium channels. Ann Neurol 2013; 73:120.
  115. Hara M, Ariño H, Petit-Pedrol M, et al. DPPX antibody-associated encephalitis: Main syndrome and antibody effects. Neurology 2017; 88:1340.
  116. Balint B, Jarius S, Nagel S, et al. Progressive encephalomyelitis with rigidity and myoclonus: a new variant with DPPX antibodies. Neurology 2014; 82:1521.
  117. Tobin WO, Lennon VA, Komorowski L, et al. DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients. Neurology 2014; 83:1797.
  118. Mas N, Saiz A, Leite MI, et al. Antiglycine-receptor encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 2011; 82:1399.
  119. Hutchinson M, Waters P, McHugh J, et al. Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 2008; 71:1291.
  120. Clerinx K, Breban T, Schrooten M, et al. Progressive encephalomyelitis with rigidity and myoclonus: resolution after thymectomy. Neurology 2011; 76:303.
  121. Turner MR, Irani SR, Leite MI, et al. Progressive encephalomyelitis with rigidity and myoclonus: glycine and NMDA receptor antibodies. Neurology 2011; 77:439.
  122. Carvajal-González A, Leite MI, Waters P, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain 2014; 137:2178.
  123. Martinez-Hernandez E, Ariño H, McKeon A, et al. Clinical and Immunologic Investigations in Patients With Stiff-Person Spectrum Disorder. JAMA Neurol 2016; 73:714.
  124. Martinez-Hernandez E, Sepulveda M, Rostásy K, et al. Antibodies to aquaporin 4, myelin-oligodendrocyte glycoprotein, and the glycine receptor α1 subunit in patients with isolated optic neuritis. JAMA Neurol 2015; 72:187.
  125. Armangué T, Sabater L, Torres-Vega E, et al. Clinical and Immunological Features of Opsoclonus-Myoclonus Syndrome in the Era of Neuronal Cell Surface Antibodies. JAMA Neurol 2016; 73:417.
  126. Tekturk P, Baykan B, Erdag E, et al. Investigation of neuronal auto-antibodies in children diagnosed with epileptic encephalopathy of unknown cause. Brain Dev 2018; 40:909.
  127. Prüss H, Rothkirch M, Kopp U, et al. Limbic encephalitis with mGluR5 antibodies and immunotherapy-responsive prosopagnosia. Neurology 2014; 83:1384.
  128. Lancaster E, Martinez-Hernandez E, Titulaer MJ, et al. Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology 2011; 77:1698.
  129. Mat A, Adler H, Merwick A, et al. Ophelia syndrome with metabotropic glutamate receptor 5 antibodies in CSF. Neurology 2013; 80:1349.
  130. Spatola M, Sabater L, Planagumà J, et al. Encephalitis with mGluR5 antibodies: Symptoms and antibody effects. Neurology 2018; 90:e1964.
  131. Spatola M, Petit Pedrol M, Maudes E, et al. Clinical features, prognostic factors, and antibody effects in anti-mGluR1 encephalitis. Neurology 2020; 95:e3012.
  132. Sillevis Smitt P, Kinoshita A, De Leeuw B, et al. Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 2000; 342:21.
  133. Lopez-Chiriboga AS, Komorowski L, Kümpfel T, et al. Metabotropic glutamate receptor type 1 autoimmunity: Clinical features and treatment outcomes. Neurology 2016; 86:1009.
  134. Gresa-Arribas N, Planagumà J, Petit-Pedrol M, et al. Human neurexin-3α antibodies associate with encephalitis and alter synapse development. Neurology 2016; 86:2235.
  135. Fang B, McKeon A, Hinson SR, et al. Autoimmune Glial Fibrillary Acidic Protein Astrocytopathy: A Novel Meningoencephalomyelitis. JAMA Neurol 2016; 73:1297.
  136. Flanagan EP, Hinson SR, Lennon VA, et al. Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: Analysis of 102 patients. Ann Neurol 2017; 81:298.
  137. Gravier-Dumonceau A, Ameli R, Rogemond V, et al. Glial Fibrillary Acidic Protein Autoimmunity: A French Cohort Study. Neurology 2022; 98:e653.
  138. Tewkesbury G, Song JW, Perrone CM. Magnetic Resonance Imaging of Autoimmune GFAP Astrocytopathy. Ann Neurol 2021; 90:691.
  139. Ruiz-García R, Martínez-Hernández E, Joubert B, et al. Paraneoplastic cerebellar ataxia and antibodies to metabotropic glutamate receptor 2. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  140. Yaguchi H, Yabe I, Takahashi H, et al. Identification of anti-Sez6l2 antibody in a patient with cerebellar ataxia and retinopathy. J Neurol 2014; 261:224.
  141. Landa J, Guasp M, Petit-Pedrol M, et al. Seizure-related 6 homolog like 2 autoimmunity: Neurologic syndrome and antibody effects. Neurol Neuroimmunol Neuroinflamm 2021; 8.
  142. Landa J, Guasp M, Míguez-Cabello F, et al. Encephalitis with Autoantibodies against the Glutamate Kainate Receptors GluK2. Ann Neurol 2021; 90:101.
  143. Gultekin SH, Rosenfeld MR, Voltz R, et al. Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients. Brain 2000; 123 ( Pt 7):1481.
  144. Dalmau J, Graus F, Rosenblum MK, Posner JB. Anti-Hu--associated paraneoplastic encephalomyelitis/sensory neuronopathy. A clinical study of 71 patients. Medicine (Baltimore) 1992; 71:59.
  145. Graus F, Keime-Guibert F, Reñe R, et al. Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain 2001; 124:1138.
  146. Sillevis Smitt P, Grefkens J, de Leeuw B, et al. Survival and outcome in 73 anti-Hu positive patients with paraneoplastic encephalomyelitis/sensory neuronopathy. J Neurol 2002; 249:745.
  147. Saiz A, Bruna J, Stourac P, et al. Anti-Hu-associated brainstem encephalitis. J Neurol Neurosurg Psychiatry 2009; 80:404.
  148. Honnorat J, Didelot A, Karantoni E, et al. Autoimmune limbic encephalopathy and anti-Hu antibodies in children without cancer. Neurology 2013; 80:2226.
  149. van Broekhoven F, de Graaf MT, Bromberg JE, et al. Human chorionic gonadotropin treatment of anti-Hu-associated paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 2010; 81:1341.
  150. Shams'ili S, de Beukelaar J, Gratama JW, et al. An uncontrolled trial of rituximab for antibody associated paraneoplastic neurological syndromes. J Neurol 2006; 253:16.
  151. Darnell RB, DeAngelis LM. Regression of small-cell lung carcinoma in patients with paraneoplastic neuronal antibodies. Lancet 1993; 341:21.
  152. Voltz R, Gultekin SH, Rosenfeld MR, et al. A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer. N Engl J Med 1999; 340:1788.
  153. Dalmau J, Graus F, Villarejo A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004; 127:1831.
  154. Dalmau J, Gultekin SH, Voltz R, et al. Ma1, a novel neuron- and testis-specific protein, is recognized by the serum of patients with paraneoplastic neurological disorders. Brain 1999; 122 ( Pt 1):27.
  155. Waragai M, Chiba A, Uchibori A, et al. Anti-Ma2 associated paraneoplastic neurological syndrome presenting as encephalitis and progressive muscular atrophy. J Neurol Neurosurg Psychiatry 2006; 77:111.
  156. Mathew RM, Vandenberghe R, Garcia-Merino A, et al. Orchiectomy for suspected microscopic tumor in patients with anti-Ma2-associated encephalitis. Neurology 2007; 68:900.
  157. Hoffmann LA, Jarius S, Pellkofer HL, et al. Anti-Ma and anti-Ta associated paraneoplastic neurological syndromes: 22 newly diagnosed patients and review of previous cases. J Neurol Neurosurg Psychiatry 2008; 79:767.
  158. Al-Thubaiti I, Al-Hayek K, Binfalah M. Anti-Ma-associated encephalitis due to dysgerminoma in a woman with Swyer syndrome. Neurology 2013; 80:1439.
  159. Blumenthal DT, Salzman KL, Digre KB, et al. Early pathologic findings and long-term improvement in anti-Ma2-associated encephalitis. Neurology 2006; 67:146.
  160. Yu Z, Kryzer TJ, Griesmann GE, et al. CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 2001; 49:146.
  161. Vernino S, Tuite P, Adler CH, et al. Paraneoplastic chorea associated with CRMP-5 neuronal antibody and lung carcinoma. Ann Neurol 2002; 51:625.
  162. Muehlschlegel S, Okun MS, Foote KD, et al. Paraneoplastic chorea with leukoencephalopathy presenting with obsessive-compulsive and behavioral disorder. Mov Disord 2005; 20:1523.
  163. Werry C, Götz F, Wurster U, et al. Paraneoplastic autoimmune encephalitis associated with CV2/CRMP-5 IgG antineuronal antibodies in a patient with thymoma. J Neurol 2009; 256:129.
  164. Rickman OB, Parisi JE, Yu Z, et al. Fulminant autoimmune cortical encephalitis associated with thymoma treated with plasma exchange. Mayo Clin Proc 2000; 75:1321.
  165. De Camilli P, Thomas A, Cofiell R, et al. The synaptic vesicle-associated protein amphiphysin is the 128-kD autoantigen of Stiff-Man syndrome with breast cancer. J Exp Med 1993; 178:2219.
  166. Murinson BB, Guarnaccia JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology 2008; 71:1955.
  167. Ariño H, Höftberger R, Gresa-Arribas N, et al. Paraneoplastic Neurological Syndromes and Glutamic Acid Decarboxylase Antibodies. JAMA Neurol 2015; 72:874.
  168. Sutton IJ, Barnett MH, Watson JD, et al. Paraneoplastic brainstem encephalitis and anti-Ri antibodies. J Neurol 2002; 249:1597.
  169. Mandel-Brehm C, Dubey D, Kryzer TJ, et al. Kelch-like Protein 11 Antibodies in Seminoma-Associated Paraneoplastic Encephalitis. N Engl J Med 2019; 381:47.
  170. Dubey D, Wilson MR, Clarkson B, et al. Expanded Clinical Phenotype, Oncological Associations, and Immunopathologic Insights of Paraneoplastic Kelch-like Protein-11 Encephalitis. JAMA Neurol 2020; 77:1420.
  171. Maudes E, Landa J, Muñoz-Lopetegi A, et al. Clinical significance of Kelch-like protein 11 antibodies. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  172. Vogrig A, Péricart S, Pinto AL, et al. Immunopathogenesis and proposed clinical score for identifying Kelch-like protein-11 encephalitis. Brain Commun 2021; 3:fcab185.
  173. Saiz A, Blanco Y, Sabater L, et al. Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 2008; 131:2553.
  174. Muñoz-Lopetegi A, de Bruijn MAAM, Boukhrissi S, et al. Neurologic syndromes related to anti-GAD65: Clinical and serologic response to treatment. Neurol Neuroimmunol Neuroinflamm 2020; 7.
  175. Graus F, Saiz A, Dalmau J. GAD antibodies in neurological disorders - insights and challenges. Nat Rev Neurol 2020; 16:353.
  176. Tüzün E, Rossi JE, Karner SF, et al. Adenylate kinase 5 autoimmunity in treatment refractory limbic encephalitis. J Neuroimmunol 2007; 186:177.
  177. Do LD, Chanson E, Desestret V, et al. Characteristics in limbic encephalitis with anti-adenylate kinase 5 autoantibodies. Neurology 2017; 88:514.
  178. Bien CI, Nehls F, Kollmar R, et al. Identification of adenylate kinase 5 antibodies during routine diagnostics in a tissue-based assay: Three new cases and a review of the literature. J Neuroimmunol 2019; 334:576975.
  179. Muñiz-Castrillo S, Hedou JJ, Ambati A, et al. Distinctive clinical presentation and pathogenic specificities of anti-AK5 encephalitis. Brain 2021; 144:2709.
  180. Dalmau J, Graus F. Diagnostic criteria for autoimmune encephalitis: utility and pitfalls for antibody-negative disease. Lancet Neurol 2023; 22:529.
  181. Dalmau J, Rosenfeld MR. Paraneoplastic syndromes of the CNS. Lancet Neurol 2008; 7:327.
  182. Blanc F, Ben Abdelghani K, Schramm F, et al. Whipple limbic encephalitis. Arch Neurol 2011; 68:1471.
  183. Blondin NA, Vortmeyer AO, Harel NY. Paraneoplastic syndrome of inappropriate antidiuretic hormone mimicking limbic encephalitis. Arch Neurol 2011; 68:1591.
  184. Moragas M, Martínez-Yélamos S, Majós C, et al. Rhombencephalitis: a series of 97 patients. Medicine (Baltimore) 2011; 90:256.
  185. Lejuste F, Thomas L, Picard G, et al. Neuroleptic intolerance in patients with anti-NMDAR encephalitis. Neurol Neuroimmunol Neuroinflamm 2016; 3:e280.
  186. Lawn ND, Westmoreland BF, Kiely MJ, et al. Clinical, magnetic resonance imaging, and electroencephalographic findings in paraneoplastic limbic encephalitis. Mayo Clin Proc 2003; 78:1363.
  187. Abboud H, Probasco JC, Irani S, et al. Autoimmune encephalitis: proposed best practice recommendations for diagnosis and acute management. J Neurol Neurosurg Psychiatry 2021; 92:757.
  188. Hacohen Y, Wright S, Waters P, et al. Paediatric autoimmune encephalopathies: clinical features, laboratory investigations and outcomes in patients with or without antibodies to known central nervous system autoantigens. J Neurol Neurosurg Psychiatry 2013; 84:748.
  189. Jarius S, Hoffmann L, Clover L, et al. CSF findings in patients with voltage gated potassium channel antibody associated limbic encephalitis. J Neurol Sci 2008; 268:74.
  190. Escudero D, Guasp M, Ariño H, et al. Antibody-associated CNS syndromes without signs of inflammation in the elderly. Neurology 2017; 89:1471.
  191. Armangue T, Olivé-Cirera G, Martínez-Hernandez E, et al. Associations of paediatric demyelinating and encephalitic syndromes with myelin oligodendrocyte glycoprotein antibodies: a multicentre observational study. Lancet Neurol 2020; 19:234.
  192. Ebright MJ, Li SH, Reynolds E, et al. Unintended consequences of Mayo paraneoplastic evaluations. Neurology 2018; 91:e2057.
  193. Flanagan EP, Geschwind MD, Lopez-Chiriboga AS, et al. Autoimmune Encephalitis Misdiagnosis in Adults. JAMA Neurol 2023; 80:30.
  194. Dalmau J, Graus F. Autoimmune Encephalitis-Misdiagnosis, Misconceptions, and How to Avoid Them. JAMA Neurol 2023; 80:12.
  195. Graus F, Vogrig A, Muñiz-Castrillo S, et al. Updated Diagnostic Criteria for Paraneoplastic Neurologic Syndromes. Neurol Neuroimmunol Neuroinflamm 2021; 8.
Topic 15759 Version 78.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟